1
|
Neuen SM, Ophelders DR, Widowski H, Hütten MC, Brokken T, van Gorp C, Nikkels PG, Severens-Rijvers CA, Sthijns MM, van Blitterswijk CA, Troost FJ, LaPointe VL, Jolani S, Seiler C, Pillow JJ, Delhaas T, Reynaert NL, Wolfs TG. Multipotent adult progenitor cells prevent functional impairment and improve development in inflammation driven detriment of preterm ovine lungs. Regen Ther 2024; 27:207-217. [PMID: 38576851 PMCID: PMC10990734 DOI: 10.1016/j.reth.2024.03.014] [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: 12/11/2023] [Revised: 03/01/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024] Open
Abstract
Background Perinatal inflammation increases the risk for bronchopulmonary dysplasia in preterm neonates, but the underlying pathophysiological mechanisms remain largely unknown. Given their anti-inflammatory and regenerative capacity, multipotent adult progenitor cells (MAPC) are a promising cell-based therapy to prevent and/or treat the negative pulmonary consequences of perinatal inflammation in the preterm neonate. Therefore, the pathophysiology underlying adverse preterm lung outcomes following perinatal inflammation and pulmonary benefits of MAPC treatment at the interface of prenatal inflammatory and postnatal ventilation exposures were elucidated. Methods Instrumented ovine fetuses were exposed to intra-amniotic lipopolysaccharide (LPS 5 mg) at 125 days gestation to induce adverse systemic and peripheral organ outcomes. MAPC (10 × 106 cells) or saline were administered intravenously two days post LPS exposure. Fetuses were delivered preterm five days post MAPC treatment and either killed humanely immediately or mechanically ventilated for 72 h. Results Antenatal LPS exposure resulted in inflammation and decreased alveolar maturation in the preterm lung. Additionally, LPS-exposed ventilated lambs showed continued pulmonary inflammation and cell junction loss accompanied by pulmonary edema, ultimately resulting in higher oxygen demand. MAPC therapy modulated lung inflammation, prevented loss of epithelial and endothelial barriers and improved lung maturation in utero. These MAPC-driven improvements remained evident postnatally, and prevented concomitant pulmonary edema and functional loss. Conclusion In conclusion, prenatal inflammation sensitizes the underdeveloped preterm lung to subsequent postnatal inflammation, resulting in injury, disturbed development and functional impairment. MAPC therapy partially prevents these changes and is therefore a promising approach for preterm infants to prevent adverse pulmonary outcomes.
Collapse
Affiliation(s)
- Sophie M.L. Neuen
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Daan R.M.G. Ophelders
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Helene Widowski
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of BioMedical Engineering, Maastricht University, Maastricht, the Netherlands
| | - Matthias C. Hütten
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Tim Brokken
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Charlotte van Gorp
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Peter G.J. Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Carmen A.H. Severens-Rijvers
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
- Department of Pathology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mireille M.J.P.E. Sthijns
- Food Innovation and Health, Department of Human Biology, Maastricht University, Venlo, the Netherlands
- NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, the Netherlands
| | | | - Freddy J. Troost
- Food Innovation and Health, Department of Human Biology, Maastricht University, Venlo, the Netherlands
| | - Vanessa L.S. LaPointe
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, the Netherlands
| | - Shahab Jolani
- Department of Methodology and Statistics, School CAPHRI, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Christof Seiler
- Department of Advanced Computing Sciences, Maastricht University, Maastricht, the Netherlands
- Mathematics Centre Maastricht, Maastricht University, the Netherlands
| | - J. Jane Pillow
- School of Human Sciences, University of Western Australia, Perth, WA, Australia
| | - Tammo Delhaas
- Department of BioMedical Engineering, Maastricht University, Maastricht, the Netherlands
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Niki L. Reynaert
- NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
- Department of Respiratory Medicine, Maastricht University, Maastricht, the Netherlands
| | - Tim G.A.M. Wolfs
- Department of Pediatrics, Maastricht University Medical Center, MosaKids Children's Hospital, Maastricht, the Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
2
|
Abdelaziz MA, Alalawy AI, Sobhi M, Alatawi OM, Alaysuy O, Alshehri MG, Mohamed ELI, Abdelaziz MM, Algrfan IA, Mohareb RM. Elaboration of chitosan nanoparticles loaded with star anise extract as a therapeutic system for lung cancer: Physicochemical and biological evaluation. Int J Biol Macromol 2024; 279:135099. [PMID: 39197631 DOI: 10.1016/j.ijbiomac.2024.135099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/13/2024] [Accepted: 08/24/2024] [Indexed: 09/01/2024]
Abstract
The research study aimed to maximize the important medical role of star anise extract (SAE) through its loading on a widely available natural polymer (chitosan, Cs). Thus, SAE loaded chitosan nanoparticles (CsNPs) was prepared. The finding illustrated the formation of spherical particles of SAE loaded CsNPs as proved by transmission electron microscope (TEM). In addition, the average particle size of CsNPs and SAE loaded CsNPs are 131.8 ± 24.63 and 318.5 ± 73.94 nm, respectively. Scanning electron microscope (SEM) showed the presence of many spherical particles deposited on the surface of CsNPs owing to the deposition of SAE on the surface and encapsulated into pores of CsNPs. It also showed the presence of elements such as sodium, potassium, copper, magnesium, zinc, calcium, and iron, as well as the elements that accompanied with CsNPs: carbon, oxygen, nitrogen, and phosphorus. The extract was rich in bioactive components, such as anethole, shikimic acid, and different flavonoids, contributing to its medicinal qualities. The bioactive molecules in SAE were assessed by chromatographic analysis. Using the agar well diffusion test, the antibacterial qualities of CsNPs and SAE loaded CsNPs were evaluated against pathogenic bacteria linked to lung illnesses. The most significant inhibition zones showed that the SAE loaded CsNPs had the most antibacterial activity. The anticancer activity using MTT assay was used in the biological assessments to determine the cytotoxicity against the NCl-H460 lung cancer cell line. The results showed that CsNPs loaded with SAE considerably decreased cell viability in a dose-dependent manner, with the most significant anticancer impact by SAE loaded CsNPs. Furthermore, in vivo tests on lung cancer therapy revealed that when compared to other treatment groups, the SAE loaded CsNPs group showed the greatest reduction in tumor biomarkers and inflammation, as seen by decreased levels of Plasma malondialdehyde (MDA), tumor protein 53 (p53), Tumor necrosis factor-alpha (TNF- alpha), and fibronectin. Results concluded that these thorough characterizations, biological assessments, and antibacterial tests have confirmed the effective integration of SAE into CsNPs. Further, SAE loaded CsNPs could be a suitable option for various biomedical applications in tackling lung cancer and the inactivation of bacterial infection.
Collapse
Affiliation(s)
- Mahmoud A Abdelaziz
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia.
| | - Adel I Alalawy
- Department of Biochemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Mohamed Sobhi
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Omar M Alatawi
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Omaymah Alaysuy
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Maryam G Alshehri
- Department of Mathematics, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - ELsiddig Idriss Mohamed
- Department of Statistics, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Maiar M Abdelaziz
- Department of Mathematics, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Ibrahim A Algrfan
- Organic Chemistry Research Laboratory, Department of Chemistry, Faculty of Science, University of Tabuk, P.O. Box 741, Tabuk 71491, Saudi Arabia
| | - Rafat M Mohareb
- Department of Chemistry, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
3
|
Kang L, Cao J, Guo W, Cui X, Wei Y, Zhang J, Liu F, Duan C, Lin Q, Lv P, Ni Z, Zuo J, Shen H. Tumor necrosis factor-α-dependent inflammation upregulates high mobility group box 1 to induce tumor promotion and anti-programmed cell death protein-1 immunotherapy resistance in lung adenocarcinoma. J Transl Med 2024:102164. [PMID: 39461427 DOI: 10.1016/j.labinv.2024.102164] [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: 04/18/2024] [Revised: 09/17/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
Tumor-associated chronic lung inflammation depends on tumor necrosis factor (TNF)-α to activate several cytokines as part of an inflammatory loop, which plays a critical role in tumor progression in lung adenocarcinoma. High mobility group box 1 (HMGB1) is a cytokine that mediates inflammation. Whether TNF-α-induced inflammation regulates HMGB1 to contribute to tumor progression and promotion in lung adenocarcinoma remains unclear. Thus, human samples and a urethane-induced inflammation-driven lung adenocarcinoma (IDLA) mouse model were used to explore the involvement of HMGB1 in tumorigenesis, tumor progression, and efficacy of anti-programmed cell death protein (PD)-1 immunotherapy. High levels of HMGB1 were observed in human lung adenocarcinoma associated with poor overall survival in patients. HMGB1 upregulation was positively correlated with TNF-α-related inflammation and TIM3+ infiltration. TNF-α upregulated intracellular and extracellular HMGB1 expression to contribute to tumor promotion in A549 cells in vitro. Using a urethane-induced IDLA mouse model, we found HMGB1 upregulation was associated with increased TIM3+ T cell infiltration. Blocking TNF-α-dependent inflammation downregulated HMGB1 expression and inhibited tumorigenesis in the IDLA. Anti-PD-1 treatment alone did not inhibit tumor growth in the TNF-α-dependent IDLA, whereas anti-PD-1 combined with TNF-α blockade overcame anti-PD-1 immunotherapy resistance. Furthermore, anti-PD-1 combined with anti-HMGB1 also inhibited tumor growth in IDLA, suggesting increased HMGB1 release by TNF-α contributes to the resistance of anti-PD-1 immunotherapy in IDLA. Thus, tumor-associated TNF-α-dependent inflammation upregulated intracellular and extracellular HMGB1 expression in an inflammatory loop, contributing to tumor promotion and anti-PD-1 immunotherapy resistance in lung adenocarcinoma.
Collapse
Affiliation(s)
- Lifei Kang
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Department of Pathology, Hebei Chest Hospital, Shijiazhuang, China
| | - Jingjing Cao
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China; Department of Pathology, Lishui Central Hospital of Zhejiang Province, The fifth affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Wenli Guo
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Department of Pathology, The Second Hospital, Hebei Medical University, Shijiazhuang, China
| | - Xiaohui Cui
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Yangxuan Wei
- Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Jiayu Zhang
- Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Feiran Liu
- Department of Oncology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, China
| | - Chenyang Duan
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China
| | - Qiang Lin
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, Renqiu, Hebei, China
| | - Ping Lv
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Zhiyu Ni
- Affiliated Hospital of Hebei Engineering University, Handan, Hebei, China; Clinical Medical College, Hebei University of Engineering, Handan, Hebei, China; Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei, China.
| | - Jing Zuo
- Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China; Department of Oncology, The Fourth Hospital, Hebei Medical University, Shijiazhuang, China; Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei, China.
| | - Haitao Shen
- Laboratory of Pathology, Hebei Medical University, Shijiazhuang, China; Center of Metabolic Diseases and Cancer Research (CMCR), Hebei Medical University, Shijiazhuang, China; Hebei Collaborative Innovation Center of Tumor Microecological Metabolism Regulation, Affiliated Hospital of Hebei University, Baoding, Hebei, China.
| |
Collapse
|
4
|
Ponholzer F, Bogensperger C, Krendl FJ, Krapf C, Dumfarth J, Schneeberger S, Augustin F. Beyond the organ: lung microbiome shapes transplant indications and outcomes. Eur J Cardiothorac Surg 2024; 66:ezae338. [PMID: 39288305 PMCID: PMC11466426 DOI: 10.1093/ejcts/ezae338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/14/2024] [Indexed: 09/19/2024] Open
Abstract
The lung microbiome plays a crucial role in the development of chronic lung diseases, which may ultimately lead to the need for lung transplantation. Also, perioperative results seem to be connected with altered lung microbiomes and its dynamic changes providing a possible target for optimizing short-term outcome after transplantation. A literature review using MEDLINE, PubMed Central and Bookshelf was performed. Chronic lung allograft dysfunction (CLAD) seems to be influenced and partly triggered by changes in the pulmonary microbiome and dysbiosis, e.g. through increased bacterial load or abundance of specific species such as Pseudomonas aeruginosa. Additionally, the specific indications for transplantation, with their very heterogeneous changes and influences on the pulmonary microbiome, influence long-term outcome. Next to composition and measurable bacterial load, dynamic changes in the allografts microbiome also possess the ability to alter long-term outcomes negatively. This review discusses the "new" microbiome after transplantation and the associations with direct postoperative outcome. With the knowledge of these principles the impact of alterations in the pulmonary microbiome in hindsight to CLAD and possible therapeutic implications are described and discussed. The aim of this review is to summarize the current literature regarding pre- and postoperative lung microbiomes and how they influence different lung diseases on their progression to failure of conservative treatment. This review provides a summary of current literature for centres looking for further options in optimizing lung transplant outcomes and highlights possible areas for further research activities investigating the pulmonary microbiome in connection to transplantation.
Collapse
Affiliation(s)
- Florian Ponholzer
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christina Bogensperger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Julius Krendl
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Krapf
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Dumfarth
- Department of Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Augustin
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
5
|
Woodrow JS, Hopster K, Palmisano M, Payette F, Kulp J, Stefanovski D, Nolen‐Walston R. Time to resolution of airway inflammation caused by bronchoalveolar lavage in healthy horses. J Vet Intern Med 2024; 38:2776-2782. [PMID: 39198933 PMCID: PMC11423487 DOI: 10.1111/jvim.17169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Bronchoalveolar lavage (BAL) is a common procedure for evaluation of the equine lower airways. Time to resolution of post-BAL inflammation has not been clearly defined. HYPOTHESIS Residual inflammation, evident by changes in immune cell populations and inflammatory cytokines, will resolve by 72 hours after BAL. ANIMALS Six adult, healthy, institution-owned horses. METHODS Randomized, complete cross-over design. Each horse underwent 3 paired BALs, including a baseline and then 48, 72, and 96 hours later, with a 7-day washout between paired BALs. Each sample underwent cytological evaluation and cytokine concentrations were determined by a commercially available multiplex bead immunoassay. Statistical analysis was performed by multilevel mixed-effects Poisson regression analysis. Data are reported as marginal means and 95% confidence interval (CI). RESULTS Neutrophil, eosinophil and mast cell percentages were not significantly different at any time points. Macrophage percentages were higher at 72 hours (45.0 [95% CI, 41.6-48.4]%) and 96 hours (45.3 [95% CI, 42.9-47.7]%) vs baseline (37.4 [95% CI, 33.5-41.4]%; P < .001 and P = .01, respectively), and at 72 hours and 96 hours vs 48 hours (31.9 [95% CI, 28.1-35.6]%; P < .001). Neutrophil percentage was not significantly increased at 48 hours (P = .11). Interleukin (IL)-6 concentration was increased at 72 hours (5.22 [95% CI, 3.44-6.99] pg/mL) vs 48 hours (4.38 [95% CI, 2.99-5.78] pg/mL; P < .001). CONCLUSIONS AND CLINICAL IMPORTANCE Significant lung inflammation was not detected at 72 and 96 hours, suggesting that repeating BAL at 72 hours or more can be done without concern of residual inflammation.
Collapse
Affiliation(s)
- Jane S. Woodrow
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Klaus Hopster
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Megan Palmisano
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Flavie Payette
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Jeaneen Kulp
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Darko Stefanovski
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| | - Rose Nolen‐Walston
- Department of Clinical Sciences‐New Bolton Center, College of Veterinary MedicineUniversity of PennsylvaniaKennett SquarePennsylvaniaUSA
| |
Collapse
|
6
|
Hernandez-Gonzalez F, Pietrocola F, Cameli P, Bargagli E, Prieto-González S, Cruz T, Mendoza N, Rojas M, Serrano M, Agustí A, Faner R, Gómez-Puerta JA, Sellares J. Exploring the Interplay between Cellular Senescence, Immunity, and Fibrosing Interstitial Lung Diseases: Challenges and Opportunities. Int J Mol Sci 2024; 25:7554. [PMID: 39062798 PMCID: PMC11276754 DOI: 10.3390/ijms25147554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Fibrosing interstitial lung diseases (ILDs) are characterized by the gradual and irreversible accumulation of scar tissue in the lung parenchyma. The role of the immune response in the pathogenesis of pulmonary fibrosis remains unclear. In recent years, substantial advancements have been made in our comprehension of the pathobiology driving fibrosing ILDs, particularly concerning various age-related cellular disturbances and immune mechanisms believed to contribute to an inadequate response to stress and increased susceptibility to lung fibrosis. Emerging studies emphasize cellular senescence as a key mechanism implicated in the pathobiology of age-related diseases, including pulmonary fibrosis. Cellular senescence, marked by antagonistic pleiotropy, and the complex interplay with immunity, are pivotal in comprehending many aspects of lung fibrosis. Here, we review progress in novel concepts in cellular senescence, its association with the dysregulation of the immune response, and the evidence underlining its detrimental role in fibrosing ILDs.
Collapse
Affiliation(s)
- Fernanda Hernandez-Gonzalez
- Department of Respiratory Medicine, Respiratory Institute, Hospital Clinic Barcelona, 08036 Barcelona, Spain; (A.A.); (J.S.)
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
| | - Federico Pietrocola
- Department of Cell and Molecular Biology, Karolinska Institutet, 17165 Solna, Sweden;
| | - Paolo Cameli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neuro-Sciences, University of Siena, 53100 Siena, Italy; (P.C.); (E.B.)
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences & Neuro-Sciences, University of Siena, 53100 Siena, Italy; (P.C.); (E.B.)
| | - Sergio Prieto-González
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Vasculitis Research Unit, Department of Autoimmune Diseases, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Tamara Cruz
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
| | - Nuria Mendoza
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
| | - Mauricio Rojas
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Manuel Serrano
- Cambridge Institute of Science, Altos Labs, Cambridge CB21 6GP, UK;
| | - Alvar Agustí
- Department of Respiratory Medicine, Respiratory Institute, Hospital Clinic Barcelona, 08036 Barcelona, Spain; (A.A.); (J.S.)
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
| | - Rosa Faner
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
- Biomedicine Department, University of Barcelona, 08036 Barcelona, Spain
| | - Jose A. Gómez-Puerta
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Rheumatology Department, Hospital Clinic Barcelona, 08036 Barcelona, Spain
| | - Jacobo Sellares
- Department of Respiratory Medicine, Respiratory Institute, Hospital Clinic Barcelona, 08036 Barcelona, Spain; (A.A.); (J.S.)
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (S.P.-G.); (T.C.); (N.M.); (R.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro Investigación Biomédica en Red Enfermedades Respiratorias (CIBERES), 08036 Barcelona, Spain
| |
Collapse
|
7
|
Laprise F, Arduini A, Duguay M, Pan Q, Liang C. SARS-CoV-2 Accessory Protein ORF8 Targets the Dimeric IgA Receptor pIgR. Viruses 2024; 16:1008. [PMID: 39066171 PMCID: PMC11281603 DOI: 10.3390/v16071008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
SARS-CoV-2 is a highly pathogenic respiratory virus that successfully initiates and establishes its infection at the respiratory mucosa. However, little is known about how SARS-CoV-2 antagonizes the host's mucosal immunity. Recent findings have shown a marked reduction in the expression of the polymeric Ig receptor (pIgR) in COVID-19 patients. This receptor maintains mucosal homeostasis by transporting the dimeric IgA (dIgA) and pentameric IgM (pIgM) across mucosal epithelial cells to neutralize the invading respiratory pathogens. By studying the interaction between pIgR and SARS-CoV-2 proteins, we discovered that the viral accessory protein Open Reading Frame 8 (ORF8) potently downregulates pIgR expression and that this downregulation activity of ORF8 correlates with its ability to interact with pIgR. Importantly, the ORF8-mediated downregulation of pIgR diminishes the binding of dIgA or pIgM, and the ORF8 proteins of the variants of concern of SARS-CoV-2 preserve the function of downregulating pIgR, indicating the importance of this conserved activity of ORF8 in SARS-CoV-2 pathogenesis. We further observed that the secreted ORF8 binds to cell surface pIgR, but that this interaction does not trigger the cellular internalization of ORF8, which requires the binding of dIgA to pIgR. These findings suggest the role of ORF8 in SARS-CoV-2 mucosal immune evasion.
Collapse
Affiliation(s)
- Frederique Laprise
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ariana Arduini
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - Mathew Duguay
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Institut de Recherche Clinique de Montréal, Montreal, QC H2W 1R7, Canada
| | - Qinghua Pan
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
8
|
Zhao M, Hou W, Pu D, Li Z, Tu L, Ow CJL, Tian J, Li W. Impact of Pulmonary microbiota on lung cancer treatment-related pneumonia. J Cancer 2024; 15:4503-4512. [PMID: 39006071 PMCID: PMC11242340 DOI: 10.7150/jca.93818] [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: 01/03/2024] [Accepted: 05/19/2024] [Indexed: 07/16/2024] Open
Abstract
Background: The use of immunotherapy is progressively expanding for the treatment of lung cancer, either alone or in combination with radiotherapy. However, treatment-related adverse events, especially pneumonia, significantly limit the drug's effectiveness in treating lung cancer. The occurrence of lung cancer, immunotherapy, and pulmonary radiotherapy can all contribute to the imbalance in the pulmonary microbiota, rendering the lungs more susceptible to inflammatory reactions. Methods: Mouse models of lung transplantation tumor were treated with either PD-1 monoclonal antibody or radiotherapy alone, or in combination. The differences in lung inflammation among the different treatment groups were regularly observed by micro-CT. Further, bronchoalveolar lavage fluid was extracted for macrogenomic and cytokine detection. The transcriptional genome of tumor-filled lung tissue was also sequenced. Results: When treated with a combination of PD-1 and radiotherapy, the CT scans showed more severe pulmonary inflammation. However, with the addition of continuously administered antibiotics, no exacerbation of pneumonia signs was observed. Moreover, the differential gene expression and cytokine profiles in the combination treatment group differed from those in the PD-1 monotherapy group and the radiotherapy monotherapy group. This discrepancy does not seem to be a straightforward superimposition of radiation-induced pneumonia and immune-related pneumonia. Further exploration of changes in pulmonary microbiota revealed specific bacterial interactions with DEGs and cytokines. Conclusions: The underlying causes of this susceptibility are intricate and may be associated with the complexity of pulmonary microbiota imbalance, along with fluctuations in the abundance of specific microbiota species.
Collapse
Affiliation(s)
- Maoyuan Zhao
- Lung cancer center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wang Hou
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Pu
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhixi Li
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Tu
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Calista Jia Ling Ow
- BSc (Hons) Biochemistry, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Units of West China, Chinese Academy of Medical Sciences, West China Hospital, Chengdu, Sichuan, China
| |
Collapse
|
9
|
Zhu M, Yi Y, Jiang K, Liang Y, Li L, Zhang F, Zheng X, Yin H. Single-cell combined with transcriptome sequencing to explore the molecular mechanism of cell communication in idiopathic pulmonary fibrosis. J Cell Mol Med 2024; 28:e18499. [PMID: 38887981 PMCID: PMC11184282 DOI: 10.1111/jcmm.18499] [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: 11/03/2023] [Revised: 05/14/2024] [Accepted: 06/08/2024] [Indexed: 06/20/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a common, chronic, and progressive lung disease that severely impacts human health and survival. However, the intricate molecular underpinnings of IPF remains elusive. This study aims to delve into the nuanced molecular interplay of cellular interactions in IPF, thereby laying the groundwork for innovative therapeutic approaches in the clinical field of IPF. Sophisticated bioinformatics methods were employed to identify crucial biomarkers essential for the progression of IPF. The GSE122960 single-cell dataset was obtained from the Gene Expression Omnibus (GEO) compendium, and intercellular communication potentialities were scrutinized via CellChat. The random survival forest paradigm was established using the GSE70866 dataset. Quintessential genes were selected through Kaplan-Meier (KM) curves, while immune infiltration examinations, functional enrichment critiques and nomogram paradigms were inaugurated. Analysis of intercellular communication revealed an intimate potential connections between macrophages and various cell types, pinpointing five cardinal genes influencing the trajectory and prognosis of IPF. The nomogram paradigm, sculpted from these seminal genes, exhibits superior predictive prowess. Our research meticulously identified five critical genes, confirming their intimate association with the prognosis, immune infiltration and transcriptional governance of IPF. Interestingly, we discerned these genes' engagement with the EPITHELIAL_MESENCHYMAL_TRANSITION signalling pathway, which may enhance our understanding of the molecular complexity of IPF.
Collapse
Affiliation(s)
- Minggao Zhu
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Yuhu Yi
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Kui Jiang
- Department of NephrologyThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Yongzhi Liang
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Lijun Li
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Feng Zhang
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Xinglong Zheng
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| | - Haiyan Yin
- Intensive Care UnitThe First Affiliated Hospital of Jinan UniversityGuangzhouGuangdongChina
| |
Collapse
|
10
|
Tapken I, Detering NT, Claus P. What could be the function of the spinal muscular atrophy-causing protein SMN in macrophages? Front Immunol 2024; 15:1375428. [PMID: 38863697 PMCID: PMC11165114 DOI: 10.3389/fimmu.2024.1375428] [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: 01/23/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Spinal Muscular Atrophy (SMA), a neurodegenerative disorder, extends its impact beyond the nervous system. The central protein implicated in SMA, Survival Motor Neuron (SMN) protein, is ubiquitously expressed and functions in fundamental processes such as alternative splicing, translation, cytoskeletal dynamics and signaling. These processes are relevant for all cellular systems, including cells of the immune system such as macrophages. Macrophages are capable of modulating their splicing, cytoskeleton and expression profile in order to fulfil their role in tissue homeostasis and defense. However, less is known about impairment or dysfunction of macrophages lacking SMN and the subsequent impact on the immune system of SMA patients. We aimed to review the potential overlaps between SMN functions and macrophage mechanisms highlighting the need for future research, as well as the current state of research addressing the role of macrophages in SMA.
Collapse
Affiliation(s)
- Ines Tapken
- SMATHERIA gGmbH – Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Nora T. Detering
- SMATHERIA gGmbH – Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Peter Claus
- SMATHERIA gGmbH – Non-Profit Biomedical Research Institute, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| |
Collapse
|
11
|
Kaur J, Sharma A, Passi G, Dey P, Khajuria A, Alajangi HK, Jaiswal PK, Barnwal RP, Singh G. Nanomedicine at the Pulmonary Frontier: Immune-Centric Approaches for Respiratory Disease Treatment. Immunol Invest 2024; 53:295-347. [PMID: 38206610 DOI: 10.1080/08820139.2023.2298398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Respiratory diseases (RD) are a group of common ailments with a rapidly increasing global prevalence, posing a significant threat to humanity, especially the elderly population, and imposing a substantial burden on society and the economy. RD represents an unmet medical need that requires the development of viable pharmacotherapies. While various promising strategies have been devised to advance potential treatments for RD, their implementation has been hindered by difficulties in drug delivery, particularly in critically ill patients. Nanotechnology offers innovative solutions for delivering medications to the inflamed organ sites, such as the lungs. Although this approach is enticing, delivering nanomedicine to the lungs presents complex challenges that require sophisticated techniques. In this context, we review the potential of novel nanomedicine-based immunomodulatory strategies that could offer therapeutic benefits in managing this pressing health condition.
Collapse
Affiliation(s)
- Jatinder Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Akanksha Sharma
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Gautam Passi
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Piyush Dey
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Hema Kumari Alajangi
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, Texas, USA
| | | | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| |
Collapse
|
12
|
Zhao S, Li H, Yang F, Yang Y, Zeng Y, An Z, Li J, Wu H, Song J, Wu W. Association of short-term PM 2.5 exposure with airway innate immune response, microbiota and metabolism alterations in human airways. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 345:123435. [PMID: 38295929 DOI: 10.1016/j.envpol.2024.123435] [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: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/04/2024]
Abstract
Exposure to fine particulate matter (PM2.5) has been associated with impaired airway innate immunity, leading to diverse lung disorders. However, the mechanisms of the adverse effects of PM2.5 on the airway innate immune system has not been adequately elucidated. This study aimed to investigate the association between short-term exposure to ambient PM2.5 and airway innate immune responses. A panel study of 53 undergraduate students was conducted in November 2020 and April 2021. Levels of airway innate immune biomarkers including interleukin-1β (IL-1β), IL-4, IL-6, IL-8, IL-17, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), myeloperoxidase (MPO), and matrix metalloproteinase-9 (MMP-9) in induced sputum were measured, and airway microbiota and metabolites examined. Linear mixed-effect model was used to evaluate the effects of short-term exposure to PM2.5 on the above-listed airway immune biomarkers. The results indicated that for every 10 μg/m3 increase in PM2.5 concentration (at lag3), was associated with an increase of 21.3 % (5.4 %-37.1 %), 26.2 % (0.30 %-52.1 %), 22.4 % (0.70 %-44.2 %), 27.4 % (6.6 %-48.3 %), 18.3 % (4.6 %-31.9 %), 3.9 % (0.20 %-7.6 %) or 2.4 % (0.10 %-4.7 %) in IL-6, TNF-α, IL-17, IL-4, IFN-γ, MPO, or MMP-9 levels, respectively. Meanwhile, exposure to higher levels of ambient PM2.5 was found to significantly modulate airway microbiota and metabolite profile. Specifically, Prevotella and Fusobacterium, as well as 96 different metabolites were associated with PM2.5 levels. The metabolic pathways associated with these metabolites mainly included amino acid biosynthesis and metabolism. Notably, PM2.5 exposure-induced alterations of some airway microbiota were significantly correlated with specific airway metabolic change. Taken together, these results demonstrated that short-term exposure to PM2.5 was associated with alterations of airway immune response, microbial dysbiosis and changes of metabolites. This study provided insights into the mechanisms underlying PM2.5-induced airway innate immune responses.
Collapse
Affiliation(s)
- Shuaiqi Zhao
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Huijun Li
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Fuyun Yang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Yishu Yang
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Yuling Zeng
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Zhen An
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Juan Li
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Hui Wu
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Jie Song
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China
| | - Weidong Wu
- Henan International Collaborative Laboratory for Health Effects and Intervention of Air Pollution, School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, China.
| |
Collapse
|
13
|
Arifin J, Sakti M, Massi N, Bukhari A, Hamid F, Winangun PA. Vertebral destruction in tuberculous spondylitis correlates with Toll-like Receptor 4 (TLR-4) levels: a cross-sectional study. Ann Med Surg (Lond) 2024; 86:800-804. [PMID: 38333260 PMCID: PMC10849308 DOI: 10.1097/ms9.0000000000001707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 12/29/2023] [Indexed: 02/10/2024] Open
Abstract
Background Tuberculous (TB) spondylitis is a hazardous infectious disease causing significant spinal deformity. Increased toll-like receptor-4 (TLR-4) activity promotes more extensive infections in patients with TB spondylitis, so it has the potential to be used as a biomarker to predict the severity. This study aims to determine the relationship between TLR-4 levels and the degree of vertebral destruction in TB spondylitis patients. Materials and methods A cross-sectional study was conducted from May to October 2023. A total of 27 TB spondylitis samples were then measured for TLR-4 serum levels. Vertebral destruction is assessed based on the Spine At Risk Signs (SARS) criteria on X-ray and MRI examinations. Moreover, the degree of sensory and motor impairment was also assessed in this study. The Spearman correlation test assessed the correlation between TLR-4 levels and vertebral destruction. Results Most of the samples in this study were less than 30 years old (10 people, 37%), female (14 people, 51.9%), had spinal destruction at 1 level (11 people, 40.7%), had paraplegia (8 people, 29.6%), and had hypoesthesia (11 people, 40.7%). TLR-4 levels had a mean value of 8254.1±1076.1 ng/ml. TLR-4 levels were positively correlated with the degree of vertebral destruction (r=0.599, P=0.001), motor disorders (r=0.632.x, P=0.000), and sensory disorders (r=0.574, P=0.002). Conclusion TLR-4 levels are associated with the severity of vertebral destruction in TB spondylitis, so it has the potential to be used as a prognostic biomarker.
Collapse
Affiliation(s)
- Jainal Arifin
- Department of Orthopedic and Traumatology, Spine Divison
| | - Muhammad Sakti
- Department of Orthopedic and Traumatology, Sports and Injury Division
| | | | | | | | - Putu A.N. Winangun
- Orthopaedic and Traumatology Study Program, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
14
|
Jannini-Sá YAP, Creyns B, Hogaboam CM, Parks WC, Hohmann MS. Macrophages in Lung Repair and Fibrosis. Results Probl Cell Differ 2024; 74:257-290. [PMID: 39406909 DOI: 10.1007/978-3-031-65944-7_10] [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] [Indexed: 11/01/2024]
Abstract
Macrophages are key regulators of tissue repair and fibrosis. Following injury, macrophages undergo marked phenotypic and functional changes to play crucial roles throughout the phases of tissue repair. Idiopathic Pulmonary Fibrosis, which is the most common fibrosing lung disease, has been described as an aberrant reparative response to repetitive alveolar epithelial injury in a genetically susceptible aging individual. The marked destruction of the lung architecture results from the excessive secretion of extracellular matrix by activated fibroblasts and myofibroblasts. Accumulating evidence suggests that macrophages have a pivotal regulatory role in pulmonary fibrosis. The origins and characteristics of macrophages in the lung and their role in regulating lung homeostasis, repair, and fibrosis are reviewed herein. We discuss recent studies that have employed single-cell RNA-sequencing to improve the identification and characterization of macrophage populations in the context of homeostatic and fibrotic conditions. We also discuss the current understanding of the macrophage-mediated mechanisms underlying the initiation and progression of pulmonary fibrosis, with a focus on the phenotypic and functional changes that aging macrophages acquire and how these changes ultimately contribute to age-related chronic lung diseases.
Collapse
Affiliation(s)
- Yago A P Jannini-Sá
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brecht Creyns
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cory M Hogaboam
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - William C Parks
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Miriam S Hohmann
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [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: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
16
|
Graf J, Trautmann-Rodriguez M, Sabnis S, Kloxin AM, Fromen CA. On the path to predicting immune responses in the lung: Modeling the pulmonary innate immune system at the air-liquid interface (ALI). Eur J Pharm Sci 2023; 191:106596. [PMID: 37770004 PMCID: PMC10658361 DOI: 10.1016/j.ejps.2023.106596] [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: 06/12/2023] [Revised: 09/01/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Chronic respiratory diseases and infections are among the largest contributors to death globally, many of which still have no cure, including chronic obstructive pulmonary disorder, idiopathic pulmonary fibrosis, and respiratory syncytial virus among others. Pulmonary therapeutics afford untapped potential for treating lung infection and disease through direct delivery to the site of action. However, the ability to innovate new therapeutic paradigms for respiratory diseases will rely on modeling the human lung microenvironment and including key cellular interactions that drive disease. One key feature of the lung microenvironment is the air-liquid interface (ALI). ALI interface modeling techniques, using cell-culture inserts, organoids, microfluidics, and precision lung slices (PCLS), are rapidly developing; however, one major component of these models is lacking-innate immune cell populations. Macrophages, neutrophils, and dendritic cells, among others, represent key lung cell populations, acting as the first responders during lung infection or injury. Innate immune cells respond to and modulate stromal cells and bridge the gap between the innate and adaptive immune system, controlling the bodies response to foreign pathogens and debris. In this article, we review the current state of ALI culture systems with a focus on innate immune cells and suggest ways to build on current models to add complexity and relevant immune cell populations.
Collapse
Affiliation(s)
- Jodi Graf
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA
| | | | - Simone Sabnis
- Department of Biomedical Engineering, University of Delaware, Newark, DE 19716, USA
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, USA.
| | - Catherine A Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
17
|
Hickman E, Rebuli ME, Robinette C, Jaspers I. Understanding the Relationship Between Neutrophil Function and Demographic Variables. RESEARCH SQUARE 2023:rs.3.rs-3622445. [PMID: 38045266 PMCID: PMC10690322 DOI: 10.21203/rs.3.rs-3622445/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Neutrophils play a crucial role in the body's defense against respiratory pathogens, and dysregulation is linked to airway diseases. The study presented here explores the association between demographic factors (age, BMI, and sex) and functional phenotypes (oxidative burst and bioenergetics) of neutrophils. We measured PMA-stimulated oxidative burst (Seahorse XF) and phagocytosis (pHrodo red S. aureus ) of human peripheral blood neutrophils and determined whether there were significant demographic associations with cellular function. There were no significant associations between neutrophil oxidative burst bioenergetic parameters or phagocytosis and BMI or age. However, our data revealed sexual dimorphism in neutrophil phagocytosis, with males exhibiting significantly higher phagocytic capacity than females. Additionally, phagocytic capacity and bioenergetic parameters were correlated in males but not in females. The study indicates potential variations in neutrophil activation pathways between males and female and emphasizes the importance of considering sex as a biological variable in respiratory host defense research.
Collapse
|
18
|
Bhat TA, Kalathil SG, Leigh N, Hutson A, Goniewicz ML, Thanavala YM. Do alternative tobacco products induce less adverse respiratory risk than cigarettes? Respir Res 2023; 24:261. [PMID: 37907902 PMCID: PMC10617138 DOI: 10.1186/s12931-023-02568-2] [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: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023] Open
Abstract
RATIONALE Due to the relatively short existence of alternative tobacco products, gaps exist in our current understanding of their long-term respiratory health effects. We therefore undertook the first-ever side-by-side comparison of the impact of chronic inhalation of aerosols emitted from electronic cigarettes (EC) and heated tobacco products (HTP), and combustible cigarettes (CC) smoke. OBJECTIVES To evaluate the potential differential effects of alternative tobacco products on lung inflammatory responses and efficacy of vaccination in comparison to CC. METHODS Mice were exposed to emissions from EC, HTP, CC, or air for 8 weeks. BAL and lung tissue were analyzed for markers of inflammation, lung damage, and oxidative stress. Another group was exposed for 12 weeks and vaccinated and challenged with a bacterial respiratory infection. Antibody titers in BAL and sera and pulmonary bacterial clearance were assessed. MAIN RESULTS EC- and HTP-aerosols significantly augmented lung immune cell infiltrates equivalent to that achieved following CC-exposure. HTP and CC significantly increased neutrophil numbers compared to EC. All products augmented numbers of B cells, T cells, and pro-inflammatory IL17A+ T cells in the lungs. Decreased lung antioxidant activity and lung epithelial and endothelial damage was induced by all products. EC and HTP differentially augmented inflammatory cytokines/chemokines in the BAL. Generation of immunity following vaccination was impaired by EC and HTP but to a lesser extent than CC, with a CC > HTP > EC hierarchy of suppression of pulmonary bacterial clearance. CONCLUSIONS HTP and EC-aerosols induced a proinflammatory pulmonary microenvironment, lung damage, and suppressed efficacy of vaccination.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Immunology, Roswell Park Comprehensive Cancer Center, 665 Elm Street, Buffalo, NY, 14263, USA
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Suresh G Kalathil
- Department of Immunology, Roswell Park Comprehensive Cancer Center, 665 Elm Street, Buffalo, NY, 14263, USA
| | - Noel Leigh
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Maciej L Goniewicz
- Department of Health Behavior, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yasmin M Thanavala
- Department of Immunology, Roswell Park Comprehensive Cancer Center, 665 Elm Street, Buffalo, NY, 14263, USA.
| |
Collapse
|
19
|
Merk VM, Phan TS, Wiedmann A, Hardy RS, Lavery GG, Brunner T. Local glucocorticoid synthesis regulates house dust mite-induced airway hypersensitivity in mice. Front Immunol 2023; 14:1252874. [PMID: 37936704 PMCID: PMC10626452 DOI: 10.3389/fimmu.2023.1252874] [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: 07/04/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Background Extra-adrenal glucocorticoid (GC) synthesis at epithelial barriers, such as skin and intestine, has been shown to be important in the local regulation of inflammation. However, the role of local GC synthesis in the lung is less well studied. Based on previous studies and the uncontentious efficacy of corticosteroid therapy in asthma patients, we here investigated the role of 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1/Hsd11b1)-dependent local GC reactivation in the regulation of allergic airway inflammation. Methods Airway inflammation in Hsd11b1-deficient and C57BL/6 wild type mice was analyzed after injection of lipopolysaccharide (LPS) and anti-CD3 antibody, and in acute and chronic models of airway hypersensitivity induced by house dust mite (HDM) extract. The role of 11β-HSD1 in normal and inflammatory conditions was assessed by high dimensional flow cytometry, histological staining, RT-qPCR analysis, ex vivo tissue cultures, GC-bioassays and protein detection by ELISA and immunoblotting. Results Here we show that lung tissue from Hsd11b1-deficient mice synthesized significantly less GC ex vivo compared with wild type animals in response to immune cell stimulation. We further observed a drastically aggravated phenotype in Hsd11b1-deficient mice treated with HDM extract compared to wild type animals. Besides eosinophilic infiltration, Hsd11b1-deficient mice exhibited aggravated neutrophilic infiltration caused by a strong Th17-type immune response. Conclusion We propose an important role of 11β-HSD1 and local GC in regulating Th17-type rather than Th2-type immune responses in HDM-induced airway hypersensitivity in mice by potentially controlling Toll-like receptor 4 (TLR4) signaling and cytokine/chemokine secretion by airway epithelial cells.
Collapse
Affiliation(s)
- Verena M. Merk
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Truong San Phan
- Department of Biology, University of Konstanz, Konstanz, Germany
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alice Wiedmann
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Rowan S. Hardy
- Institute of Clinical Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Gareth G. Lavery
- Department of Biosciences, Nottingham Trent University, Nottingham, United Kingdom
| | - Thomas Brunner
- Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
20
|
Mohammadi Ebli F, Heshmatipour Z, Daneshjou K, Siadat SD. Simultaneous Genetically Detection of Streptococcuspyogenes, Streptococcuspneumoniae and Haemophilusinfluenzae in Patients with Treatment-Resistant Respiratory Infection. IRANIAN JOURNAL OF PATHOLOGY 2023; 18:356-362. [PMID: 37942203 PMCID: PMC10628374 DOI: 10.30699/ijp.2023.1991067.3075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/15/2023] [Indexed: 11/10/2023]
Abstract
Background & Objective Streptococcus pneumoniae, Haemophilus influenzae and Streptococcus pyogenes are among the most important causes of infection in human. Inventing rapid methods to identify these species can help in providing appropriate and effective treatment options. Therefore, the current study aimed to develop a multiplex touch-down PCR method to identify rapidly the aforementioned species patients' sputum samples, simultaneously. Methods A total of 50 sputum samples of patients with respiratory infections resistant to treatment were collected. After DNA extraction and primer design, the complete capsule (CAP) region II, capsular polysaccharide biosynthesis (cpsA) and the structural regulator of transcription (spy) genes were amplified for detecting H. influenzae, S. pneumoniae and S. pyogenes by multiplex touch-down PCR. Results Among 50 samples prepared from patients with different diseases, 27 samples were positive for amplified genes. The frequency of presence of pathogens in the collected samples included 14% H. influenzae, 20% S. pneumoniae and 20% S. pyogenes. Also, in some patients, the simultaneous presence of two or three pathogens were observed. Conclusion In general, it can be concluded that the PCR touchdown method developed in the present study is an effective and fast method for the simultaneous identification of H. influenzae, S. pneumoniae and S. pyogenes pathogens in clinical samples of patients.
Collapse
Affiliation(s)
- Farzad Mohammadi Ebli
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Zoheir Heshmatipour
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Khadijeh Daneshjou
- Department of Pediatrics, Imam Khomeini Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
21
|
Andrenacci B, De Filippo M, Votto M, Prevedoni Gorone MS, De Amici M, La Grutta S, Marseglia GL, Licari A. Severe pediatric asthma endotypes: current limits and future perspectives. Expert Rev Respir Med 2023; 17:675-690. [PMID: 37647343 DOI: 10.1080/17476348.2023.2254234] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/24/2023] [Accepted: 08/29/2023] [Indexed: 09/01/2023]
Abstract
INTRODUCTION Although rare, pediatric severe therapy-resistant asthma (STRA) is a highly heterogeneous, resource-demanding disease that differs significantly from severe adult asthma and whose pathogenesis is still poorly understood. AREAS COVERED This review summarizes the latest 10 years of English-written studies defining pediatric STRA endotypes using lung-specific techniques such as bronchoalveolar lavage and endobronchial biopsy. Results of the studies and limits on the field are discussed, together with some future perspectives. EXPERT OPINION Over the years, it has become increasingly clear that 'one size does not fit all" in asthma. However, "Does an extremely tailored size fit more than one?'. Only using multicentric, longitudinal pediatric studies, will we be able to answer. Three issues could be particularly critical for future research. First, to provide, if existing, a distinction between prepuberal STRA and puberal STRA endotypes to understand the transition from pediatric to adult STRA and to design effective, tailored therapies in adolescents, usually suffering from poorer asthma control. Second, design early treatments for pediatric airway remodeling to preserve lifelong good lung function. Finally, to better characterize inflammation before and during biological therapies, to provide clues on whether to stop or change treatments.
Collapse
Affiliation(s)
- Beatrice Andrenacci
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Maria De Filippo
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Martina Votto
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Maria Sole Prevedoni Gorone
- Pediatric Radiology Unit, Department of Diagnostic and Interventional Radiology and Neuroradiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Mara De Amici
- Immuno-Allergology Laboratory, Clinical Chemistry Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Stefania La Grutta
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
| | - Gian Luigi Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Amelia Licari
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Palermo, Italy
| |
Collapse
|
22
|
Jacenik D, Hikisz P, Beswick EJ, Fichna J. The clinical relevance of the adhesion G protein-coupled receptor F5 for human diseases and cancers. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166683. [PMID: 36878303 PMCID: PMC10164118 DOI: 10.1016/j.bbadis.2023.166683] [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: 11/30/2022] [Revised: 01/30/2023] [Accepted: 02/28/2023] [Indexed: 03/07/2023]
Abstract
Among the numerous adhesion G protein-coupled receptors (GPCRs), adhesion G protein-coupled estrogen receptor F5 (ADGRF5) contains unique domains in the long N-terminal tail which can determine cell-cell and cell-matrix interaction as well as cell adhesion. Nevertheless, the biology of ADGRF5 is complex and still poorly explored. Accumulating evidence suggests that the ADGRF5 activity is fundamental in health and disease. For instance, ADGRF5 is essential in the proper function of lungs and kidney as well as the endocrine system, and its signification in vascularization and tumorigenesis has been demonstrated. The most recent studies have provided findings about the diagnostic potential of ADGRF5 in osteoporosis and cancers, and ongoing studies suggest other diseases as well. Here, we elaborate on the current state of knowledge about the ADGRF5 in the physiology and pathophysiology of human diseases and highlight its high potential as a novel target in various therapeutic areas.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| | - Pawel Hikisz
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| | - Ellen J Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States.
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
23
|
Gopallawa I, Dehinwal R, Bhatia V, Gujar V, Chirmule N. A four-part guide to lung immunology: Invasion, inflammation, immunity, and intervention. Front Immunol 2023; 14:1119564. [PMID: 37063828 PMCID: PMC10102582 DOI: 10.3389/fimmu.2023.1119564] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/09/2023] [Indexed: 04/03/2023] Open
Abstract
Lungs are important respiratory organs primarily involved in gas exchange. Lungs interact directly with the environment and their primary function is affected by several inflammatory responses caused by allergens, inflammatory mediators, and pathogens, eventually leading to disease. The immune architecture of the lung consists of an extensive network of innate immune cells, which induce adaptive immune responses based on the nature of the pathogen(s). The balance of immune responses is critical for maintaining immune homeostasis in the lung. Infection by pathogens and physical or genetic dysregulation of immune homeostasis result in inflammatory diseases. These responses culminate in the production of a plethora of cytokines such as TSLP, IL-9, IL-25, and IL-33, which have been implicated in the pathogenesis of several inflammatory and autoimmune diseases. Shifting the balance of Th1, Th2, Th9, and Th17 responses have been the targets of therapeutic interventions in the treatment of these diseases. Here, we have briefly reviewed the innate and adaptive i3mmune responses in the lung. Genetic and environmental factors, and infection are the major causes of dysregulation of various functions of the lung. We have elaborated on the impact of inflammatory and infectious diseases, advances in therapies, and drug delivery devices on this critical organ. Finally, we have provided a comprehensive compilation of different inflammatory and infectious diseases of the lungs and commented on the pros and cons of different inhalation devices for the management of lung diseases. The review is intended to provide a summary of the immunology of the lung, with an emphasis on drug and device development.
Collapse
Affiliation(s)
- Indiwari Gopallawa
- Clinical Pharmacology & Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Ruchika Dehinwal
- Department of Microbiology, Division of Infectious Disease, Brigham Women’s Hospital, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, United States
| | | | - Vikramsingh Gujar
- Department of Anatomy and Cell Biology, Oklahoma State University Center for Health Sciences, Tulsa, OK, United States
| | - Narendra Chirmule
- R&D Department, SymphonyTech Biologics, Philadelphia, PA, United States
- *Correspondence: Narendra Chirmule,
| |
Collapse
|
24
|
Molecular Characterization and Selection of Indigenous SARS-CoV-2 Delta Variant for the Development of the First Inactivated SARS-CoV-2 Vaccine of Pakistan. Vaccines (Basel) 2023; 11:vaccines11030607. [PMID: 36992191 DOI: 10.3390/vaccines11030607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Vaccines are one of the efficient means available so far for preventing and controlling the infection rate of COVID-19. Several researchers have focused on the whole virus’s (SARS-CoV-2) inactivated vaccines which are economically efficient to produce. In Pakistan, multiple variants of SARS-CoV-2 have been reported since the start of the pandemic in February 2020. Due to the continuous evolution of the virus and economic recessions, the present study was designed to develop an indigenous inactivated SARS-CoV-2 vaccine that might help not only to prevent the COVID-19 in Pakistan, it will also save the country’s economic resources. The SARS-CoV-2 were isolated and characterized using the Vero-E6 cell culture system. The seed selection was carried out using cross-neutralization assay and phylogenetic analysis. The selected isolate of SARS-CoV-2 (hCoV-19/Pakistan/UHSPK3-UVAS268/2021) was inactivated using beta-propiolactone followed by vaccine formulation using Alum adjuvant, keeping the S protein concentration as 5 μg/dose. The vaccine efficacy was evaluated by in vivo immunogenicity testing in laboratory animals and in in vitro microneutralization test. The phylogenetic analysis revealed that all the SARS-CoV-2 isolates reported from Pakistan nested into different clades, representing multiple introductions of the virus into Pakistan. The antisera raised against various isolates from different waves in Pakistan showed a varied level of neutralization titers. However, the antisera produced against a variant (hCoV-19/Pakistan/UHSPK3-UVAS268/2021; fourth wave) efficiently neutralized (1:64–1:512) all the tested SARS-CoV-2 isolates. The inactivated whole virus vaccine of SARS-CoV-2 was safe and it also elicited a protective immune response in rabbits and rhesus macaques on the 35th-day post-vaccination. The activity of neutralizing antibodies of vaccinated animals was found at 1:256–1:1024 at 35 days post-vaccination, indicating the effectiveness of the double-dose regime of the indigenous SARS-CoV-2 vaccine.
Collapse
|
25
|
Kennedy K, Khaddour K, Ramnath N, Weinberg F. The Lung Microbiome in Carcinogenesis and Immunotherapy Treatment. Cancer J 2023; 29:61-69. [PMID: 36957975 DOI: 10.1097/ppo.0000000000000644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023]
Abstract
ABSTRACT Lung cancer is the leading cause of cancer-related deaths. Over the past 10 years, significant advances in treatment modalities, including immune checkpoint inhibitor (ICI) blockade, have led to improved outcomes. Elucidating predicative biomarkers in responders and nonresponders to ICI will lead to development of therapeutic targets that could enhance ICI efficacy. Recently, the gut microbiome was identified as a predictive biomarker for ICI in patients with multiple cancer types. However, it is unclear how other host microbiomes influence tumorigenesis and response to ICI. Other groups have explored the lung microbiome as it relates to carcinogenesis and immunotherapy efficacy. In this review, we explore the role of the lung microbiome in health and disease. We also review the current state of lung microbiome research as it relates to tumorigenesis and treatments and provide potential insights into how the lung microbiome could improve outcomes in patients with cancer.
Collapse
Affiliation(s)
- Kathleen Kennedy
- From the Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Karam Khaddour
- From the Department of Medicine, University of Illinois Chicago, Chicago, IL
| | | | - Frank Weinberg
- From the Department of Medicine, University of Illinois Chicago, Chicago, IL
| |
Collapse
|
26
|
Pyung YJ, Park DJ, Kim CG, Yun CH. Remodeling and Restraining Lung Tissue Damage Through the Regulation of Respiratory Immune Responses. Tissue Eng Regen Med 2023; 20:329-339. [PMID: 36763280 PMCID: PMC9913030 DOI: 10.1007/s13770-022-00516-7] [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: 11/15/2022] [Revised: 12/13/2022] [Accepted: 12/26/2022] [Indexed: 02/11/2023] Open
Abstract
Tissue damage caused by various stimuli under certain conditions, such as biological and environmental cues, can actively induce systemic and/or local immune responses. Therefore, understanding the immunological perspective would be critical to not only regulating homeostasis of organs and tissues but also to restrict and remodel their damage. Lungs serve as one of the key immunological organs, and thus, in the present article, we focus on the innate and adaptive immune systems involved in remodeling and engineering lung tissue. Innate immune cells are known to react immediately to damage. Macrophages, one of the most widely studied types of innate immune cells, are known to be involved in tissue damage and remodeling, while type 2 innate lymphoid cells (ILC2s) have recently been revealed as an important cell type responsible for tissue remodeling. On the other hand, adaptive immune cells are also involved in damage control. In particular, resident memory T cells in the lung prevent prolonged disease that causes tissue damage. In this review, we first outlined the structure of the respiratory system with biological and environmental cues and the innate/adaptive immune responses in the lung. It is our hope that understanding an immunological perspective for tissue remodeling and damage control in the lung will be beneficial for stakeholders in this area.
Collapse
Affiliation(s)
- Young Jin Pyung
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Da-Jeong Park
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol Gyun Kim
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- Center for Food and Bioconvergence, Seoul National University, Seoul, 08826, Republic of Korea.
- Institutes of Green-Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-Do, 25354, Republic of Korea.
- Interdisciplinary Programs in Agricultural Genomics, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
27
|
Ataide MA, Manin GZ, Oliveira SS, Guerra RO, Zamboni DS. Inflammasome activation and CCR2-mediated monocyte-derived dendritic cell recruitment restrict Legionella pneumophila infection. Eur J Immunol 2023; 53:e2249985. [PMID: 36427489 DOI: 10.1002/eji.202249985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/22/2022] [Accepted: 11/24/2022] [Indexed: 11/27/2022]
Abstract
Flagellin-induced NAIP/NLRC4 inflammasome activation and pyroptosis are critical events restricting Legionella pneumophila infection. However, the cellular and molecular dynamics of the in vivo responses against this bacterium are still unclear. We have found temporal coordination of two independent innate immunity pathways in controlling Legionella infection, the inflammasome activation and the CCR2-mediated Mo-DC recruitment. Inflammasome activation was an important player at the early stage of infection by lowering the numbers of bacteria for an efficient bacterial clearance conferred by the Mo-DC at the late stage of the infection. Mo-DC emergence highly depended on CCR2-signaling and dispensed inflammasome activation and pyroptosis. Also, Mo-DC compartment did not rely on the inflammasome machinery to deliver proper immune responses and was the most abundant cytokine-producing among the monocyte-derived cells in the infected lung. Importantly, when the CCR2- and NLRC4-dependent axes of response were simultaneously ablated, we observed an aggravated bacterial burden in the lung of infected mice. Taken together, we showed that inflammasome activation and CCR2-mediated immune response interplay in distinct pathways to restrict pulmonary bacterial infection. These findings extend our understanding of the in vivo integration and cooperation of different innate immunity arms in controlling infectious agents.
Collapse
Affiliation(s)
- Marco A Ataide
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Graziele Z Manin
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Samuel S Oliveira
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rhanoica O Guerra
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Dario S Zamboni
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
28
|
Rothen-Rutishauser B, Gibb M, He R, Petri-Fink A, Sayes CM. Human lung cell models to study aerosol delivery - considerations for model design and development. Eur J Pharm Sci 2023; 180:106337. [PMID: 36410570 DOI: 10.1016/j.ejps.2022.106337] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Human lung tissue models range from simple monolayer cultures to more advanced three-dimensional co-cultures. Each model system can address the interactions of different types of aerosols and the choice of the model and the mode of aerosol exposure depends on the relevant scenario, such as adverse outcomes and endpoints of interest. This review focuses on the functional, as well as structural, aspects of lung tissue from the upper airway to the distal alveolar compartments as this information is relevant for the design of a model as well as how the aerosol properties determine the interfacial properties with the respiratory wall. The most important aspects on how to design lung models are summarized with a focus on (i) choice of appropriate scaffold, (ii) selection of cell types for healthy and diseased lung models, (iii) use of culture condition and assembly, (iv) aerosol exposure methods, and (v) endpoints and verification process. Finally, remaining challenges and future directions in this field are discussed.
Collapse
Affiliation(s)
- Barbara Rothen-Rutishauser
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland.
| | - Matthew Gibb
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA
| | - Ruiwen He
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- BioNanomaterials, Adolphe Merkle Institute, University Fribourg, Chemin des Verdiers 4 CH-1700, Fribourg, Switzerland
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, One Bear Place #97266, Waco, TX 76798-7266, USA.
| |
Collapse
|
29
|
Nagai J, Lin J, Boyce JA. Macrophage P2Y6 Receptor Signaling Selectively Activates NFATC2 and Suppresses Allergic Lung Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:2293-2303. [PMID: 36307120 PMCID: PMC9719840 DOI: 10.4049/jimmunol.2200452] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/05/2022] [Indexed: 12/14/2022]
Abstract
Innate immune responses to innocuous Ags can either prevent or facilitate adaptive type 2 allergic inflammation, but the mechanisms are incompletely understood. We now demonstrate that macrophage UDP-specific type 6 purinergic (P2Y6) receptors selectively activate NFATC2, a member of the NFAT family, to drive an innate IL-12/IFN-γ axis that prevents type 2 allergic inflammation. UDP priming potentiated IL-12p40 production in bone marrow-derived macrophages (BMMs) stimulated by the house dust mite Dermatophagoides farinae (Df) in a P2Y6-dependent manner. Inhibitions of phospholipase C, calcium increase, and calcineurin eliminated UDP-potentiated Df-induced IL-12p40 production. UDP specifically induced nuclear translocation of NFATC2, but not NFATC1 and NFATC3, in BMMs in a P2Y6-dependent manner. UDP-potentiated IL-12p40 production by BMMs and Df-induced IL-12p40 gene expression by alveolar macrophages were abrogated in cells from Nfatc2 knockout mice. Pulmonary transplantation of wild-type but not Nfatc2 knockout macrophages increased Df-induced IL-12 production and IFN-γ expression in P2ry6 fl/fl/Cre/+ recipient mice. Finally, Nfatc2 knockout mice showed significantly increased indices of type 2 immunopathology in response to Df challenge, similar to P2ry6 fl/fl/Cre/+ mice. Thus, macrophage P2Y6 receptor signaling selectively utilizes NFATC2 to potentiate an innate IL-12/IFN-γ axis, a potential mechanism that protects against inappropriate type 2 immune responses.
Collapse
Affiliation(s)
- Jun Nagai
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, MA
| | - Junrui Lin
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, MA
| | - Joshua A. Boyce
- Department of Medicine, Harvard Medical School, Boston, MA
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA; and
- Jeff and Penny Vinik Center for Allergic Disease Research, Boston, MA
| |
Collapse
|
30
|
Tanner L, Bergwik J, Bhongir RKV, Puthia M, Lång P, Ali MN, Welinder C, Önnerfjord P, Erjefält JS, Palmberg L, Andersson G, Egesten A. Tartrate resistant acid phosphatase 5 (TRAP5) mediates immune cell recruitment in a murine model of pulmonary bacterial infection. Front Immunol 2022; 13:1079775. [PMID: 36569898 PMCID: PMC9779928 DOI: 10.3389/fimmu.2022.1079775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction During airway infection, upregulation of proinflammatory cytokines and subsequent immune cell recruitment is essential to mitigate bacterial infection. Conversely, during prolonged and non-resolving airway inflammation, neutrophils contribute to tissue damage and remodeling. This occurs during diseases including cystic fibrosis (CF) and COPD where bacterial pathogens, not least Pseudomonas aeruginosa, contribute to disease progression through long-lasting infections. Tartrate-resistant acid phosphatase (TRAP) 5 is a metalloenzyme expressed by alveolar macrophages and one of its target substrates is the phosphoglycoprotein osteopontin (OPN). Methods We used a knockout mouse strain (Trap5-/-) and BALB/c-Tg (Rela-luc)31Xen mice paired with siRNA administration or functional protein add-back to elucidate the role of Trap5 during bacterial infection. In a series of experiments, Trap5-/- and wild-type control mice received intratracheal administration of P.aerugniosa (Xen41) or LPS, with mice monitored using intravital imaging (IVIS). In addition, multiplex cytokine immunoassays, flow cytometry, multispectral analyses, histological staining were performed. Results In this study, we found that Trap5-/- mice had impaired clearance of P. aeruginosa airway infection and reduced recruitment of immune cells (i.e. neutrophils and inflammatory macrophages). Trap5 knockdown using siRNA resulted in a decreased activation of the proinflammatory transcription factor NF-κB in reporter mice and a subsequent decrease of proinflammatory gene expression. Add-back experiments of enzymatically active TRAP5 to Trap5-/- mice restored immune cell recruitment and bacterial killing. In human CF lung tissue, TRAP5 of alveolar macrophages was detected in proximity to OPN to a higher degree than in normal lung tissue, indicating possible interactions. Discussion Taken together, the findings of this study suggest a key role for TRAP5 in modulating airway inflammation. This could have bearing in diseases such as CF and COPD where excessive neutrophilic inflammation could be targeted by pharmacological inhibitors of TRAP5.
Collapse
Affiliation(s)
- Lloyd Tanner
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Jesper Bergwik
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Ravi K. V. Bhongir
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Manoj Puthia
- Department of Dermatology and Venereology, Lund University and Skåne University Hospital, Lund, Sweden,Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Pernilla Lång
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mohamad N. Ali
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden
| | - Charlotte Welinder
- Swedish National Infrastructure for Biological Mass Spectrometry (BioMS), Lund University, Lund, Sweden
| | - Patrik Önnerfjord
- Molecular Skeletal Biology, Section for Rheumatology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Jonas S. Erjefält
- Unit of Airway Inflammation, Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Lena Palmberg
- Work Environment Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Göran Andersson
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Arne Egesten
- Respiratory Medicine, Allergology & Palliative Medicine, Department of Clinical Sciences Lund, Lund University and Skåne University Hospital, Lund, Sweden,*Correspondence: Arne Egesten,
| |
Collapse
|
31
|
Kulle A, Thanabalasuriar A, Cohen TS, Szydlowska M. Resident macrophages of the lung and liver: The guardians of our tissues. Front Immunol 2022; 13:1029085. [PMID: 36532044 PMCID: PMC9750759 DOI: 10.3389/fimmu.2022.1029085] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
Resident macrophages play a unique role in the maintenance of tissue function. As phagocytes, they are an essential first line defenders against pathogens and much of the initial characterization of these cells was focused on their interaction with viral and bacterial pathogens. However, these cells are increasingly recognized as contributing to more than just host defense. Through cytokine production, receptor engagement and gap junction communication resident macrophages tune tissue inflammatory tone, influence adaptive immune cell phenotype and regulate tissue structure and function. This review highlights resident macrophages in the liver and lung as they hold unique roles in the maintenance of the interface between the circulatory system and the external environment. As such, we detail the developmental origin of these cells, their contribution to host defense and the array of tools these cells use to regulate tissue homeostasis.
Collapse
Affiliation(s)
- Amelia Kulle
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | | | - Taylor S. Cohen
- Late Stage Development, Vaccines and Immune Therapies (V&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| | - Marta Szydlowska
- Bacteriology and Vaccine Discovery, Research and Early Development, Vaccines and Immune Therapies (V&I), BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, United States
| |
Collapse
|
32
|
Santopolo G, Clemente A, Rojo-Molinero E, Fernández S, Álvarez MC, Oliver A, de la Rica R. Improved cytometric analysis of untouched lung leukocytes by enzymatic liquefaction of sputum samples. Biol Proced Online 2022; 24:17. [PMID: 36396988 PMCID: PMC9673301 DOI: 10.1186/s12575-022-00181-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
Background Phenotyping sputum-resident leukocytes and evaluating their functional status are essential analyses for exploring the cellular basis of pathological processes in the lungs, and flow cytometry is widely recognized as the gold-standard technique to address them. However, sputum-resident leukocytes are found in respiratory samples which need to be liquefied prior to cytometric analysis. Traditional liquefying procedures involve the use of a reducing agent such as dithiothreitol (DTT) in temperature-controlled conditions, which does not homogenize respiratory samples efficiently and impairs cell viability and functionality. Methods Here we propose an enzymatic method that rapidly liquefies samples by means of generating O2 bubbles with endogenous catalase. Sputum specimens from patients with suspected pulmonary infection were treated with DTT, the enzymatic method or PBS. We used turbidimetry to compare the liquefaction degree and cell counts were determined using a hemocytometer. Finally, we conducted a comparative flow cytometry study for evaluating frequencies of sputum-resident neutrophils, eosinophils and lymphocytes and their activation status after liquefaction. Results Enzymatically treated samples were better liquefied than those treated with DTT or PBS, which resulted in a more accurate cytometric analysis. Frequencies of all cell subsets analyzed within liquefied samples were comparable between liquefaction methods. However, the gentle cell handling rendered by the enzymatic method improves cell viability and retains in vivo functional characteristics of sputum-resident leukocytes (with regard to HLA-DR, CD63 and CD11b expression). Conclusion In conclusion, the proposed enzymatic liquefaction method improves the cytometric analysis of respiratory samples and leaves the cells widely untouched for properly addressing functional analysis of lung leukocytes. Supplementary Information The online version contains supplementary material available at 10.1186/s12575-022-00181-z.
Collapse
|
33
|
Ryan KM, Siegler E. Pyogenic brain abscess associated with an incidental pulmonary arteriovenous malformation. BMJ Case Rep 2022; 15:e252794. [PMID: 36384884 PMCID: PMC9670921 DOI: 10.1136/bcr-2022-252794] [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] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arteriovenous malformations (PAVMs) are rare and often asymptomatic vascular anomalies that can be associated with serious neurological consequences due to right-to-left shunting. We report a case of a woman in her 80s without substantial medical history who presented with a headache, weakness and personality changes, and was found to have a pyogenic brain abscess requiring emergent neurosurgical evacuation. The abscess grew oral flora, suspected to have reached the brain via an incidentally discovered PAVM. With drainage and antibiotics, the patient achieved a full recovery and the PAVM was embolised. To our knowledge, this is the oldest presentation of a PAVM-associated brain abscess in the published literature. Older patients may present without the typical signs and symptoms of a given illness, which complicates accurate diagnosis and treatment. Primary care physicians can help facilitate timely care and positive clinical outcomes.
Collapse
Affiliation(s)
- Kara Morgan Ryan
- Internal Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Eugenia Siegler
- Geriatrics, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
34
|
Patel M, Mazumder R, Mishra R, Kant Kaushik K. Potential of Nanotechnology-based Formulations in Combating Pulmonary Infectious Diseases: A Current Scenario. Curr Pharm Des 2022; 28:3413-3427. [PMID: 36397631 DOI: 10.2174/1381612829666221116143138] [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: 04/20/2022] [Revised: 07/09/2022] [Accepted: 10/19/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Pulmonary microbial infection is mainly caused by microbes like atypical bacteria, viruses, and fungi, on both the upper and lower respiratory tracts. One of the demands of the present is the use of nanotechnology-based treatments to fight various lung infections. AIM The main aim of the study is to explore all pulmonary infectious diseases and to compare the advanced and novel treatment approaches with the conventional methods which are available to treat infections. METHODS This work sheds light on pulmonary infectious diseases with their conventional and present treatment approaches along with a focus on the advantageous roles of nano-based formulations. In the literature, it has been reported that the respiratory system is the key target of various infectious diseases which gives rise to various challenges in the treatment of pulmonary infections. RESULTS The present review article describes the global situation of pulmonary infections and the different strategies which are available for their management, along with their limitations. The article also highlights the advantages and different examples of nanoformulations currently combating the limitations of conventional therapies. CONCLUSION The content of the present article further reflects on the summary of recently published research and review works on pulmonary infections, conventional methods of treatment with their limitations, and the role of nano-based approaches to combat the existing infectious diseases which will jointly help the researchers to produce effective drug formulations with desired pharmacological activities.
Collapse
Affiliation(s)
- Manisha Patel
- Pharmacy Institute, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, Uttar Pradesh-201 306, India
| | - Rupa Mazumder
- Pharmacy Institute, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, Uttar Pradesh-201 306, India
| | - Rakhi Mishra
- Pharmacy Institute, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, Uttar Pradesh-201 306, India
| | - Kamal Kant Kaushik
- Pharmacy Institute, Noida Institute of Engineering and Technology, 19, Knowledge Park-II, Institutional Area, Greater Noida, Uttar Pradesh-201 306, India
| |
Collapse
|
35
|
Meoli A, Eickmeier O, Pisi G, Fainardi V, Zielen S, Esposito S. Impact of CFTR Modulators on the Impaired Function of Phagocytes in Cystic Fibrosis Lung Disease. Int J Mol Sci 2022; 23:12421. [PMID: 36293274 PMCID: PMC9604330 DOI: 10.3390/ijms232012421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Cystic fibrosis (CF), the most common genetically inherited disease in Caucasian populations, is a multi-systemic life-threatening autosomal recessive disorder caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. In 2012, the arrival of CFTR modulators (potentiators, correctors, amplifiers, stabilizers, and read-through agents) revolutionized the therapeutic approach to CF. In this review, we examined the physiopathological mechanism of chronic dysregulated innate immune response in the lungs of CF patients with pulmonary involvement with particular reference to phagocytes, critically analyzing the role of CFTR modulators in influencing and eventually restoring their function. Our literature review highlighted that the role of CFTR in the lungs is crucial not only for the epithelial function but also for host defense, with particular reference to phagocytes. In macrophages and neutrophils, the CFTR dysfunction compromises both the intricate process of phagocytosis and the mechanisms of initiation and control of inflammation which then reverberates on the epithelial environment already burdened by the chronic colonization of pathogens leading to irreversible tissue damage. In this context, investigating the impact of CFTR modulators on phagocytic functions is therefore crucial not only for explaining the underlying mechanisms of pleiotropic effects of these molecules but also to better understand the physiopathological basis of this disease, still partly unexplored, and to develop new complementary or alternative therapeutic approaches.
Collapse
Affiliation(s)
- Aniello Meoli
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Olaf Eickmeier
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Giovanna Pisi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Valentina Fainardi
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Stefan Zielen
- Division of Allergy, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, University Hospital, 60431 Frankfurt, Germany
| | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| |
Collapse
|
36
|
Hastak PS, Andersen CR, Kelleher AD, Sasson SC. Frontline workers: Mediators of mucosal immunity in community acquired pneumonia and COVID-19. Front Immunol 2022; 13:983550. [PMID: 36211412 PMCID: PMC9539803 DOI: 10.3389/fimmu.2022.983550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
The current COVID-19 pandemic has highlighted a need to further understand lung mucosal immunity to reduce the burden of community acquired pneumonia, including that caused by the SARS-CoV-2 virus. Local mucosal immunity provides the first line of defence against respiratory pathogens, however very little is known about the mechanisms involved, with a majority of literature on respiratory infections based on the examination of peripheral blood. The mortality for severe community acquired pneumonia has been rising annually, even prior to the current pandemic, highlighting a significant need to increase knowledge, understanding and research in this field. In this review we profile key mediators of lung mucosal immunity, the dysfunction that occurs in the diseased lung microenvironment including the imbalance of inflammatory mediators and dysbiosis of the local microbiome. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality.
Collapse
Affiliation(s)
- Priyanka S. Hastak
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Christopher R. Andersen
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
- Intensive Care Unit, Royal North Shore Hospital, Sydney, NSW, Australia
- Critical Care and Trauma Division, The George Institute for Global Health, Sydney, NSW, Australia
| | - Anthony D. Kelleher
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Sarah C. Sasson
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
37
|
Juarez VM, Montalbine AN, Singh A. Microbiome as an immune regulator in health, disease, and therapeutics. Adv Drug Deliv Rev 2022; 188:114400. [PMID: 35718251 PMCID: PMC10751508 DOI: 10.1016/j.addr.2022.114400] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/11/2022] [Accepted: 06/12/2022] [Indexed: 11/27/2022]
Abstract
New discoveries in drugs and drug delivery systems are focused on identifying and delivering a pharmacologically effective agent, potentially targeting a specific molecular component. However, current drug discovery and therapeutic delivery approaches do not necessarily exploit the complex regulatory network of an indispensable microbiota that has been engineered through evolutionary processes in humans or has been altered by environmental exposure or diseases. The human microbiome, in all its complexity, plays an integral role in the maintenance of host functions such as metabolism and immunity. However, dysregulation in this intricate ecosystem has been linked with a variety of diseases, ranging from inflammatory bowel disease to cancer. Therapeutics and bacteria have an undeniable effect on each other and understanding the interplay between microbes and drugs could lead to new therapies, or to changes in how existing drugs are delivered. In addition, targeting the human microbiome using engineered therapeutics has the potential to address global health challenges. Here, we present the challenges and cutting-edge developments in microbiome-immune cell interactions and outline novel targeting strategies to advance drug discovery and therapeutics, which are defining a new era of personalized and precision medicine.
Collapse
Affiliation(s)
- Valeria M Juarez
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Alyssa N Montalbine
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Ankur Singh
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, United States.
| |
Collapse
|
38
|
Stevenson ER, Wilkinson ML, Abramova E, Guo C, Gow AJ. Intratracheal Administration of Acyl Coenzyme A Acyltransferase-1 Inhibitor K-604 Reduces Pulmonary Inflammation Following Bleomycin-Induced Lung Injury. J Pharmacol Exp Ther 2022; 382:356-365. [PMID: 35970601 PMCID: PMC9426763 DOI: 10.1124/jpet.122.001284] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/28/2022] [Indexed: 01/19/2023] Open
Abstract
Acute lung injury (ALI) is characterized by epithelial damage, barrier dysfunction, and pulmonary edema. Macrophage activation and failure to resolve play a role in ALI; thus, macrophage phenotype modulation is a rational target for therapeutic intervention. Large, lipid-laden macrophages have been observed in various injury models, including intratracheal bleomycin (ITB), suggesting that lipid storage may play a role in ALI severity. The endoplasmic reticulum-associated enzyme acyl coenzyme A acyltransferase-1 (Acat-1/Soat1) is highly expressed in macrophages, where it catalyzes the esterification of cholesterol, leading to intracellular lipid accumulation. We hypothesize that inhibition of Acat-1 will reduce macrophage activation and improve outcomes of lung injury in ITB. K-604, a selective inhibitor of Acat-1, was used to reduce cholesterol esterification and hence lipid accumulation in response to ITB. Male and female C57BL6/J mice (n = 16-21/group) were administered control, control + K-604, ITB, or ITB + K-604 on d0, control or K-604 on d3, and were sacrificed on day 7. ITB caused significant body weight loss and an increase in cholesterol accumulation in bronchoalveolar lavage cells. These changes were mitigated by Acat-1 inhibition. K-604 also significantly reduced ITB-induced alveolar thickening. Surfactant composition was normalized as indicated by a significant decrease in phospholipid: SP-B ratio in ITB+K-604 compared with ITB. K-604 administration preserved mature alveolar macrophages, decreased activation in response to ITB, and decreased the percentage mature and pro-fibrotic interstitial macrophages. These results show that inhibition of Acat-1 in the lung is associated with reduced inflammatory response to ITB-mediated lung injury. SIGNIFICANCE STATEMENT: Acyl coenzyme A acyltransferase-1 (Acat-1) is critical to lipid droplet formation, and thus inhibition of Acat-1 presents as a pharmacological target. Intratracheal administration of K-604, an Acat-1 inhibitor, reduces intracellular cholesterol ester accumulation in lung macrophages, attenuates inflammation and macrophage activation, and normalizes mediators of surface-active function after intratracheal bleomycin administration in a rodent model. The data presented within suggest that inhibition of Acat-1 in the lung improves acute lung injury outcomes.
Collapse
Affiliation(s)
- Emily R Stevenson
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Melissa L Wilkinson
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Elena Abramova
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Changjiang Guo
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Andrew J Gow
- Ernest Mario School of Pharmacy, Department of Pharmacology & Toxicology, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| |
Collapse
|
39
|
Mifflin KA, Brennan FH, Guan Z, Kigerl KA, Filous AR, Mo X, Schwab JM, Popovich PG. Spinal Cord Injury Impairs Lung Immunity in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:157-170. [PMID: 35697382 PMCID: PMC9246940 DOI: 10.4049/jimmunol.2200192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/22/2022] [Indexed: 06/15/2023]
Abstract
Pulmonary infection is a leading cause of morbidity and mortality after spinal cord injury (SCI). Although SCI causes atrophy and dysfunction in primary and secondary lymphoid tissues with a corresponding decrease in the number and function of circulating leukocytes, it is unknown whether this SCI-dependent systemic immune suppression also affects the unique tissue-specific antimicrobial defense mechanisms that protect the lung. In this study, we tested the hypothesis that SCI directly impairs pulmonary immunity and subsequently increases the risk for developing pneumonia. Using mouse models of severe high-level SCI, we find that recruitment of circulating leukocytes and transcriptional control of immune signaling in the lung is impaired after SCI, creating an environment that is permissive for infection. Specifically, we saw a sustained loss of pulmonary leukocytes, a loss of alveolar macrophages at chronic time points postinjury, and a decrease in immune modulatory genes, especially cytokines, needed to eliminate pulmonary infections. Importantly, this injury-dependent impairment of pulmonary antimicrobial defense is only partially overcome by boosting the recruitment of immune cells to the lung with the drug AMD3100, a Food and Drug Administration-approved drug that mobilizes leukocytes and hematopoietic stem cells from bone marrow. Collectively, these data indicate that the immune-suppressive effects of SCI extend to the lung, a unique site of mucosal immunity. Furthermore, preventing lung infection after SCI will likely require novel strategies, beyond the use of orthodox antibiotics, to reverse or block tissue-specific cellular and molecular determinants of pulmonary immune surveillance.
Collapse
Affiliation(s)
- Katherine A Mifflin
- Department of Neuroscience, The Ohio State University, Columbus, OH
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
| | - Faith H Brennan
- Department of Neuroscience, The Ohio State University, Columbus, OH
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
| | - Zhen Guan
- Department of Neuroscience, The Ohio State University, Columbus, OH
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
| | - Kristina A Kigerl
- Department of Neuroscience, The Ohio State University, Columbus, OH
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
| | - Angela R Filous
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
- Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH; and
| | - Xiaokui Mo
- Department of Biomedical Informatics, The Ohio State University, Center for Biostatistics, Columbus, OH
| | - Jan M Schwab
- Department of Neuroscience, The Ohio State University, Columbus, OH
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
- Department of Neurology, The Ohio State University, Wexner Medical Center, Columbus, OH; and
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University, Columbus, OH;
- Belford Center for Spinal Cord Injury, The Ohio State University, Columbus, OH
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH
| |
Collapse
|
40
|
Griesser E, Schönberger T, Stierstorfer B, Wyatt H, Rist W, Lamla T, Thomas MJ, Lamb D, Geillinger-Kästle KE. Characterization of a flexible AAV-DTR/DT mouse model of acute epithelial lung injury. Am J Physiol Lung Cell Mol Physiol 2022; 323:L206-L218. [PMID: 35762632 DOI: 10.1152/ajplung.00364.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Animal models are important to mimic certain pathways or biological aspects of human pathologies including acute and chronic pulmonary diseases. We developed a novel and flexible mouse model of acute epithelial lung injury based on adeno-associated virus (AAV) variant 6.2 mediated expression of the human diphtheria toxin receptor (DTR). Following intratracheal administration of diphtheria toxin (DT), a cell-specific death of bronchial and alveolar epithelial cells can be observed. In contrast to other lung injury models, the here described mouse model provides the possibility of targeted injury using specific tropisms of AAV vectors or cell type specific promotors to drive the human DTR expression. Also, generation of cell specific mouse lines is not required. Detailed characterization of the AAV-DTR/DT mouse model including titration of viral genome (vg) load and administered DT amount revealed increasing cell numbers in bronchoalveolar lavage (BAL; macrophages, neutrophils, and unspecified cells) and elevation of degenerated cells and infiltrated leukocytes in lung tissue, dependent of vg load and DT dose. Cytokine levels in BAL fluid showed different patterns with higher vg load, e.g. IFNγ, TNFα, and IP10 increasing and IL-5 and IL-6 decreasing, while lung function was not affected. Additionally, laser-capture microdissection (LCM)-based proteomics of bronchial epithelium and alveolar tissue revealed upregulated immune and inflammatory response in all regions and extracellular matrix deposition in infiltrated alveoli. Overall, our novel AAV-DTR/DT model allows investigation of repair mechanisms following epithelial injury and resembles specific mechanistic aspects of acute and chronic pulmonary diseases.
Collapse
Affiliation(s)
- Eva Griesser
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Tanja Schönberger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Birgit Stierstorfer
- Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Hannah Wyatt
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Wolfgang Rist
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Thorsten Lamla
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Matthew James Thomas
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany.,University of Bath, Bath, United Kingdom
| | - David Lamb
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany
| | - Kerstin E Geillinger-Kästle
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
41
|
Flow Cytometric Analysis of Leukocyte Populations in the Lung Tissue of Dromedary Camels. Vet Sci 2022; 9:vetsci9060287. [PMID: 35737339 PMCID: PMC9231337 DOI: 10.3390/vetsci9060287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/23/2022] Open
Abstract
Respiratory tract infections are among the most common infections in dromedary camels, with a high impact on animal health, production, and welfare. Tissue-specific distribution of immune cells is one of the important factors that influence the nature and outcome of the immune response to pathogens. Several protocols have recently been described for the flow cytometric analysis of immune cells in the lung tissue of several species. However, no such protocol currently exists for dromedary camels. The aim of the present study was, therefore, to establish a flow cytometric protocol for the identification of immune cell populations in the camel lung tissue and the evaluation of some of their phenotypic and functional properties. Combined staining of camel lung leukocytes with monoclonal antibodies to the pan-leukocyte marker CD45 and the myeloid cell marker CD172a allowed the identification of myeloid cells (CD45+CD172a+) and lymphoid cells (CD45+CD172a−) in the lung of healthy camels. The cell adhesion molecules CD11a and CD18 were found in a higher abundance on myeloid cells compared to lymphoid cells. Based on their differential expression of the LPS receptor CD14, macrophages (CD172a+CD14high cells) were identified as the most abundant immune cell population in the camel lung tissue. In contrast to their dominance in camel peripheral blood, granulocytes (CD172a+CD14low) presented only a minor population in the lung tissue. The higher frequency of γδ T cells in the lung tissue than in peripheral blood suggests a role for these cells in the pulmonary immune system. Flow cytometric analysis of bacterial phagocytosis and ROS production upon bacterial stimulation revealed high antimicrobial activity of camel lung phagocytes, which was comparable with the antimicrobial activity of blood granulocytes. Comparative analysis of immune cell distribution between the cranial and caudal lobes of the camel lung revealed a higher frequency of granulocytes and a lower frequency of macrophages in the cranial compared to the caudal lung lobe. In addition, the higher frequency of cells expressing the M2 macrophage marker CD163 in the caudal lung tissue, with a slightly higher fraction of MHCII-positive cells (M1 phenotype) in the cranial lung tissue, may suggest the distribution of different macrophage subtypes in the different lobes of the camel lung. Such differences between lung lobes could influence the effectiveness of the immune response to infection or vaccination with respiratory pathogens. Collectively, the present study identified some similarities and differences between camels and other farm animals regarding the distribution of the main immune cell populations in their lungs. Further studies are required for comprehensive immunophenotyping of the cellular pulmonary immune system in camels.
Collapse
|
42
|
Lu JY, Simon M, Zhao Y, Ablaeva J, Corson N, Choi Y, Yamada KYH, Schork NJ, Hood WR, Hill GE, Miller RA, Seluanov A, Gorbunova V. Comparative transcriptomics reveals circadian and pluripotency networks as two pillars of longevity regulation. Cell Metab 2022; 34:836-856.e5. [PMID: 35580607 PMCID: PMC9364679 DOI: 10.1016/j.cmet.2022.04.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/24/2022] [Accepted: 04/22/2022] [Indexed: 01/24/2023]
Abstract
Mammals differ more than 100-fold in maximum lifespan. Here, we conducted comparative transcriptomics on 26 species with diverse lifespans. We identified thousands of genes with expression levels negatively or positively correlated with a species' maximum lifespan (Neg- or Pos-MLS genes). Neg-MLS genes are primarily involved in energy metabolism and inflammation. Pos-MLS genes show enrichment in DNA repair, microtubule organization, and RNA transport. Expression of Neg- and Pos-MLS genes is modulated by interventions, including mTOR and PI3K inhibition. Regulatory networks analysis showed that Neg-MLS genes are under circadian regulation possibly to avoid persistent high expression, whereas Pos-MLS genes are targets of master pluripotency regulators OCT4 and NANOG and are upregulated during somatic cell reprogramming. Pos-MLS genes are highly expressed during embryogenesis but significantly downregulated after birth. This work provides targets for anti-aging interventions by defining pathways correlating with longevity across mammals and uncovering circadian and pluripotency networks as central regulators of longevity.
Collapse
Affiliation(s)
- J Yuyang Lu
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew Simon
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Yang Zhao
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Julia Ablaeva
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Nancy Corson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Yongwook Choi
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - KayLene Y H Yamada
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Nicholas J Schork
- Quantitative Medicine and Systems Biology Division, Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Wendy R Hood
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Geoffrey E Hill
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY 14627, USA.
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
43
|
Porto GD, Haupenthal DPDS, Souza PS, Silveira GDB, Nesi RT, Feuser PE, Possato JC, de Andrade VM, Pinho RA, Silveira PCL. Effects of the intranasal application of gold nanoparticles on the pulmonary tissue after acute exposure to industrial cigarette smoke. J Biomed Mater Res B Appl Biomater 2022; 110:1234-1244. [PMID: 34894049 DOI: 10.1002/jbm.b.34994] [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: 03/17/2021] [Revised: 11/15/2021] [Accepted: 12/05/2021] [Indexed: 11/09/2022]
Abstract
Inhalation of harmful particles appears as a primary factor for the onset and establishment of chronic obstructive pulmonary disease (COPD). Cigarette smoke acutely promotes an exacerbated inflammatory response with oxidative stress induction with DNA damage. Administration of Gold Nanoparticles (GNPs) with 20 nm in different concentrations can revert damages caused by external aggravations. The effects of GNPs in a COPD process have not been observed until now. The objective of this work was to evaluate the therapeutic effects of intranasal administration of different doses of GNPs after acute exposure to industrial cigarette smoke. Thirty male Swiss mice were randomly divided into five groups: Sham; cigarette smoke (CS); CS + GNPs 2.5 mg/L; CS + GNPs 7.5 mg/L and CS + GNPs 22.5 mg/L. The animals were exposed to the commercial cigarette with filter in an acrylic inhalation chamber and treated with intranasal GNPs for five consecutive days. The results demonstrate that exposure to CS causes an increase in inflammatory cytokines, histological changes, oxidative and nitrosive damage in the lung, as well as increased damage to the DNA of liver cells, blood plasma and lung. Among the three doses of GNPs (2.5, 7.5, and 22.5 mg/L) used, the highest dose had better anti-inflammatory effects. However, GNPs at a dose of 7.5 mg/L showed better efficacies in reducing ROS formation, alveolar diameter, and the number of inflammatory cells in histology, in addition to significantly reduced rate of DNA damage in lung cells without additional systemic genotoxicity already caused by cigarette smoke.
Collapse
Affiliation(s)
- Germano Duarte Porto
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | | | - Priscila Soares Souza
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Gustavo de Bem Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Renata Tiscoski Nesi
- Biochemistry in Health, Graduate Program in Health Sciences, Medicine School, Pontifícia Universidade Católica do Paraná, Puerto Rico, Brazil
| | - Paulo Emilio Feuser
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Jonathann Corrêa Possato
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Vanessa Moraes de Andrade
- Translational Biomedicine Laboratory, Graduate Program of Health Sciences, Department of Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Ricardo Aurino Pinho
- Biochemistry in Health, Graduate Program in Health Sciences, Medicine School, Pontifícia Universidade Católica do Paraná, Puerto Rico, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| |
Collapse
|
44
|
Palmer CS, Kimmey JM. Neutrophil Recruitment in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2022; 12:894644. [PMID: 35646729 PMCID: PMC9136017 DOI: 10.3389/fcimb.2022.894644] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 04/18/2022] [Indexed: 01/19/2023] Open
Abstract
Streptococcus pneumoniae (Spn) is the primary agent of community-acquired pneumonia. Neutrophils are innate immune cells that are essential for bacterial clearance during pneumococcal pneumonia but can also do harm to host tissue. Neutrophil migration in pneumococcal pneumonia is therefore a major determinant of host disease outcomes. During Spn infection, detection of the bacterium leads to an increase in proinflammatory signals and subsequent expression of integrins and ligands on both the neutrophil as well as endothelial and epithelial cells. These integrins and ligands mediate the tethering and migration of the neutrophil from the bloodstream to the site of infection. A gradient of host-derived and bacterial-derived chemoattractants contribute to targeted movement of neutrophils. During pneumococcal pneumonia, neutrophils are rapidly recruited to the pulmonary space, but studies show that some of the canonical neutrophil migratory machinery is dispensable. Investigation of neutrophil migration is necessary for us to understand the dynamics of pneumococcal infection. Here, we summarize what is known about the pathways that lead to migration of the neutrophil from the capillaries to the lung during pneumococcal infection.
Collapse
Affiliation(s)
| | - Jacqueline M. Kimmey
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
45
|
Melo-González F, Sepúlveda-Alfaro J, Schultz BM, Suazo ID, Boone DL, Kalergis AM, Bueno SM. Distal Consequences of Mucosal Infections in Intestinal and Lung Inflammation. Front Immunol 2022; 13:877533. [PMID: 35572549 PMCID: PMC9095905 DOI: 10.3389/fimmu.2022.877533] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/28/2022] [Indexed: 11/17/2022] Open
Abstract
Infectious diseases are one of the leading causes of morbidity and mortality worldwide, affecting high-risk populations such as children and the elderly. Pathogens usually activate local immune responses at the site of infection, resulting in both protective and inflammatory responses, which may lead to local changes in the microbiota, metabolites, and the cytokine environment. Although some pathogens can disseminate and cause systemic disease, increasing evidence suggests that local infections can affect tissues not directly invaded. In particular, diseases occurring at distal mucosal barriers such as the lung and the intestine seem to be linked, as shown by epidemiological studies in humans. These mucosal barriers have bidirectional interactions based mainly on multiple signals derived from the microbiota, which has been termed as the gut-lung axis. However, the effects observed in such distal places are still incompletely understood. Most of the current research focuses on the systemic impact of changes in microbiota and bacterial metabolites during infection, which could further modulate immune responses at distal tissue sites. Here, we describe how the gut microbiota and associated metabolites play key roles in maintaining local homeostasis and preventing enteric infection by direct and indirect mechanisms. Subsequently, we discuss recent murine and human studies linking infectious diseases with changes occurring at distal mucosal barriers, with particular emphasis on bacterial and viral infections affecting the lung and the gastrointestinal tract. Further, we discuss the potential mechanisms by which pathogens may cause such effects, promoting either protection or susceptibility to secondary infection.
Collapse
Affiliation(s)
- Felipe Melo-González
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara M. Schultz
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Isidora D. Suazo
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David L. Boone
- Department of Microbiology and Immunology, Indiana University School of Medicine-South Bend, South Bend, IN, United States
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
46
|
Anderson S, Atkins P, Bäckman P, Cipolla D, Clark A, Daviskas E, Disse B, Entcheva-Dimitrov P, Fuller R, Gonda I, Lundbäck H, Olsson B, Weers J. Inhaled Medicines: Past, Present, and Future. Pharmacol Rev 2022; 74:48-118. [PMID: 34987088 DOI: 10.1124/pharmrev.120.000108] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/06/2021] [Indexed: 12/21/2022] Open
Abstract
The purpose of this review is to summarize essential pharmacological, pharmaceutical, and clinical aspects in the field of orally inhaled therapies that may help scientists seeking to develop new products. After general comments on the rationale for inhaled therapies for respiratory disease, the focus is on products approved approximately over the last half a century. The organization of these sections reflects the key pharmacological categories. Products for asthma and chronic obstructive pulmonary disease include β -2 receptor agonists, muscarinic acetylcholine receptor antagonists, glucocorticosteroids, and cromones as well as their combinations. The antiviral and antibacterial inhaled products to treat respiratory tract infections are then presented. Two "mucoactive" products-dornase α and mannitol, which are both approved for patients with cystic fibrosis-are reviewed. These are followed by sections on inhaled prostacyclins for pulmonary arterial hypertension and the challenging field of aerosol surfactant inhalation delivery, especially for prematurely born infants on ventilation support. The approved products for systemic delivery via the lungs for diseases of the central nervous system and insulin for diabetes are also discussed. New technologies for drug delivery by inhalation are analyzed, with the emphasis on those that would likely yield significant improvements over the technologies in current use or would expand the range of drugs and diseases treatable by this route of administration. SIGNIFICANCE STATEMENT: This review of the key aspects of approved orally inhaled drug products for a variety of respiratory diseases and for systemic administration should be helpful in making judicious decisions about the development of new or improved inhaled drugs. These aspects include the choices of the active ingredients, formulations, delivery systems suitable for the target patient populations, and, to some extent, meaningful safety and efficacy endpoints in clinical trials.
Collapse
Affiliation(s)
- Sandra Anderson
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Paul Atkins
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Per Bäckman
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - David Cipolla
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Andrew Clark
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Evangelia Daviskas
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Bernd Disse
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Plamena Entcheva-Dimitrov
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Rick Fuller
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Igor Gonda
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Hans Lundbäck
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Bo Olsson
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| | - Jeffry Weers
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia (S.A.); Inhaled Delivery Solutions LLC, Durham, North Carolina (P.A.); Emmace Consulting AB Medicon Village, Lund, Sweden (P.B., H.L., B.O.); Insmed Inc., Bridgewater, New Jersey (D.C.); Aerogen Pharma Corporation, San Mateo, California (A.C.); Woolcock Institute of Medical Research, Glebe, New South Wales, Australia (E.D.); Drug Development, Pharmacology and Clinical Pharmacology Consulting, Mainz, Germany (B.D.); Preferred Regulatory Consulting, San Mateo, California (P.E-.D.); Clayton, CA (R.F.); Respidex LLC, Dennis, Massachusetts (I.G.); and cystetic Medicines, Inc., Burlingame, California (J.W.)
| |
Collapse
|
47
|
Tang W, Zhang Y, Zhu G. Pulmonary delivery of mucosal nanovaccines. NANOSCALE 2022; 14:263-276. [PMID: 34918733 PMCID: PMC8734613 DOI: 10.1039/d1nr06512b] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 11/29/2021] [Indexed: 06/14/2023]
Abstract
Mucosal vaccination can elicit both systemic and mucosal immunity, and therefore has the potential to not only treat mucosal immune diseases, prevent the pathogen infection at the mucosal entry sites, but also treat distant or systemic immune disorders. However, only a few mucosal vaccines have been approved for human use in the clinic. Effective mucosal immunization requires the delivery of immunogenic agents to appropriate mucosal surfaces, which remains significantly challenging due to the essential biological barriers presenting at mucosal tissues. In the past decade, remarkable progress has been made in the development of pulmonary mucosal nanovaccines. The nanovaccines leverage advanced nanoparticle-based pulmonary delivery technologies on the characteristics of large surface area and rich antigen presentation cell environment of the lungs for triggering robust immune protection against various mucosal diseases. Herein, we review current methods and formulations of pulmonary delivery, discuss the design strategies of mucosal nanovaccines for potent and long-lasting immune responses, and highlight recent advances in the application of lipid-based pulmonary nanovaccines against mucosal diseases. These advances promise to accelerate the development of novel mucosal nanovaccines for the prophylaxis and therapy of infectious diseases, and cancer, as well as autoimmune disorders at mucosal tissues.
Collapse
Affiliation(s)
- Wei Tang
- Department of Pharmacy and Department of Diagnostic Radiology, Faculty of Science and Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore.
| | - Yu Zhang
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Richmond, VA 23298, USA.
| | - Guizhi Zhu
- Department of Pharmaceutics and Center for Pharmaceutical Engineering and Sciences, Institute for Structural Biology and Drug Discovery, School of Pharmacy, The Developmental Therapeutics Program, Massey Cancer Center, Richmond, VA 23298, USA.
| |
Collapse
|
48
|
Wang Y, Chang RYK, Britton WJ, Chan HK. Advances in the development of antimicrobial peptides and proteins for inhaled therapy. Adv Drug Deliv Rev 2022; 180:114066. [PMID: 34813794 DOI: 10.1016/j.addr.2021.114066] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 12/13/2022]
Abstract
Antimicrobial peptides and proteins (APPs) are becoming increasingly important in targeting multidrug-resistant (MDR) bacteria. APPs is a rapidly emerging area with novel molecules being produced and further optimised to enhance antimicrobial efficacy, while overcoming issues associated with biologics such as potential toxicity and low bioavailability resulting from short half-life. Inhalation delivery of these agents can be an effective treatment of respiratory infections owing to the high local drug concentration in the lungs with lower exposure to systemic circulation hence reducing systemic toxicity. This review describes the recent studies on inhaled APPs, including in vitro and in vivo antimicrobial activities, toxicity assessments, and formulation strategies whenever available. The review also includes studies on combination of APPs with other antimicrobial agents to achieve enhanced synergistic antimicrobial effect. Since different APPs have different biological and chemical stabilities, a targeted formulation strategy should be considered for developing stable and inhalable antimicrobial peptides and proteins. These strategies include the use of sodium chloride to reduce electrostatic interaction between APP and extracellular DNA in sputum, the use of D-enantiomers or dendrimers to minimise protease-mediated degradation and or the use of prodrugs to reduce toxicity. Although great effort has been put towards optimising the biological functions of APPs, studies assessing biological stability in inhalable aerosols are scarce, particularly for novel molecules. As such, formulation and manufacture of inhalable liquid and powder formulations of APPs are underexplored, yet they are crucial areas of research for clinical translation.
Collapse
|
49
|
Feyaerts D, Urbschat C, Gaudillière B, Stelzer IA. Establishment of tissue-resident immune populations in the fetus. Semin Immunopathol 2022; 44:747-766. [PMID: 35508672 PMCID: PMC9067556 DOI: 10.1007/s00281-022-00931-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/17/2022] [Indexed: 12/15/2022]
Abstract
The immune system establishes during the prenatal period from distinct waves of stem and progenitor cells and continuously adapts to the needs and challenges of early postnatal and adult life. Fetal immune development not only lays the foundation for postnatal immunity but establishes functional populations of tissue-resident immune cells that are instrumental for fetal immune responses amidst organ growth and maturation. This review aims to discuss current knowledge about the development and function of tissue-resident immune populations during fetal life, focusing on the brain, lung, and gastrointestinal tract as sites with distinct developmental trajectories. While recent progress using system-level approaches has shed light on the fetal immune landscape, further work is required to describe precise roles of prenatal immune populations and their migration and adaptation to respective organ environments. Defining points of prenatal susceptibility to environmental challenges will support the search for potential therapeutic targets to positively impact postnatal health.
Collapse
Affiliation(s)
- Dorien Feyaerts
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| | - Christopher Urbschat
- grid.13648.380000 0001 2180 3484Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Brice Gaudillière
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Ina A. Stelzer
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| |
Collapse
|
50
|
Souza TN, Valdez AF, Rizzo J, Zamith-Miranda D, Guimarães AJ, Nosanchuk JD, Nimrichter L. Host cell membrane microdomains and fungal infection. Cell Microbiol 2021; 23:e13385. [PMID: 34392593 PMCID: PMC8664998 DOI: 10.1111/cmi.13385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/14/2021] [Accepted: 07/24/2021] [Indexed: 01/13/2023]
Abstract
Lipid microdomains or lipid rafts are dynamic and tightly ordered regions of the plasma membrane. In mammalian cells, they are enriched in cholesterol, glycosphingolipids, Glycosylphosphatidylinositol-anchored and signalling-related proteins. Several studies have suggested that mammalian pattern recognition receptors are concentrated or recruited to lipid domains during host-pathogen association to enhance the effectiveness of host effector processes. However, pathogens have also evolved strategies to exploit these domains to invade cells and survive. In fungal organisms, a complex cell wall network usually mediates the first contact with the host cells. This cell wall may contain virulence factors that interfere with the host membrane microdomains dynamics, potentially impacting the infection outcome. Indeed, the microdomain disruption can dampen fungus-host cell adhesion, phagocytosis and cellular immune responses. Here, we provide an overview of regulatory strategies employed by pathogenic fungi to engage with and potentially subvert the lipid microdomains of host cells. TAKE AWAY: Lipid microdomains are ordered regions of the plasma membrane enriched in cholesterol, glycosphingolipids (GSL), GPI-anchored and signalling-related proteins. Pathogen recognition by host immune cells can involve lipid microdomain participation. During this process, these domains can coalesce in larger complexes recruiting receptors and signalling proteins, significantly increasing their signalling abilities. The antifungal innate immune response is mediated by the engagement of pathogen-associated molecular patterns to pattern recognition receptors (PRRs) at the plasma membrane of innate immune cells. Lipid microdomains can concentrate or recruit PRRs during host cell-fungi association through a multi-interactive mechanism. This association can enhance the effectiveness of host effector processes. However, virulence factors at the fungal cell surface and extracellular vesicles can re-assembly these domains, compromising the downstream signalling and favouring the disease development. Lipid microdomains are therefore very attractive targets for novel drugs to combat fungal infections.
Collapse
Affiliation(s)
- Taiane N Souza
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro F Valdez
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juliana Rizzo
- Unité Biologie des ARN des Pathogènes Fongiques, Département de Mycologie, Institut Pasteur, Paris, France
| | - Daniel Zamith-Miranda
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Allan Jefferson Guimarães
- Departamento de Microbiologia e Parasitologia-MIP, Instituto Biomédico, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Leonardo Nimrichter
- Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|