1
|
Driesschaert B, Mergan L, Lucci C, Simon C, Santos D, De Groef L, Temmerman L. The role of phagocytic cells in aging: insights from vertebrate and invertebrate models. Biogerontology 2024; 25:1301-1314. [PMID: 39168928 DOI: 10.1007/s10522-024-10131-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
While the main role of phagocytic scavenger cells consists of the neutralization and elimination of pathogens, they also keep the body fluids clean by taking up and breaking down waste material. Since a build-up of waste is thought to contribute to the aging process, these cells become particularly pertinent in the research field of aging. Nevertheless, a direct link between their scavenging functions and the aging process has yet to be established. Integrative approaches involving various model organisms hold promise to elucidate this potential, but are lagging behind since the diversity and evolutionary relationship of these cells across animal species remain unclear. In this perspective, we review the current knowledge associating phagocytic scavenger cells with aging in vertebrate and invertebrate animals, as well as put forward important questions for further exploration. Additionally, we highlight future challenges and propose a constructive approach for tackling them.
Collapse
Affiliation(s)
- Brecht Driesschaert
- Molecular and Functional Neurobiology, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Lucas Mergan
- Molecular and Functional Neurobiology, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Cristiano Lucci
- Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, Naamsestraat 61 - Box 2464, B-3000, Leuven, Belgium
| | - Caroline Simon
- Molecular Developmental Physiology and Signal Transduction, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Dulce Santos
- Molecular Developmental Physiology and Signal Transduction, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration, Department of Biology, KU Leuven, Naamsestraat 61 - Box 2464, B-3000, Leuven, Belgium
| | - Liesbet Temmerman
- Molecular and Functional Neurobiology, Department of Biology, KU Leuven, Naamsestraat 59 - Box 2465, B-3000, Leuven, Belgium.
| |
Collapse
|
2
|
Yuan JQ, Li XY, Fan YN, Fang N, Li P, Wen XZ, Hou Q, Zhang ZQ, Lin MB. Rosmarinic acid suppresses the progression of COPD via Syk by modulating airway inflammation and epithelial apoptosis in vivo and in vitro. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-15. [PMID: 39312447 DOI: 10.1080/10286020.2024.2403617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
Rosmarinic acid (RosA), a hydrophilic phenolic compound found in various plants, has several biological effects such as anti-inflammatory and anti-apoptosis activities. However, its potential impact on chronic obstructive pulmonary disease (COPD) and its underlying mechanism has not been investigated. In this study, we explored the potential therapeutic effects and mechanism of RosA on COPD airway inflammation and alveolar epithelial apoptosis in vivo and in vitro. Our data suggested that RosA may be a therapeutic candidate for COPD with low toxicity. The corresponding mechanism lies in its anti-inflammatory effect on macrophage and bronchial epithelial cells, as well as protective effect on lung epithelial apoptosis via the jointly cross-target spleen tyrosine kinase (Syk).
Collapse
Affiliation(s)
- Ji-Qiao Yuan
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Xu-Yu Li
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yan-Nan Fan
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Nan Fang
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ping Li
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xin-Zhu Wen
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qi Hou
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Zi-Qian Zhang
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ming-Bao Lin
- Laboratory of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
3
|
Croitoru GA, Pîrvulescu DC, Niculescu AG, Epistatu D, Rădulescu M, Grumezescu AM, Nicolae CL. Nanomaterials in Immunology: Bridging Innovative Approaches in Immune Modulation, Diagnostics, and Therapy. J Funct Biomater 2024; 15:225. [PMID: 39194663 DOI: 10.3390/jfb15080225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/30/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
The intersection of immunology and nanotechnology has provided significant advancements in biomedical research and clinical applications over the years. Immunology aims to understand the immune system's defense mechanisms against pathogens. Nanotechnology has demonstrated its potential to manipulate immune responses, as nanomaterials' properties can be modified for the desired application. Research has shown that nanomaterials can be applied in diagnostics, therapy, and vaccine development. In diagnostics, nanomaterials can be used for biosensor development, accurately detecting biomarkers even at very low concentrations. Therapeutically, nanomaterials can act as efficient carriers for delivering drugs, antigens, or genetic material directly to targeted cells or tissues. This targeted delivery improves therapeutic efficacy and reduces the adverse effects on healthy cells and tissues. In vaccine development, nanoparticles can improve vaccine durability and extend immune responses by effectively delivering adjuvants and antigens to immune cells. Despite these advancements, challenges regarding the safety, biocompatibility, and scalability of nanomaterials for clinical applications are still present. This review will cover the fundamental interactions between nanomaterials and the immune system, their potential applications in immunology, and their safety and biocompatibility concerns.
Collapse
Affiliation(s)
- George-Alexandru Croitoru
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania
| | - Diana-Cristina Pîrvulescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Dragoș Epistatu
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania
| | - Marius Rădulescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Faculty of Chemical Engineering and Biotechnology, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania
- Research Institute of the University of Bucharest-ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Carmen-Larisa Nicolae
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania
| |
Collapse
|
4
|
Chevalier C, Chica C, Matheau J, Pain A, Connor MG, Hamon MA. Epithelial cells maintain memory of prior infection with Streptococcus pneumoniae through di-methylation of histone H3. Nat Commun 2024; 15:5545. [PMID: 38956024 PMCID: PMC11219877 DOI: 10.1038/s41467-024-49347-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 05/30/2024] [Indexed: 07/04/2024] Open
Abstract
Epithelial cells are the first point of contact for bacteria entering the respiratory tract. Streptococcus pneumoniae is an obligate human pathobiont of the nasal mucosa, carried asymptomatically but also the cause of severe pneumoniae. The role of the epithelium in maintaining homeostatic interactions or mounting an inflammatory response to invasive S. pneumoniae is currently poorly understood. However, studies have shown that chromatin modifications, at the histone level, induced by bacterial pathogens interfere with the host transcriptional program and promote infection. Here, we uncover a histone modification induced by S. pneumoniae infection maintained for at least 9 days upon clearance of bacteria with antibiotics. Di-methylation of histone H3 on lysine 4 (H3K4me2) is induced in an active manner by bacterial attachment to host cells. We show that infection establishes a unique epigenetic program affecting the transcriptional response of epithelial cells, rendering them more permissive upon secondary infection. Our results establish H3K4me2 as a unique modification induced by infection, distinct from H3K4me3 or me1, which localizes to enhancer regions genome-wide. Therefore, this study reveals evidence that bacterial infection leaves a memory in epithelial cells after bacterial clearance, in an epigenomic mark, thereby altering cellular responses to subsequent infections and promoting infection.
Collapse
Affiliation(s)
- Christine Chevalier
- Institut Pasteur, Université Paris Cité, Chromatin and Infection Laboratory, F-75015, Paris, France
| | - Claudia Chica
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015, Paris, France
| | - Justine Matheau
- Institut Pasteur, Université Paris Cité, Chromatin and Infection Laboratory, F-75015, Paris, France
- Bio Sorbonne Paris Cité doctoral school, Department of Infectiology Microbiology, Université Paris Cité, F-75006, Paris, France
| | - Adrien Pain
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, F-75015, Paris, France
| | - Michael G Connor
- Institut Pasteur, Université Paris Cité, Chromatin and Infection Laboratory, F-75015, Paris, France
| | - Melanie A Hamon
- Institut Pasteur, Université Paris Cité, Chromatin and Infection Laboratory, F-75015, Paris, France.
| |
Collapse
|
5
|
Guallar-Garrido S, Soldati T. Exploring host-pathogen interactions in the Dictyostelium discoideum-Mycobacterium marinum infection model of tuberculosis. Dis Model Mech 2024; 17:dmm050698. [PMID: 39037280 DOI: 10.1242/dmm.050698] [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: 07/23/2024] Open
Abstract
Mycobacterium tuberculosis is a pathogenic mycobacterium that causes tuberculosis. Tuberculosis is a significant global health concern that poses numerous clinical challenges, particularly in terms of finding effective treatments for patients. Throughout evolution, host immune cells have developed cell-autonomous defence strategies to restrain and eliminate mycobacteria. Concurrently, mycobacteria have evolved an array of virulence factors to counteract these host defences, resulting in a dynamic interaction between host and pathogen. Here, we review recent findings, including those arising from the use of the amoeba Dictyostelium discoideum as a model to investigate key mycobacterial infection pathways. D. discoideum serves as a scalable and genetically tractable model for human phagocytes, providing valuable insights into the intricate mechanisms of host-pathogen interactions. We also highlight certain similarities between M. tuberculosis and Mycobacterium marinum, and the use of M. marinum to more safely investigate mycobacteria in D. discoideum.
Collapse
Affiliation(s)
- Sandra Guallar-Garrido
- Department of Biochemistry, Faculty of Science, University of Geneva, 30 quai Ernest-Ansermet, Science II, 1211 Geneva-4, Switzerland
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, 30 quai Ernest-Ansermet, Science II, 1211 Geneva-4, Switzerland
| |
Collapse
|
6
|
Brozat JF, Harbalioğlu N, Hohlstein P, Abu Jhaisha S, Pollmanns MR, Adams JK, Wirtz TH, Hamesch K, Yagmur E, Weiskirchen R, Tacke F, Trautwein C, Koch A. Elevated Serum KIM-1 in Sepsis Correlates with Kidney Dysfunction and the Severity of Multi-Organ Critical Illness. Int J Mol Sci 2024; 25:5819. [PMID: 38892009 PMCID: PMC11172102 DOI: 10.3390/ijms25115819] [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: 04/20/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
The kidney injury molecule (KIM)-1 is shed from proximal tubular cells in acute kidney injury (AKI), relaying tubular epithelial proliferation. Additionally, KIM-1 portends complex immunoregulation and is elevated after exposure to lipopolysaccharides. It thus may represent a biomarker in critical illness, sepsis, and sepsis-associated AKI (SA-AKI). To characterise and compare KIM-1 in these settings, we analysed KIM-1 serum concentrations in 192 critically ill patients admitted to the intensive care unit. Irrespective of kidney dysfunction, KIM-1 serum levels were significantly higher in patients with sepsis compared with other critical illnesses (191.6 vs. 132.2 pg/mL, p = 0.019) and were highest in patients with urogenital sepsis, followed by liver failure. Furthermore, KIM-1 levels were significantly elevated in critically ill patients who developed AKI within 48 h (273.3 vs. 125.8 pg/mL, p = 0.026) or later received renal replacement therapy (RRT) (299.7 vs. 146.3 pg/mL, p < 0.001). KIM-1 correlated with markers of renal function, inflammatory parameters, hematopoietic function, and cholangiocellular injury. Among subcomponents of the SOFA score, KIM-1 was elevated in patients with hyperbilirubinaemia (>2 mg/dL, p < 0.001) and thrombocytopenia (<150/nL, p = 0.018). In univariate and multivariate regression analyses, KIM-1 predicted sepsis, the need for RRT, and multi-organ dysfunction (MOD, SOFA > 12 and APACHE II ≥ 20) on the day of admission, adjusting for relevant comorbidities, bilirubin, and platelet count. Additionally, KIM-1 in multivariate regression was able to predict sepsis in patients without prior (CKD) or present (AKI) kidney injury. Our study suggests that next to its established role as a biomarker in kidney dysfunction, KIM-1 is associated with sepsis, biliary injury, and critical illness severity. It thus may offer aid for risk stratification in these patients.
Collapse
Affiliation(s)
- Jonathan Frederik Brozat
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Neval Harbalioğlu
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Philipp Hohlstein
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Samira Abu Jhaisha
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Maike Rebecca Pollmanns
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Jule Katharina Adams
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Theresa Hildegard Wirtz
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Karim Hamesch
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Eray Yagmur
- Institute of Laboratory Medicine, Western Palatine Hospital, Hellmut-Hartert-Straße 1, 67655 Kaiserslautern, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité—Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Augustenburger Platz 1, 13353 Berlin, Germany;
| | - Christian Trautwein
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| | - Alexander Koch
- Department for Gastroenterology, Metabolic Disorders and Intensive Care Medicine, University Hospital RWTH Aachen, RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany; (J.F.B.); (N.H.); (P.H.); (S.A.J.); (M.R.P.); (J.K.A.); (T.H.W.); (K.H.); (C.T.)
| |
Collapse
|
7
|
Li H, Wang H, Cui L, Liu K, Guo L, Li J, Dong J. The effect of selenium on the proliferation of bovine endometrial epithelial cells in a lipopolysaccharide-induced damage model. BMC Vet Res 2024; 20:109. [PMID: 38500165 PMCID: PMC10946195 DOI: 10.1186/s12917-024-03958-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Endometritis is a common bovine postpartum disease. Rapid endometrial repair is beneficial for forming natural defense barriers and lets cows enter the next breeding cycle as soon as possible. Selenium (Se) is an essential trace element closely related to growth and development in animals. This study aims to observe the effect of Se on the proliferation of bovine endometrial epithelial cells (BEECs) induced by lipopolysaccharide (LPS) and to elucidate the possible underlying mechanism. RESULTS In this study, we developed a BEECs damage model using LPS. Flow cytometry, cell scratch test and EdU proliferation assay were used to evaluate the cell cycle, migration and proliferation. The mRNA transcriptions of growth factors were detected by quantitative reverse transcription-polymerase chain reaction. The activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and Wnt/β-catenin pathways were detected by Western blotting and immunofluorescence. The results showed that the cell viability and BCL-2/BAX protein ratio were significantly decreased, and the cell apoptosis rate was significantly increased in the LPS group. Compared with the LPS group, Se promoted cell cycle progression, increased cell migration and proliferation, and significantly increased the gene expressions of TGFB1, TGFB3 and VEGFA. Se decreased the BCL-2/BAX protein ratio, promoted β-catenin translocation from the cytoplasm to the nucleus and activated the Wnt/β-catenin and PI3K/AKT signaling pathways inhibited by LPS. CONCLUSIONS In conclusion, Se can attenuate LPS-induced damage to BEECs and promote cell proliferation and migration in vitro by enhancing growth factors gene expression and activating the PI3K/AKT and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Hanqing Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China
| | - Heng Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China
| | - Luying Cui
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China
| | - Kangjun Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China
| | - Long Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China
| | - Jianji Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China.
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China.
| | - Junsheng Dong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine , Yangzhou University, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, 12 East Wenhui Rd, Yangzhou, 225009, China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou, 225009, China.
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
8
|
Jakubczyk D, Leszczyńska K, Pacyga-Prus K, Kozakiewicz D, Kazana-Płuszka W, Gełej D, Migdał P, Kruszakin R, Zabłocka A, Górska S. What happens to Bifidobacterium adolescentis and Bifidobacterium longum ssp. longum in an experimental environment with eukaryotic cells? BMC Microbiol 2024; 24:60. [PMID: 38373929 PMCID: PMC10875879 DOI: 10.1186/s12866-023-03179-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/29/2023] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND The impact of probiotic strains on host health is widely known. The available studies on the interaction between bacteria and the host are focused on the changes induced by bacteria in the host mainly. The studies determining the changes that occurred in the bacteria cells are in the minority. Within this paper, we determined what happens to the selected Bifidobacterium adolescentis and Bifidobacterium longum ssp. longum in an experimental environment with the intestinal epithelial layer. For this purpose, we tested the bacteria cells' viability, redox activity, membrane potential and enzymatic activity in different environments, including CaCo-2/HT-29 co-culture, cell culture medium, presence of inflammatory inductor (TNF-α) and oxygen. RESULTS We indicated that the external milieu impacts the viability and vitality of bacteria. Bifidobacterium adolescentis decrease the size of the live population in the cell culture medium with and without TNF-α (p < 0.001 and p < 0.01 respectively). In contrast, Bifidobacterium longum ssp. longum significantly increased survivability in contact with the eukaryotic cells and cell culture medium (p < 0.001). Bifidobacterium adolescentis showed significant changes in membrane potential, which was decreased in the presence of eukaryotic cells (p < 0.01), eukaryotic cells in an inflammatory state (p < 0.01), cell culture medium (p < 0.01) and cell culture medium with TNF-α (p < 0.05). In contrast, Bifidobacterium longum ssp. longum did not modulate membrane potential. Instead, bacteria significantly decreased the redox activity in response to milieus such as eukaryotic cells presence, inflamed eukaryotic cells as well as the culture medium (p < 0.001). The redox activity was significantly different in the cells culture medium vs the presence of eukaryotic cells (p < 0.001). The ability to β-galactosidase production was different for selected strains: Bifidobacterium longum ssp. longum indicated 91.5% of positive cells, whereas Bifidobacterium adolescentis 4.34% only. Both strains significantly reduced the enzyme production in contact with the eukaryotic milieu but not in the cell culture media. CONCLUSION The environmental-induced changes may shape the probiotic properties of bacterial strains. It seems that the knowledge of the sensitivity of bacteria to the external environment may help to select the most promising probiotic strains, reduce research costs, and contribute to greater reproducibility of the obtained probiotic effects.
Collapse
Affiliation(s)
- Dominika Jakubczyk
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland.
| | - Katarzyna Leszczyńska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Katarzyna Pacyga-Prus
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Dominika Kozakiewicz
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Wioletta Kazana-Płuszka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Dominika Gełej
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Paweł Migdał
- Inter-Departmental Laboratory of Instrumental Analysis and Preparation, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Roksana Kruszakin
- Inter-Departmental Laboratory of Instrumental Analysis and Preparation, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Agnieszka Zabłocka
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Sabina Górska
- Laboratory of Microbiome Immunobiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
9
|
Zhao Y, Li M, Mao J, Su Y, Huang X, Xia W, Leng X, Zan T. Immunomodulation of wound healing leading to efferocytosis. SMART MEDICINE 2024; 3:e20230036. [PMID: 39188510 PMCID: PMC11235971 DOI: 10.1002/smmd.20230036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/18/2023] [Indexed: 08/28/2024]
Abstract
Effectively eliminating apoptotic cells is precisely controlled by a variety of signaling molecules and a phagocytic effect known as efferocytosis. Abnormalities in efferocytosis may bring about the development of chronic conditions, including angiocardiopathy, chronic inflammatory diseases and autoimmune diseases. During wound healing, failure of efferocytosis leads to the collection of apoptosis, the release of necrotic material and chronic wounds that are difficult to heal. In addition to the traditional phagocytes-macrophages, other important cell species including dendritic cells, neutrophils, vascular endothelial cells, fibroblasts and keratinocytes contribute to wounding healing. This review summarizes how efferocytosis-mediated immunomodulation plays a repair-promoting role in wound healing, providing new insights for patients suffering from various cutaneous wounds.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Cosmetic and Plastic SurgeryAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Minxiong Li
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jiayi Mao
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yinghong Su
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin Huang
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenzheng Xia
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiangfeng Leng
- Department of Cosmetic and Plastic SurgeryAffiliated Hospital of Qingdao UniversityQingdaoChina
| | - Tao Zan
- Department of Plastic and Reconstructive SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
10
|
Iso-Touru T, Panitz F, Fischer D, Kyläniemi MK, Taponen S, Tabell J, Virta A, Vilkki J. Genes and pathways revealed by whole transcriptome analysis of milk derived bovine mammary epithelial cells after Escherichia coli challenge. Vet Res 2024; 55:13. [PMID: 38303095 PMCID: PMC10835992 DOI: 10.1186/s13567-024-01269-y] [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: 05/23/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
Mastitis, inflammation of the mammary gland, is the costliest disease in dairy cattle and a major animal welfare concern. Mastitis is usually caused by bacteria, of which staphylococci, streptococci and Escherichia coli are most frequently isolated from bovine mastitis. Bacteria activate the mammary immune system in variable ways, thereby influencing the severity of the disease. Escherichia coli is a common cause of mastitis in cattle causing both subclinical and clinical mastitis. Understanding of the molecular mechanisms that activate and regulate the host response would be central to effective prevention of mastitis and breeding of cows more resistant to mastitis. We used primary bovine mammary epithelial cell cultures extracted noninvasively from bovine milk samples to monitor the cellular responses to Escherichia coli challenge. Differences in gene expression between control and challenged cells were studied by total RNA-sequencing at two time points post-challenge. In total, 150 and 440 (Padj < 0.05) differentially expressed genes were identified at 3 h and 24 h post-challenge, respectively. The differentially expressed genes were mostly upregulated at 3 h (141/150) and 24 h (424/440) post-challenge. Our results are in line with known effects of E. coli infection, with a strong early inflammatory response mediated by pathogen receptor families. Among the most significantly enriched early KEGG pathways were the TNF signalling pathway, the cytokine-cytokine receptor interaction, and the NF-kappa B signalling pathway. At 24 h post-challenge, most significantly enriched were the Influenza A, the NOD-like receptor signalling, and the IL-17 signaling pathway.
Collapse
Affiliation(s)
- Terhi Iso-Touru
- Natural Resources Institute Finland (Luke), Jokioinen, Finland.
| | - Frank Panitz
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Daniel Fischer
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Minna K Kyläniemi
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Suvi Taponen
- Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Jonna Tabell
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Anneli Virta
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| | - Johanna Vilkki
- Natural Resources Institute Finland (Luke), Jokioinen, Finland
| |
Collapse
|
11
|
Burgher-Pulgaron Y, Provost C, Alvarez F, Meza-Serrano E, Pesant MJ, Price CA, Gagnon CA. DUSP1 mRNA modulation during porcine circovirus type 2 and porcine reproductive and respiratory syndrome virus co-infection regulates viruses replication. Virus Res 2024; 339:199282. [PMID: 37995964 PMCID: PMC10711501 DOI: 10.1016/j.virusres.2023.199282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023]
Abstract
The effects of porcine circovirus type 2b (PCV2b) and porcine reproductive and respiratory syndrome virus (PRRSV) co-infection in epithelial cells of the swine respiratory tract is unknown. In the present study, the newborn pig trachea cell line NPTr-CD163, which is permissive to both viruses, was persistently infected with PCV2b and then with PRRSV. Viral replication, cell viability, cytokines' mRNA expression, and modulation of cellular genes expression were evaluated in infected cells. In NPTr-CD163 co-infection model, PCV2b replication was enhanced while PRRSV replication was suppressed. Cell viability was significantly decreased during PCV2b single infection and co-infection compared to mock-infected and PRRSV single infected cells. However, no difference was observed in cell viability between PCV2b and PCV2b/PRRSV infected cells. The IL6, IL8 and IL10 mRNA expression was significantly higher in co-infected cells compared to PCV2b and PRRSV single infected cells. Moreover, the IFN-α/β expression was significantly reduced in co-infected cells compared to PCV2b infected cells whereas it remained higher compared to PRRSV infected cells. The differential gene expression analysis revealed that the mRNA expression level of the cellular gene DUSP1 was significantly higher in all PRRSV infection models compared to PCV2b single infected cells. Knockdown of DUSP1 expression in co-infected cells significantly reduced PCV2b replication, suggesting a role for DUSP1 in PCV2b/PRRSV pathogenesis.
Collapse
Affiliation(s)
- Yaima Burgher-Pulgaron
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA-FRQNT), Faculté de Médecine Vétérinaire (FMV), Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Chantale Provost
- Molecular Diagnostic Laboratory, Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), FMV, Canada
| | - Fernando Alvarez
- Infectious Diseases and Immunity in Global Health (IDIGH), McGill University, 1001 Décarie, Montréal, Québec, Canada, H4A 3J1
| | - Europa Meza-Serrano
- Centre de Recherche en Reproduction Animale, FMV, Université de Montréal, Canada
| | - Marie-Jeanne Pesant
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA-FRQNT), Faculté de Médecine Vétérinaire (FMV), Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2
| | - Christopher A Price
- Centre de Recherche en Reproduction Animale, FMV, Université de Montréal, Canada
| | - Carl A Gagnon
- The Swine and Poultry Infectious Diseases Research Centre (CRIPA-FRQNT), Faculté de Médecine Vétérinaire (FMV), Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, Québec, Canada, J2S 2M2; Molecular Diagnostic Laboratory, Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), FMV, Canada.
| |
Collapse
|
12
|
Chala D, Sabadashka M, Morozovych A, Krychowiak-Maśnicka M, Królicka A, Sybirna N. Immunomodulatory and antibacterial effect of red wine concentrate rich in a natural complex of polyphenols under diabetes mellitus. Biomed Pharmacother 2024; 170:116023. [PMID: 38104417 DOI: 10.1016/j.biopha.2023.116023] [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: 10/16/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/19/2023] Open
Abstract
Changes in immunocompetent cells influence the course of diabetes mellitus and contribute to its complications. Thus, correction of diabetes-induced immune system disorders is vital for normalizing the state of the organism. Red wine polyphenols due to their biological activities could be considered a potential remedy for correcting diabetes. The study aimed to evaluate the antimicrobial potential and the influence of red wine polyphenols on immune system in streptozotocin-induced diabetes. We studied immunological parameters, i.e. quantity of white blood cells in peripheral blood and peritoneal macrophages, the bactericidal activity of phagocytes of blood, the activity of myeloperoxidase, and the level of cationic proteins in these cells after the administration of the polyphenol-rich red wine concentrate (PC concentrate) of known composition, obtained from Ukrainian wine, for 14th day to rats with streptozotocin-induced diabetes. The Minimal Bactericidal Concentration (MBC) of the PC concentrate was determined with the Broth Microdilution method. The PC concentrate normalized the quantity and functional activity of peripheral blood neutrophils and peritoneal macrophages, and decreased the quantity of lymphocytes under diabetes, as well as possessed the antibacterial activity against Staphylococcus aureus and Escherichia coli. Our results indicate the significant biological potential of the PC concentrate and its therapeutic relevance to correct diabetes-induced disorders.
Collapse
Affiliation(s)
- Dariya Chala
- Ivan Franko National University of Lviv, Hrushevskogo 4, Lviv 79005, Ukraine.
| | - Mariya Sabadashka
- Ivan Franko National University of Lviv, Hrushevskogo 4, Lviv 79005, Ukraine
| | | | - Marta Krychowiak-Maśnicka
- University of Gdansk, Intercollegiate Faculty of Biotechnology UG & MUG, Laboratory of Biologically Active Compounds, Abrahama 58, 80-307 Gdansk, Poland
| | - Aleksandra Królicka
- University of Gdansk, Intercollegiate Faculty of Biotechnology UG & MUG, Laboratory of Biologically Active Compounds, Abrahama 58, 80-307 Gdansk, Poland
| | - Nataliia Sybirna
- Ivan Franko National University of Lviv, Hrushevskogo 4, Lviv 79005, Ukraine
| |
Collapse
|
13
|
Li Y, Jia G, Li T, Zhou X, Zhao H, Cao J, Guan Z, Zhao R. Preparation Optimization and Immunological Activity Studies of Portulaca oleracea L. Polysaccharides Liposomes. Curr Pharm Des 2024; 30:786-797. [PMID: 38385493 DOI: 10.2174/0113816128279071231204071210] [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: 09/16/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 02/23/2024]
Abstract
AIMS This study combines traditional Chinese medicine polysaccharides with nanomaterials to enhance drug bioavailability and immunological activity. BACKGROUND The study of polysaccharide preparation, structure identification, pharmacological activity, and mechanism of action is deepening, but the research combined with the new drug delivery system is relatively weak, so the application of polysaccharides is still facing great limitations. In order to prolong the action time of polysaccharides and improve their bioavailability, liposome has become the most promising delivery carrier. OBJECTIVES The purpose of this study was to optimize the preparation process of Portulaca oleracea L. polysaccharides liposomes (POL-PL) and evaluate the immunoactivity in vitro. METHODS POL-PL was prepared by reverse evaporation, and the preparation process was optimized using the response surface methodology. The characteristic analysis of POL-PL was detected by the indicators including morphology, particle size, zeta potential, encapsulation efficiency, release, and stability. The effects of POL-PL on the proliferation and immunological activity of mouse spleen lymphocytes and RAW264.7 cells were evaluated in vitro. RESULTS POL-PL is highly homogeneous in morphology and particle size, and its sustained release improves the bioavailability of Portulaca oleracea L. polysaccharides (POL-P). Moreover, POL-PL treatment significantly enhanced the proliferation and phagocytic activity of RAW264.7 cells and increased the secretion of IL-6, TNF-α, IL-1β, and NO. CONCLUSION This study suggested that POL-PL were prepared successfully by reverse evaporation method, and POL-PL had immunoenhancing activity in vitro. The results provided a theoretical basis for further application of POL-PL.
Collapse
Affiliation(s)
- Yan Li
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| | - Guiyan Jia
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| | - Tao Li
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| | - Xiechen Zhou
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| | - Hui Zhao
- Centers for Disease Control and Prevention, Daqing City People's Hospital, Daqing, Heilongjiang Province 163319, P.R. China
| | - Junyang Cao
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| | - Zijan Guan
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| | - Rui Zhao
- College of Life Science & Biotechnology, Heilongjiang Bayi Agricultural University, Daqing High-Tech Industrial Development Zone, Daqing, Heilongjiang Province 163319, P.R. China
| |
Collapse
|
14
|
Baker P, Huang C, Radi R, Moll SB, Jules E, Arbiser JL. Skin Barrier Function: The Interplay of Physical, Chemical, and Immunologic Properties. Cells 2023; 12:2745. [PMID: 38067173 PMCID: PMC10706187 DOI: 10.3390/cells12232745] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
An intact barrier function of the skin is important in maintaining skin health. The regulation of the skin barrier depends on a multitude of molecular and immunological signaling pathways. By examining the regulation of a healthy skin barrier, including maintenance of the acid mantle and appropriate levels of ceramides, dermatologists can better formulate solutions to address issues that are related to a disrupted skin barrier. Conversely, by understanding specific skin barrier disruptions that are associated with specific conditions, such as atopic dermatitis or psoriasis, the development of new compounds could target signaling pathways to provide more effective relief for patients. We aim to review key factors mediating skin barrier regulation and inflammation, including skin acidity, interleukins, nuclear factor kappa B, and sirtuin 3. Furthermore, we will discuss current and emerging treatment options for skin barrier conditions.
Collapse
Affiliation(s)
- Paola Baker
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.B.); (C.H.); (R.R.); (S.B.M.); (E.J.)
| | - Christina Huang
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.B.); (C.H.); (R.R.); (S.B.M.); (E.J.)
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Rakan Radi
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.B.); (C.H.); (R.R.); (S.B.M.); (E.J.)
| | - Samara B. Moll
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.B.); (C.H.); (R.R.); (S.B.M.); (E.J.)
| | - Emmanuela Jules
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA 30322, USA; (P.B.); (C.H.); (R.R.); (S.B.M.); (E.J.)
| | - Jack L. Arbiser
- Metroderm/United Derm Partners, 875 Johnson Ferry Road, Atlanta, GA 30342, USA
| |
Collapse
|
15
|
Bowen J, Cross C. The Role of the Innate Immune Response in Oral Mucositis Pathogenesis. Int J Mol Sci 2023; 24:16314. [PMID: 38003503 PMCID: PMC10670995 DOI: 10.3390/ijms242216314] [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: 10/07/2023] [Revised: 11/11/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Oral mucositis (OM) is a significant complication of cancer therapy with limited management strategies. Whilst inflammation is a central feature of destructive and ultimately ulcerative pathology, to date, attempts to mitigate damage via this mechanism have proven limited. A relatively underexamined aspect of OM development is the contribution of elements of the innate immune system. In particular, the role played by barriers, pattern recognition systems, and microbial composition in early damage signaling requires further investigation. As such, this review highlights the innate immune response as a potential focus for research to better understand OM pathogenesis and development of interventions for patients treated with radiotherapy and chemotherapy. Future areas of evaluation include manipulation of microbial-mucosal interactions to alter cytotoxic sensitivity, use of germ-free models, and translation of innate immune-targeted agents interrogated for mucosal injury in other regions of the alimentary canal into OM-based clinical trials.
Collapse
Affiliation(s)
- Joanne Bowen
- School of Biomedicine, University of Adelaide, Adelaide 5005, Australia;
| | | |
Collapse
|
16
|
Slattery O, Dahle MK, Sundaram AYM, Nowak BF, Gjessing MC, Solhaug A. Functional and molecular characterization of the Atlantic salmon gill epithelium cell line ASG-10; a tool for in vitro gill research. Front Mol Biosci 2023; 10:1242879. [PMID: 37916189 PMCID: PMC10616884 DOI: 10.3389/fmolb.2023.1242879] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/08/2023] [Indexed: 11/03/2023] Open
Abstract
Fish gills are not only the respiratory organ, but also essential for ion-regulation, acid-base control, detoxification, waste excretion and host defense. Multifactorial gill diseases are common in farmed Atlantic salmon, and still poorly understood. Understanding gill pathophysiology is of paramount importance, but the sacrifice of large numbers of experimental animals for this purpose should be avoided. Therefore, in vitro models, such as cell lines, are urgently required to replace fish trials. An Atlantic salmon gill epithelial cell line, ASG-10, was established at the Norwegian Veterinary institute in 2018. This cell line forms a monolayer expressing cytokeratin, e-cadherin and desmosomes, hallmarks of a functional epithelial barrier. To determine the value of ASG-10 for comparative studies of gill functions, the characterization of ASG-10 was taken one step further by performing functional assays and comparing the cell proteome and transcriptome with those of gills from juvenile freshwater Atlantic salmon. The ASG-10 cell line appear to be a homogenous cell line consisting of epithelial cells, which express tight junction proteins. We demonstrated that ASG-10 forms a barrier, both alone and in co-culture with the Atlantic salmon gill fibroblast cell line ASG-13. ASG-10 cells can phagocytose and express several ATP-binding cassette transport proteins. Additionally, ASG-10 expresses genes involved in biotransformation of xenobiotics and immune responses. Taken together, this study provides an overview of functions that can be studied using ASG-10, which will be an important contribution to in vitro gill epithelial research of Atlantic salmon.
Collapse
Affiliation(s)
- Orla Slattery
- Marine and Freshwater Research Centre, Atlantic Technological University, Galway, Ireland
| | | | - Arvind Y. M. Sundaram
- Norwegian Veterinary Institute, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Barbara F. Nowak
- Institute of Marine and Antarctic Studies, University of Tasmania, Hobart, TAS, Australia
| | | | | |
Collapse
|
17
|
Taya T, Teruyama F, Gojo S. Host-directed therapy for bacterial infections -Modulation of the phagolysosome pathway. Front Immunol 2023; 14:1227467. [PMID: 37841276 PMCID: PMC10570837 DOI: 10.3389/fimmu.2023.1227467] [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: 05/23/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Bacterial infections still impose a significant burden on humanity, even though antimicrobial agents have long since been developed. In addition to individual severe infections, the f fatality rate of sepsis remains high, and the threat of antimicrobial-resistant bacteria grows with time, putting us at inferiority. Although tremendous resources have been devoted to the development of antimicrobial agents, we have yet to recover from the lost ground we have been driven into. Looking back at the evolution of treatment for cancer, which, like infectious diseases, has the similarity that host immunity eliminates the lesion, the development of drugs to eliminate the tumor itself has shifted from a single-minded focus on drug development to the establishment of a treatment strategy in which the de-suppression of host immunity is another pillar of treatment. In infectious diseases, on the other hand, the development of therapies that strengthen and support the immune system has only just begun. Among innate immunity, the first line of defense that bacteria encounter after invading the host, the molecular mechanisms of the phagolysosome pathway, which begins with phagocytosis to fusion with lysosome, have been elucidated in detail. Bacteria have a large number of strategies to escape and survive the pathway. Although the full picture is still unfathomable, the molecular mechanisms have been elucidated for some of them, providing sufficient clues for intervention. In this article, we review the host defense mechanisms and bacterial evasion mechanisms and discuss the possibility of host-directed therapy for bacterial infection by intervening in the phagolysosome pathway.
Collapse
Affiliation(s)
- Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Fumiya Teruyama
- Pharmacology Research Department, Tokyo New Drug Research Laboratories, Kowa Company, Ltd., Tokyo, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Keramidas P, Papachristou E, Papi RM, Mantsou A, Choli-Papadopoulou T. Inhibition of PERK Kinase, an Orchestrator of the Unfolded Protein Response (UPR), Significantly Reduces Apoptosis and Inflammation of Lung Epithelial Cells Triggered by SARS-CoV-2 ORF3a Protein. Biomedicines 2023; 11:1585. [PMID: 37371681 DOI: 10.3390/biomedicines11061585] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
SARS-CoV-2 ORF3a accessory protein was found to be involved in virus release, immunomodulation and exhibited a pro-apoptotic character. In order to unravel a potential ORF3a-induced apoptotic and inflammatory death mechanism, lung epithelial cells (A549) were transfected with in vitro synthesized ORF3a mRNA. The protein's dynamic involvement as "stress factor" for the endoplasmic reticulum, causing the activation of PERK kinase and other UPR-involved proteins and therefore the upregulation of their signaling pathway executioners (ATF6, XBP-1s, PERK, phospho eIF2a, ATF4, CHOP, GADD34), has been clearly demonstrated. Furthermore, the overexpression of BAX and BH3-only pro-apoptotic protein PUMA, the upregulation of Bcl-2 family genes (BAX, BAK, BID, BAD), the reduced expression of Bcl-2 in mRNA and protein levels, and lastly, the cleavage of PARP-1 and caspase family members (caspase-3,-8 and -9) indicate that ORF3a displays its apoptotic character through the mitochondrial pathway of apoptosis. Moreover, the upregulation of NFκB, phosphorylation of p65 and IκΒα and the elevated expression of pro-inflammatory cytokines (IL-1b, IL-6, IL-8 and IL-18) in transfected cells with ORF3a mRNA indicate that this protein causes the inflammatory response through NFκB activation and therefore triggers lung injury. An intriguing finding of our study is that upon treatment of the ORF3a-transfected cells with GSK2606414, a selective PERK inhibitor, both complications (apoptosis and inflammatory response) were neutralized, and cell survival was favored, whereas treatment of transfected cells with z-VAD (a pan-caspase inhibitor) despite inhibiting cell death, could not ameliorate the inflammatory response of transfected A549 cells. Given the above, we point out that PERK kinase is a "master tactician" and its activation constitutes the main stimulus for the emergence of ORF3a apoptotic and inflammatory nature and therefore could serve as potential target for developing novel therapeutic approaches against COVID-19.
Collapse
Affiliation(s)
- Panagiotis Keramidas
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleni Papachristou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Rigini M Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Aglaia Mantsou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Theodora Choli-Papadopoulou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
19
|
Burgher Pulgaron Y, Provost C, Pesant MJ, Gagnon CA. Porcine Circovirus Modulates Swine Influenza Virus Replication in Pig Tracheal Epithelial Cells and Porcine Alveolar Macrophages. Viruses 2023; 15:v15051207. [PMID: 37243291 DOI: 10.3390/v15051207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/15/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
The pathogenesis of porcine circovirus type 2b (PCV2b) and swine influenza A virus (SwIV) during co-infection in swine respiratory cells is poorly understood. To elucidate the impact of PCV2b/SwIV co-infection, newborn porcine tracheal epithelial cells (NPTr) and immortalized porcine alveolar macrophages (iPAM 3D4/21) were co-infected with PCV2b and SwIV (H1N1 or H3N2 genotype). Viral replication, cell viability and cytokine mRNA expression were determined and compared between single-infected and co-infected cells. Finally, 3'mRNA sequencing was performed to identify the modulation of gene expression and cellular pathways in co-infected cells. It was found that PCV2b significantly decreased or improved SwIV replication in co-infected NPTr and iPAM 3D4/21 cells, respectively, compared to single-infected cells. Interestingly, PCV2b/SwIV co-infection synergistically up-regulated IFN expression in NPTr cells, whereas in iPAM 3D4/21 cells, PCV2b impaired the SwIV IFN induced response, both correlating with SwIV replication modulation. RNA-sequencing analyses revealed that the modulation of gene expression and enriched cellular pathways during PCV2b/SwIV H1N1 co-infection is regulated in a cell-type-dependent manner. This study revealed different outcomes of PCV2b/SwIV co-infection in porcine epithelial cells and macrophages and provides new insights on porcine viral co-infections pathogenesis.
Collapse
Affiliation(s)
- Yaima Burgher Pulgaron
- Swine and Poultry Infectious Diseases Research Center (CRIPA-FRQ), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Chantale Provost
- Molecular Diagnostic Laboratory, Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Marie-Jeanne Pesant
- Swine and Poultry Infectious Diseases Research Center (CRIPA-FRQ), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Carl A Gagnon
- Swine and Poultry Infectious Diseases Research Center (CRIPA-FRQ), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Molecular Diagnostic Laboratory, Centre de Diagnostic Vétérinaire de l'Université de Montréal (CDVUM), Saint-Hyacinthe, QC J2S 2M2, Canada
| |
Collapse
|
20
|
Liu R, Armstrong E, Constable S, Buchanan LB, Mohammadi A, Galiwango RM, Huibner S, Perry MC, Prodger JL, Coburn B, Kaul R. Soluble E-cadherin: A marker of genital epithelial disruption. Am J Reprod Immunol 2023; 89:e13674. [PMID: 36593681 DOI: 10.1111/aji.13674] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 01/04/2023] Open
Abstract
PROBLEM The genital epithelial barrier is a crucial first line of defence against HIV, and epithelial disruption may enhance HIV susceptibility. Assessment of genital epithelial integrity requires biopsies, but their collection is not practical in many research settings. A validated biomarker of genital epithelial barrier integrity would therefore be useful. The purpose of this study was to evaluate soluble E-cadherin (sE-cad) as a marker of genital epithelial disruption. METHOD OF STUDY Using in vitro models of endocervical and foreskin epithelial cells, we assessed changes in sE-cad, IL-6, IL-1β, and IL-1α levels following mechanical disruption. We also assessed changes in sE-cad levels in vivo in cervicovaginal secretions after epithelial disruption by endocervical cytobrush sampling in Canadian women, and assessed the relationship between levels of sE-cad in coronal sulcus swabs to membrane-bound E-cadherin in the overlying foreskin tissue in Ugandan men. RESULTS sE-cad levels immediately increased after in vitro epithelial physical disruption with the degree of elevation dependent on the extent of disruption, as did levels of IL-1β and IL-1α; this was followed by a delayed increase in IL-6 levels. In vivo results confirmed that sE-cad levels in cervicovaginal secretions were elevated 6 h after cytobrush sampling when compared to baseline. Furthermore, levels of sE-cad in the prepuce were inversely correlated with the amount of membrane-bound E-cadherin of overlying tissue. CONCLUSION Our results validate the use of sE-cad as a marker of epithelial disruption and demonstrate that the processes of physical disruption and inflammation in the genital tract are strongly intertwined.
Collapse
Affiliation(s)
- Rachel Liu
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eric Armstrong
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Shirley Constable
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Lane B Buchanan
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Avid Mohammadi
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Sanja Huibner
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Marie C Perry
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jessica L Prodger
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Bryan Coburn
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University Health Network, Toronto, Ontario, Canada
| | - Rupert Kaul
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Shen J, Yang F, Wang G, Mou X, Li J, Ding X, Wang X, Li H. Paeoniflorin alleviates inflammation in bovine mammary epithelial cells induced by Staphylococcus haemolyticus through TLR2/NF-κB signaling pathways. Res Vet Sci 2023; 156:95-103. [PMID: 36796241 DOI: 10.1016/j.rvsc.2023.01.022] [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: 08/13/2022] [Revised: 01/04/2023] [Accepted: 01/29/2023] [Indexed: 02/12/2023]
Abstract
Staphylococcus haemolyticus (S. haemolyticus) is one of the most common coagulase-negative staphylococci (CoNS) isolates from bovine mastitis. Paeoniflorin (PF) shows anti-inflammatory effects on different inflammatory diseases in vitro studies and in vivo animal experiments. In this study, the viability of bovine mammary epithelial cells (bMECs) was detected by the cell counting kit-8 experiment. Subsequently, bMECs were induced with S. haemolyticus, and the induction dosage was determined. The expression of pro-inflammatory cytokines and toll-like receptor (TLR2) and nuclear factor kappa-B (NF-κB) signaling pathway-related genes were investigated by quantitative real-time PCR. The critical pathway proteins were detected by western blot. The results showed that the multiplicity of infection (MOI; the ratio of bacteria to bMECs) 5:1 of S. haemolyticus for 12 h could cause cellular inflammation, which was selected to establish the inflammatory model. Incubation with 50 μg/ml PF for 12 h was the best intervention condition for cells stimulated by S. hemolyticus. Quantitative real-time PCR and western blot analysis showed that PF inhibited the activation of TLR2 and NF-κB pathway-related genes and the expression of related proteins. Western blot results showed that PF suppressed the expression of NF-κB unit p65, NF-κB unit p50, and MyD88 in bMECs stimulated by S. haemolyticus. The inflammatory response pathway and molecular mechanism caused by S. haemolyticus on bMECs are related to TLR2-mediated NF-κB signaling pathways. The anti-inflammatory mechanism of PF may also be through this pathway. Therefore, PF is expected to develop potential drugs against CoNS-induced bovine mastitis.
Collapse
Affiliation(s)
- Jirao Shen
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Feng Yang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Guibo Wang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xiaoqing Mou
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Jinyu Li
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xuezhi Ding
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China
| | - Xurong Wang
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China.
| | - Hongsheng Li
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agricultural Sciences, Lanzhou, People's Republic of China.
| |
Collapse
|
22
|
Tischenko K, Brill-Karniely Y, Steinberg E, Segev-Yekutiel H, Benny O. Surface physical cues mediate the uptake of foreign particles by cancer cells. APL Bioeng 2023; 7:016113. [PMID: 36960390 PMCID: PMC10030191 DOI: 10.1063/5.0138245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 03/02/2023] [Indexed: 03/22/2023] Open
Abstract
Cancer phenotypes are often associated with changes in the mechanical states of cells and their microenvironments. Numerous studies have established correlations between cancer cell malignancy and cell deformability at the single-cell level. The mechanical deformation of cells is required for the internalization of large colloidal particles. Compared to normal epithelial cells, cancer cells show higher capacities to distort their shapes during the engulfment of external particles, thus performing phagocytic-like processes more efficiently. This link between cell deformability and particle uptake suggests that the cell's adherence state may affect this particle uptake, as cells become stiffer when plated on a more rigid substrate and vice versa. Based on this, we hypothesized that cancer cells of the same origin, which are subjected to external mechanical cues through attachment to surfaces with varying rigidities, may express different capacities to uptake foreign particles. The effects of substrate rigidity on cancer cell uptake of inert particles (0.8 and 2.4 μm) were examined using surfaces with physiologically relevant rigidities (from 0.5 to 64 kPa). Our data demonstrate a wave-like ("meandering") dependence of cell uptake on the rigidity of the culture substrate explained by a superposition of opposing physical and biological effects. The uptake patterns were inversely correlated with the expression of phosphorylated paxillin, indicating that the initial passive particle absorbance is the primary limiting step toward complete uptake. Overall, our findings may provide a foundation for mechanical rationalization of particle uptake design.
Collapse
Affiliation(s)
- Katerina Tischenko
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Yifat Brill-Karniely
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Eliana Steinberg
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Hadas Segev-Yekutiel
- The Core Research Facility, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| | - Ofra Benny
- Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001 Israel
| |
Collapse
|
23
|
Suzuki Y, Kami D, Taya T, Sano A, Ogata T, Matoba S, Gojo S. ZLN005 improves the survival of polymicrobial sepsis by increasing the bacterial killing via inducing lysosomal acidification and biogenesis in phagocytes. Front Immunol 2023; 14:1089905. [PMID: 36820088 PMCID: PMC9938763 DOI: 10.3389/fimmu.2023.1089905] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/23/2023] [Indexed: 02/07/2023] Open
Abstract
Polymicrobial sepsis still has a high mortality rate despite the development of antimicrobial agents, elaborate strategies to protect major organs, and the investment of numerous medical resources. Mitochondrial dysfunction, which acts as the center of energy metabolism, is clearly the basis of pathogenesis. Drugs that act on PGC1α, the master regulator of mitochondrial biosynthesis, have shown useful effects in the treatment of sepsis; therefore, we investigated the efficacy of ZLN005, a PGC1α agonist, and found significant improvement in overall survival in an animal model. The mode of action of this effect was examined, and it was shown that the respiratory capacity of mitochondria was enhanced immediately after administration and that the function of TFEB, a transcriptional regulator that promotes lysosome biosynthesis and mutually enhances PGC1α, was enhanced, as was the physical contact between mitochondria and lysosomes. ZLN005 strongly supported immune defense in early sepsis by increasing lysosome volume and acidity and enhancing cargo degradation, resulting in a significant reduction in bacterial load. ZLN005 rapidly acted on two organelles, mitochondria and lysosomes, against sepsis and interactively linked the two to improve the pathogenesis. This is the first demonstration that acidification of lysosomes by a small molecule is a mechanism of action in the therapeutic strategy for sepsis, which will have a significant impact on future drug discovery.
Collapse
Affiliation(s)
- Yosuke Suzuki
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Kami
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshihiko Taya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Arata Sano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pathology and Cell Regulation, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoshi Gojo
- Department of Regenerative Medicine, Graduate School of Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
24
|
Cohen-Kedar S, Shaham Barda E, Rabinowitz KM, Keizer D, Abu-Taha H, Schwartz S, Kaboub K, Baram L, Sadot E, White I, Wasserberg N, Wolff-Bar M, Levy-Barda A, Dotan I. Human intestinal epithelial cells can internalize luminal fungi via LC3-associated phagocytosis. Front Immunol 2023; 14:1142492. [PMID: 36969163 PMCID: PMC10030769 DOI: 10.3389/fimmu.2023.1142492] [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: 01/11/2023] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Background Intestinal epithelial cells (IECs) are the first to encounter luminal microorganisms and actively participate in intestinal immunity. We reported that IECs express the β-glucan receptor Dectin-1, and respond to commensal fungi and β-glucans. In phagocytes, Dectin-1 mediates LC3-associated phagocytosis (LAP) utilizing autophagy components to process extracellular cargo. Dectin-1 can mediate phagocytosis of β-glucan-containing particles by non-phagocytic cells. We aimed to determine whether human IECs phagocytose β-glucan-containing fungal particles via LAP. Methods Colonic (n=18) and ileal (n=4) organoids from individuals undergoing bowel resection were grown as monolayers. Fluorescent-dye conjugated zymosan (β-glucan particle), heat-killed- and UV inactivated C. albicans were applied to differentiated organoids and to human IEC lines. Confocal microscopy was used for live imaging and immuno-fluorescence. Quantification of phagocytosis was carried out with a fluorescence plate-reader. Results zymosan and C. albicans particles were phagocytosed by monolayers of human colonic and ileal organoids and IEC lines. LAP was identified by LC3 and Rubicon recruitment to phagosomes and lysosomal processing of internalized particles was demonstrated by co-localization with lysosomal dyes and LAMP2. Phagocytosis was significantly diminished by blockade of Dectin-1, actin polymerization and NAPDH oxidases. Conclusions Our results show that human IECs sense luminal fungal particles and internalize them via LAP. This novel mechanism of luminal sampling suggests that IECs may contribute to the maintenance of mucosal tolerance towards commensal fungi.
Collapse
Affiliation(s)
- Sarit Cohen-Kedar
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- *Correspondence: Iris Dotan, ; Sarit Cohen-Kedar,
| | - Efrat Shaham Barda
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Keren Masha Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Danielle Keizer
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Hanan Abu-Taha
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shoshana Schwartz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Kawsar Kaboub
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Liran Baram
- Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Sadot
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Ian White
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Wasserberg
- Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Meirav Wolff-Bar
- Department of Pathology, Rabin Medical Center, Petah-Tikva, Israel
| | | | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Iris Dotan, ; Sarit Cohen-Kedar,
| |
Collapse
|
25
|
Yi M, Liu S, Jiao B, Niu Y, Shen M, Duan H, Dai Y. Effect of trichloroethanol on TLR2 and TLR4/NF-κB-mediated antigen processing and presentation in HLA-B* 13:01-transfected antigen-presenting cells. Toxicol Lett 2022; 373:123-131. [PMID: 36423715 DOI: 10.1016/j.toxlet.2022.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 10/28/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Trichloroethanol (TCOH), as a metabolite of trichloroethylene, has sensitization in the pathogenesis of trichloroethylene-induced hypersensitivity dermatitis (TIHD) which the human leukocyte antigen (HLA)-B∗13:01 gene is strongly associated with it. However, it is still obscure how TCOH participates in the pathogenesis of TIHD. Here, we demonstrate that TLR2 and TLR4 signaling through MyD88 and TRAF6-dependent pathway could activate NF-κB by promoting degradation of the inhibitor IκB-α to stimulate the process of NF-κB nuclear translocation. Besides, the crucial molecules of antigen processing and presentation, including TAP1, LMP2, LMP7, and HLA-B* 13:01, were all enhanced and the abundance of HLA-B* 13:01 on the surface of CIR-B* 13:01 cells was also up-regulated with the TCOH concentration increasing. Notably, we used 50 μM pyrrolidinedithiocarbamate (ammonium) to effectively inhibit the activation of NF-κB, which could effectively reverse the stimulation of antigen processing and presentation in TCOH-treated CIR-B* 13:01 cells. Taken together, we speculated that TCOH could promote the abundance of HLA complex on the antigen-presenting cells via TLR2 and TLR4/NF-κB to induce the severe reactivation of T lymphocytes, leading to the extreme immune response.
Collapse
Affiliation(s)
- Mengnan Yi
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Shuai Liu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Bo Jiao
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yong Niu
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Meili Shen
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Huawei Duan
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yufei Dai
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China.
| |
Collapse
|
26
|
Zemanova M, Langova L, Novotná I, Dvorakova P, Vrtkova I, Havlicek Z. Immune mechanisms, resistance genes, and their roles in the prevention of mastitis in dairy cows. Arch Anim Breed 2022; 65:371-384. [PMID: 36415759 PMCID: PMC9673033 DOI: 10.5194/aab-65-371-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/05/2022] [Indexed: 01/25/2023] Open
Abstract
Mastitis is one of the most important diseases of the mammary gland. The increased incidence of this disease in cows is due to the breeding of dairy cattle for higher yields, which is accompanied by an increased susceptibility to mastitis. Therefore, the difficulty involved with preventing this disease has increased. An integral part of current research is the elimination of mastitis in order to reduce the consumption of antibiotic drugs, thereby reducing the resistance of microorganisms and decreasing companies' economic losses due to mastitis (i.e. decreased milk yield, increased drug costs, and reduced milk supply). Susceptibility to mastitis is based on dairy cows' immunity, health, nutrition, and welfare. Thus, it is important to understand the immune processes in the body in order to increase the resistance of animals. Recently, various studies have focused on the selection of mastitis resistance genes. An important point is also the prevention of mastitis. This publication aims to describe the physiology of the mammary gland along with its immune mechanisms and to approximate their connection with potential mastitis resistance genes. This work describes various options for mastitis elimination and focuses on genetic selection and a closer specification of resistance genes to mastitis. Among the most promising resistance genes for mastitis, we consider CD14, CXCR1, lactoferrin, and lactoglobulin.
Collapse
|
27
|
Rainard P, Gilbert FB, Germon P. Immune defenses of the mammary gland epithelium of dairy ruminants. Front Immunol 2022; 13:1031785. [PMID: 36341445 PMCID: PMC9634088 DOI: 10.3389/fimmu.2022.1031785] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/03/2022] [Indexed: 11/17/2022] Open
Abstract
The epithelium of the mammary gland (MG) fulfills three major functions: nutrition of progeny, transfer of immunity from mother to newborn, and its own defense against infection. The defense function of the epithelium requires the cooperation of mammary epithelial cells (MECs) with intraepithelial leucocytes, macrophages, DCs, and resident lymphocytes. The MG is characterized by the secretion of a large amount of a nutrient liquid in which certain bacteria can proliferate and reach a considerable bacterial load, which has conditioned how the udder reacts against bacterial invasions. This review presents how the mammary epithelium perceives bacteria, and how it responds to the main bacterial genera associated with mastitis. MECs are able to detect the presence of actively multiplying bacteria in the lumen of the gland: they express pattern recognition receptors (PRRs) that recognize microbe-associated molecular patterns (MAMPs) released by the growing bacteria. Interactions with intraepithelial leucocytes fine-tune MECs responses. Following the onset of inflammation, new interactions are established with lymphocytes and neutrophils recruited from the blood. The mammary epithelium also identifies and responds to antigens, which supposes an antigen-presenting capacity. Its responses can be manipulated with drugs, plant extracts, probiotics, and immune modifiers, in order to increase its defense capacities or reduce the damage related to inflammation. Numerous studies have established that the mammary epithelium is a genuine effector of both innate and adaptive immunity. However, knowledge gaps remain and newly available tools offer the prospect of exciting research to unravel and exploit the multiple capacities of this particular epithelium.
Collapse
|
28
|
AKBULUT S, KÜÇÜKAKÇALI Z, ÇOLAK C. XGBoost modeli ile gen dizileme verilerine dayalı kolorektal kanserin sınıflandırılması: Bir halk sağlığı bilişimi uygulaması. CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1128653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Amaç: Bu çalışma, bir makine öğrenmesi yöntemi olan XGBoost yöntemi ile açık erişimli kolorektal kanser gen verilerini sınıflandırmayı ve temel genleri tanımlamayı amaçlamaktadır.
Gereç ve Yöntem: Çalışmada açık erişimli kolorektal kanser gen veri seti kullanıldı. Veri seti, sağlıklı kontrollerden 10 mukozanın ve kolorektal kanserli 12 hastanın kolon mukozasının gen dizileme sonuçlarını içeriyordu. Hastalığı sınıflandırmak için makine öğrenmesi yöntemlerinden biri olan XGboost kullanıldı. Model performansı için doğruluk, dengelenmiş doğruluk, duyarlılık, seçicilik, pozitif tahmin değeri ve negatif tahmin değeri performans metrikleri değerlendirildi.
Bulgular: Değişken seçim yöntemine göre 17 gen seçilmiş ve bu girdi değişkenleri ile modelleme yapılmıştır. Modelleme sonuçlarından elde edilen doğruluk, dengeli doğruluk, duyarlılık, özgüllük, pozitif tahmin değeri, negatif tahmin değeri ve F1 puanı sırasıyla %95.5, %95.8, %91.7, %1, %1 ve %90.9 ve %95.7 idi. XGboost tekniği sonucundan elde edilen değişken önemliliklerine göre, CYR61, NR4A, FOSB ve NR4A2 genleri kolorektal kanser için biyolojik belirteçler olarak kullanılabilir.
Sonuç: Bu araştırma sonucunda kolorektal kanserle bağlantılı olabilecek genlerin yanı sıra hastalığa yönelik genetik biyobelirteçler de belirlendi. Gelecekte, tespit edilen genlerin güvenilirliği doğrulanabilir, bu genlere dayalı olarak terapötik prosedürler oluşturulabilir ve klinik pratikteki yararları belgelenebilir.
Collapse
|
29
|
Vins N, Sugathan S, Al Menhali A, Karam SM. Overgrowth of Squamocolumnar Junction and Dysregulation of Stem Cell Lineages in the Stomach of Vitamin A-Deficient Mice. Nutrients 2022; 14:nu14163334. [PMID: 36014840 PMCID: PMC9412427 DOI: 10.3390/nu14163334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
Junctional epithelia are common sites for pathological transformations. In mice, the stratified epithelium of the forestomach joins the simple glandular epithelium of the cardia at the limiting ridge. We previously demonstrated the expression of vitamin A receptors in the gastric stem/progenitor cells and their progeny and found that excess retinoic acid enhances cellular dynamics of gastric epithelium. This study examines how deficiency of vitamin A would alter gastric epithelial stem cell lineages. Three-week-old mice of both genders were weaned and fed with a vitamin A deficient (VAD) diet for 4 or 8 months. Sex- and weight-matched littermate mice received a standard (control) diet. To label S-phase cells, all mice received a single intraperitoneal injection of 5-bromo-2-deoxyuridine before being euthanized. Stomach tissues were processed for microscopic examination and protein analysis to investigate stem cell lineages using different stains, lectins, or antibodies. The Student’s t-test was used to compare quantified data showing differences between control and VAD groups. Eight-month-vitamin-A deficiency caused enlarged forestomach and overgrowth of the squamocolumnar junction with metaplastic and dysplastic cardiac glands, formation of intramucosal cysts, loss of surface mucosal integrity, increased amount of luminal surface mucus, and upregulation of trefoil factor 1 and H+,K+-ATPase. These changes were associated with decreased cell proliferation and upregulation of p63. In conclusion, vitamin A is necessary for maintaining gastric epithelial integrity and its deficiency predisposes the mouse stomach to precancerous lesions.
Collapse
Affiliation(s)
- Neethu Vins
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Subi Sugathan
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Centre for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Sherif M. Karam
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Bin Sultan Centre for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Correspondence: ; Tel.: +971-3-713-7493
| |
Collapse
|
30
|
Lungova V, Wendt K, Thibeault SL. Exposure to e-cigarette vapor extract induces vocal fold epithelial injury and triggers intense mucosal remodeling. Dis Model Mech 2022; 15:dmm049476. [PMID: 35770504 PMCID: PMC9438930 DOI: 10.1242/dmm.049476] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 06/24/2022] [Indexed: 11/20/2022] Open
Abstract
Vaping has been reported to cause acute epiglottitis, a life-threatening airway obstruction induced by direct epithelial injury and subsequent inflammatory reaction. Here, we show that we were able to recapitulate this phenomenon in vitro. Exposure of human engineered vocal fold (VF) mucosae to 0.5% and 5% electronic cigarette (e-cigarette) vapor extract (ECVE) for 1 week induced cellular damage of luminal cells, disrupting homeostasis and innate immune responses. Epithelial erosion was likely caused by accumulation of solvents and lipid particles in the cytosol and intercellular spaces, which altered lipid metabolism and plasma membrane properties. Next, we investigated how the mucosal cells responded to the epithelial damage. We withdrew the ECVE from the experimental system and allowed VF mucosae to regenerate for 1, 3 and 7 days, which triggered intense epithelial remodeling. The epithelial changes included expansion of P63 (TP63)-positive basal cells and cytokeratin 14 (KRT14) and laminin subunit α-5 (LAMA5) deposition, which might lead to local basal cell hyperplasia, hyperkeratinization and basement membrane thickening. In summary, vaping presents a threat to VF mucosal health and airway protection, thereby raising further concerns over the safety of e-cigarette use. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Vlasta Lungova
- Department of Surgery, University of Wisconsin-Madison, 5105 WIMR Madison, WI 53705, USA
| | - Kristy Wendt
- Department of Surgery, University of Wisconsin-Madison, 5105 WIMR Madison, WI 53705, USA
| | - Susan L. Thibeault
- Department of Surgery, University of Wisconsin-Madison, 5103 WIMR, Madison, WI 53705, USA
| |
Collapse
|
31
|
Herminghaus A, Kozlov AV, Szabó A, Hantos Z, Gylstorff S, Kuebart A, Aghapour M, Wissuwa B, Walles T, Walles H, Coldewey SM, Relja B. A Barrier to Defend - Models of Pulmonary Barrier to Study Acute Inflammatory Diseases. Front Immunol 2022; 13:895100. [PMID: 35874776 PMCID: PMC9300899 DOI: 10.3389/fimmu.2022.895100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Andrey V. Kozlov
- L Boltzmann Institute for Traumatology in Cooperation with AUVA and Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department of Human Pathology , IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Zoltán Hantos
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Mahyar Aghapour
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Bianka Wissuwa
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, Magdeburg University Medicine, Magdeburg, Germany
| | - Heike Walles
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- *Correspondence: Borna Relja,
| |
Collapse
|
32
|
Perilla Fruit Water Extract Attenuates Inflammatory Responses and Alleviates Neutrophil Recruitment via MAPK/JNK-AP-1/c-Fos Signaling Pathway in ARDS Animal Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4444513. [PMID: 35815275 PMCID: PMC9262517 DOI: 10.1155/2022/4444513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 06/10/2022] [Indexed: 11/18/2022]
Abstract
Airway respiratory distress syndrome (ARDS) is usually caused by a severe pulmonary infection. However, there is currently no effective treatment for ARDS. Traditional Chinese medicine (TCM) has been shown to effectively treat inflammatory lung diseases, but a clear mechanism of action of TCM is not available. Perilla fruit water extract (PFWE) has been used to treat cough, excessive mucus production, and some pulmonary diseases. Thus, we propose that PFWE may be able to reduce lung inflammation and neutrophil infiltration in a lipopolysaccharide (LPS)-stimulated murine model. C57BL/6 mice were stimulated with LPS (10 μg/mouse) by intratracheal (IT) injection and treated with three doses of PFWE (2, 5, and 8 g/kg) by intraperitoneal (IP) injections. To investigate possible mechanisms, A549 cells were treated with PFWE and stimulated with LPS. Our results showed that PFWE decreased airway resistance, neutrophil infiltration, vessel permeability, and interleukin (IL)-6 and chemokine (C-C motif) ligand 2 (CCL2/MCP-1) expressions in vivo. In addition, the PFWE inhibited the expression of IL-6, CCL2/MCP-1, chemokine (CXC motif) ligand 1 (CXCL1/GROα), and IL-8 in vitro. Moreover, PFWE also inhibited the MAPK/JNK-AP-1/c-Fos signaling pathway in A549 cells. In conclusion, we demonstrated that PFWE attenuated pro-inflammatory cytokine and chemokine levels and downregulated neutrophil recruitment through the MAPK/JNK-AP-1/c-Fos pathway. Thus, PFWE can be a potential drug to assist the treatment of ARDS.
Collapse
|
33
|
Moreno-Mendieta S, Guillén D, Vasquez-Martínez N, Hernández-Pando R, Sánchez S, Rodríguez-Sanoja R. Understanding the Phagocytosis of Particles: the Key for Rational Design of Vaccines and Therapeutics. Pharm Res 2022; 39:1823-1849. [PMID: 35739369 DOI: 10.1007/s11095-022-03301-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/23/2022] [Indexed: 12/17/2022]
Abstract
A robust comprehension of phagocytosis is crucial for understanding its importance in innate immunity. A detailed description of the molecular mechanisms that lead to the uptake and clearance of endogenous and exogenous particles has helped elucidate the role of phagocytosis in health and infectious or autoimmune diseases. Furthermore, knowledge about this cellular process is important for the rational design and development of particulate systems for the administration of vaccines or therapeutics. Depending on these specific applications and the required biological responses, particles must be designed to encourage or avoid their phagocytosis and prolong their circulation time. Functionalization with specific polymers or ligands and changes in the size, shape, or surface of particles have important effects on their recognition and internalization by professional and nonprofessional phagocytes and have a major influence on their fate and safety. Here, we review the phagocytosis of particles intended to be used as carrier or delivery systems for vaccines or therapeutics, the cells involved in this process depending on the route of administration, and the strategies employed to obtain the most desirable particles for each application through the manipulation of their physicochemical characteristics. We also offer a view of the challenges and potential opportunities in the field and give some recommendations that we expect will enable the development of improved approaches for the rational design of these systems.
Collapse
Affiliation(s)
- Silvia Moreno-Mendieta
- Consejo Nacional de Ciencia y Tecnología (CONACyT), Ciudad de México, Mexico. .,Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| | - Daniel Guillén
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Nathaly Vasquez-Martínez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.,Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Sergio Sánchez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico
| | - Romina Rodríguez-Sanoja
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, 04510, Ciudad de México, Mexico.
| |
Collapse
|
34
|
Shu Y, Jiang H, Yuen CNT, Wang W, He J, Zhang H, Liu G, Wei L, Chen L, Wu H. Microcystin-leucine arginine induces skin barrier damage and reduces resistance to pathogenic bacteria in Lithobates catesbeianus tadpoles. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113584. [PMID: 35512477 DOI: 10.1016/j.ecoenv.2022.113584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 06/14/2023]
Abstract
Despite the importance of the skin mucosal barrier and commensal microbiota for the health of amphibians, the potential of environmental contaminants to disrupt the skin mucosal barrier and microbiota have rarely been studied in toxicology. In this study, tadpoles (Lithobates catesbeianus) were exposed to 0, 0.5, and 2 μg/L of microcystin-leucine arginine (MC-LR) for 30 days to explore the impacts of environmentally realistic MC-LR concentrations on the physical skin barrier, immune barrier, commensal microbiota, and skin resistance to pathogenic bacterial invasion. MC-LR exposure significantly reduced the collagen fibrils in the dermis of skin tissues and down-regulated tight junction and stratum corneum-related gene transcriptions, suggesting the damage caused by MC-LR to the physical barrier of the skin. Increased skin eosinophils and upregulated transcriptions of inflammation-related genes in the exposed tadpoles underline the development of skin inflammation resulting from MC-LR exposure even at environmentally realistic concentrations. Comparative transcriptome and immunobiochemical analyses found that antimicrobial peptides (Brevinin-1PLc, Brevinin-2GHc, and Ranatuerin-2PLa) and lysozyme were down-regulated in the exposed groups, while complement, pattern recognition receptor, and specific immune processes were up-regulated. However, the content of endotoxin lipopolysaccharide produced by bacteria increased in a dose-dependent pattern. The disc diffusion test showed a reduced ability of skin supernatant to inhibit pathogenic bacteria in the exposed groups. Analysis of microbial 16 S rRNA gene by high-throughput sequencing revealed that MC-LR interfered with the abundance, composition, and diversity of the skin commensal microbiota, which favored the growth of pathogen-containing genera Rhodococcus, Acinetobacter, and Gordonibacter. In summary, the current study provides the first clues about the impact of MC-LR on the integrity and function of skin barrier of amphibians. These new toxicological evidences can facilitate a more comprehensive evaluation of the ecological risk of MC-LR to amphibians.
Collapse
Affiliation(s)
- Yilin Shu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Huiling Jiang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Calista N T Yuen
- State Key Laboratory in Marine Pollution, City University of Hong Kong, Department of Chemistry, Hong Kong, China
| | - Wenchao Wang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Jun He
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Huijuan Zhang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Guangxuan Liu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Luting Wei
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China
| | - Lianguo Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| | - Hailong Wu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241002, China.
| |
Collapse
|
35
|
RNA Sequencing Reveals the Upregulation of FOXO Signaling Pathway in Porphyromonas gingivalis Persister-Treated Human Gingival Epithelial Cells. Int J Mol Sci 2022; 23:ijms23105728. [PMID: 35628542 PMCID: PMC9146424 DOI: 10.3390/ijms23105728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 02/04/2023] Open
Abstract
Porphyromonas gingivalis as the keystone periodontopathogen plays a critical role in the pathogenesis of periodontitis, and crucially accounts for inflammatory comorbidities such as cardiovascular disease and Alzheimer's disease. We recently identified the existence of P. gingivalis persisters and revealed the unforeseen perturbation of innate response in human gingival epithelial cells (HGECs) due to these noxious persisters. Herein, RNA sequencing revealed how P. gingivalis persisters affected the expression profile of cytokine genes and related signaling pathways in HGECs. Results showed that metronidazole-treated P. gingivalis persisters (M-PgPs) impaired the innate host defense of HGECs, in a similar fashion to P. gingivalis. Notably, over one thousand differentially expressed genes were identified in HGECs treated with M-PgPs or P. gingivalis with reference to the controls. Gene Ontology and KEGG pathway analysis demonstrated significantly enriched signaling pathways, such as FOXO. Importantly, the FOXO1 inhibitor rescued the M-PgP-induced disruption of cytokine expression. This study suggests that P. gingivalis persisters may perturb innate host defense, through the upregulation of the FOXO signaling pathway. Thus, the current findings could contribute to developing new approaches to tackling P. gingivalis persisters for the effective control of periodontitis and P. gingivalis-related inflammatory comorbidities.
Collapse
|
36
|
Liu J, Wei Q, Wang Z, Sun X, He QY. Proteomic Study of the Adaptive Mechanism of Ciprofloxacin-Resistant Staphylococcus aureus to the Host Environment. J Proteome Res 2022; 21:1537-1547. [PMID: 35594371 DOI: 10.1021/acs.jproteome.2c00140] [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: 11/28/2022]
Abstract
Antibiotic-resistant bacteria can escape host immune killing and settle in the host to form persistent infections. In this study we investigated the adaptive mechanism of resistant Staphylococcus aureus to the host environment by data-independent acquisition-based quantitative proteomics and functional validation. The growth curve and minimum inhibitory concentration (MIC) indicated that ciprofloxacin-resistant (Cip-R) S. aureus showed a survival advantage over sensitive strains. Cip-R also exhibited a stronger invasion and biofilm formation ability than sensitive bacteria. Cip-R stimulation resulted in the improved production of inflammatory factors of the host cells. Proteomics study combined with biochemical validations showed that Cip-R obtained adaptability to the host via upregulation of the tricarboxylic acid cycle (TCA cycle) and downregulation of ribosome metabolism and protein folding to maintain energy to support Cip-R's survival. Thus, this study will help us to further explain the growth strategy of resistant bacteria to adapt to the host environment, and provide important information for the development of new antibacterial drugs.
Collapse
Affiliation(s)
- Jiajia Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuxia Wei
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhen Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xuesong Sun
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
37
|
Williams TC, Loo SL, Nichol KS, Reid AT, Veerati PC, Esneau C, Wark PAB, Grainge CL, Knight DA, Vincent T, Jackson CL, Alton K, Shimkets RA, Girkin JL, Bartlett NW. IL-25 blockade augments antiviral immunity during respiratory virus infection. Commun Biol 2022; 5:415. [PMID: 35508632 PMCID: PMC9068710 DOI: 10.1038/s42003-022-03367-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
IL-25 is implicated in the pathogenesis of viral asthma exacerbations. However, the effect of IL-25 on antiviral immunity has yet to be elucidated. We observed abundant expression and colocalization of IL-25 and IL-25 receptor at the apical surface of uninfected airway epithelial cells and rhinovirus infection increased IL-25 expression. Analysis of immune transcriptome of rhinovirus-infected differentiated asthmatic bronchial epithelial cells (BECs) treated with an anti-IL-25 monoclonal antibody (LNR125) revealed a re-calibrated response defined by increased type I/III IFN and reduced expression of type-2 immune genes CCL26, IL1RL1 and IL-25 receptor. LNR125 treatment also increased type I/III IFN expression by coronavirus infected BECs. Exogenous IL-25 treatment increased viral load with suppressed innate immunity. In vivo LNR125 treatment reduced IL-25/type 2 cytokine expression and increased IFN-β expression and reduced lung viral load. We define a new immune-regulatory role for IL-25 that directly inhibits virus induced airway epithelial cell innate anti-viral immunity.
Collapse
Affiliation(s)
- Teresa C Williams
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Su-Ling Loo
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Kristy S Nichol
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Andrew T Reid
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Punnam C Veerati
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Camille Esneau
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Peter A B Wark
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Christopher L Grainge
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Darryl A Knight
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
- UBC Providence Health Care Research Institute, Vancouver, BC, Canada
- Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Thomas Vincent
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Kirby Alton
- Abeome Corporation/Lanier Biotherapeutics, Athens, GA, USA
| | | | - Jason L Girkin
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Nathan W Bartlett
- The University of Newcastle and Hunter Medical Research Institute, Newcastle, NSW, Australia.
| |
Collapse
|
38
|
Garcia-Arcos I, Park SS, Mai M, Alvarez-Buve R, Chow L, Cai H, Baumlin-Schmid N, Agudelo CW, Martinez J, Kim MD, Dabo AJ, Salathe M, Goldberg IJ, Foronjy RF. LRP1 loss in airway epithelium exacerbates smoke-induced oxidative damage and airway remodeling. J Lipid Res 2022; 63:100185. [PMID: 35202607 PMCID: PMC8953659 DOI: 10.1016/j.jlr.2022.100185] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
The LDL receptor-related protein 1 (LRP1) partakes in metabolic and signaling events regulated in a tissue-specific manner. The function of LRP1 in airways has not been studied. We aimed to study the function of LRP1 in smoke-induced disease. We found that bronchial epithelium of patients with chronic obstructive pulmonary disease and airway epithelium of mice exposed to smoke had increased LRP1 expression. We then knocked out LRP1 in human bronchial epithelial cells in vitro and in airway epithelial club cells in mice. In vitro, LRP1 knockdown decreased cell migration and increased transforming growth factor β activation. Tamoxifen-inducible airway-specific LRP1 knockout mice (club Lrp1-/-) induced after complete lung development had increased inflammation in the bronchoalveolar space and lung parenchyma at baseline. After 6 months of smoke exposure, club Lrp1-/- mice showed a combined restrictive and obstructive phenotype, with lower compliance, inspiratory capacity, and forced expiratory volume0.05/forced vital capacity than WT smoke-exposed mice. This was associated with increased values of Ashcroft fibrotic index. Proteomic analysis of room air exposed-club Lrp1-/- mice showed significantly decreased levels of proteins involved in cytoskeleton signaling and xenobiotic detoxification as well as decreased levels of glutathione. The proteome fingerprint created by smoke eclipsed many of the original differences, but club Lrp1-/- mice continued to have decreased lung glutathione levels and increased protein oxidative damage and airway cell proliferation. Therefore, LRP1 deficiency leads to greater lung inflammation and damage and exacerbates smoke-induced lung disease.
Collapse
Affiliation(s)
- Itsaso Garcia-Arcos
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA.
| | - Sangmi S Park
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michelle Mai
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Roger Alvarez-Buve
- Respiratory Department, Hospital University Arnau de Vilanova and Santa Maria, IRB Lleida, University of Lleida, Lleida, Catalonia, Spain
| | - Lillian Chow
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Huchong Cai
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | | | - Christina W Agudelo
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Jennifer Martinez
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Michael D Kim
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Abdoulaye J Dabo
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| | - Matthias Salathe
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Ira J Goldberg
- Department of Medicine, NYU Langone School of Medicine, New York, NY, USA
| | - Robert F Foronjy
- Departments of Medicine and Cell Biology, SUNY Downstate Medical Center, New York, NY, USA
| |
Collapse
|
39
|
Kang MS, Kim SH, Yang MJ, Kim HY, Kim IH, Kang JW, Choi HS, Jin SW, Park EJ. Polyhexamethylene guanidine phosphate-induced necrosis may be linked to pulmonary fibrosis. Toxicol Lett 2022; 362:1-16. [DOI: 10.1016/j.toxlet.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/05/2022] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
40
|
Inhibition of the IFN-α JAK/STAT Pathway by MERS-CoV and SARS-CoV-1 Proteins in Human Epithelial Cells. Viruses 2022; 14:v14040667. [PMID: 35458397 PMCID: PMC9032603 DOI: 10.3390/v14040667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/04/2022] [Accepted: 03/17/2022] [Indexed: 12/10/2022] Open
Abstract
Coronaviruses (CoVs) have caused several global outbreaks with relatively high mortality rates, including Middle East Respiratory Syndrome coronavirus (MERS)-CoV, which emerged in 2012, and Severe Acute Respiratory Syndrome (SARS)-CoV-1, which appeared in 2002. The recent emergence of SARS-CoV-2 highlights the need for immediate and greater understanding of the immune evasion mechanisms used by CoVs. Interferon (IFN)-α is the body's natural antiviral agent, but its Janus kinase/signal transducer and activators of transcription (JAK/STAT) signalling pathway is often antagonized by viruses, thereby preventing the upregulation of essential IFN stimulated genes (ISGs). Therapeutic IFN-α has disappointingly weak clinical responses in MERS-CoV and SARS-CoV-1 infected patients, indicating that these CoVs inhibit the IFN-α JAK/STAT pathway. Here we show that in lung alveolar A549 epithelial cells expression of MERS-CoV-nsp2 and SARS-CoV-1-nsp14, but not MERS-CoV-nsp5, increased basal levels of total and phosphorylated STAT1 & STAT2 protein, but reduced IFN-α-mediated phosphorylation of STAT1-3 and induction of MxA. While MERS-CoV-nsp2 and SARS-CoV-1-nsp14 similarly increased basal levels of STAT1 and STAT2 in bronchial BEAS-2B epithelial cells, unlike in A549 cells, they did not enhance basal pSTAT1 nor pSTAT2. However, both viral proteins reduced IFN-α-mediated induction of pSTAT1-3 and ISGs (MxA, ISG15 and PKR) in BEAS-2B cells. Furthermore, even though IFN-α-mediated induction of pSTAT1-3 was not affected by MERS-CoV-nsp5 expression in BEAS-2B cells, downstream ISG induction was reduced, revealing that MERS-CoV-nsp5 may use an alternative mechanism to reduce antiviral ISG induction in this cell line. Indeed, we subsequently discovered that all three viral proteins inhibited STAT1 nuclear translocation in BEAS-2B cells, unveiling another layer of inhibition by which these viral proteins suppress responses to Type 1 IFNs. While these observations highlight cell line-specific differences in the immune evasion effects of MERS-CoV and SARS-CoV-1 proteins, they also demonstrate the broad spectrum of immune evasion strategies these deadly coronaviruses use to stunt antiviral responses to Type IFN.
Collapse
|
41
|
Saenz-de-Juano MD, Silvestrelli G, Weber A, Röhrig C, Schmelcher M, Ulbrich SE. Inflammatory Response of Primary Cultured Bovine Mammary Epithelial Cells to Staphylococcus aureus Extracellular Vesicles. BIOLOGY 2022; 11:biology11030415. [PMID: 35336789 PMCID: PMC8944978 DOI: 10.3390/biology11030415] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 11/22/2022]
Abstract
Simple Summary Mastitis, the inflammation of the mammary gland, is one of the most common and costly diseases worldwide, and Staphylococcus aureus (S. aureus) is among the most prevalent microorganisms that cause it. To obtain new insights into S. aureus mammary gland infections, we have isolated S. aureus extracellular vesicles to challenge in vitro primary bovine mammary epithelial cells. Despite the toxic content of the vesicles, we observed only a minor pro-inflammatory response. The latter can contribute to the explanation of how S. aureus evades mammary epithelial defence mechanisms and successfully colonizes the mammary gland. Abstract In dairy cows, Staphylococcus aureus (S. aureus) is among the most prevalent microorganisms worldwide, causing mastitis, an inflammation of the mammary gland. Production of extracellular vesicles (EVs) is a common feature of S. aureus strains, which contributes to its pathogenesis by delivering bacterial effector molecules to host cells. In the current study, we evaluated the differences between five S. aureus mastitis isolates regarding their EV production. We found that different mastitis-related S. aureus strains differ in their behaviour of shedding EVs, with M5512VL producing the largest amount of EVs containing alpha-haemolysin, a strong cytotoxic agent. We stimulated primary cultured bovine mammary epithelial cells (pbMECs) with EVs from the S. aureus strain M5512VL. After 24 h of incubation, we observed a moderate increase in gene expression of tumour necrosis factor-alpha (TNF-α) but, surprisingly, a lack of an associated pronounced pro-inflammatory response. Our results contribute to understanding the damaging nature of S. aureus in its capacity to effectively affect mammary epithelial cells.
Collapse
Affiliation(s)
- Mara D. Saenz-de-Juano
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
| | - Giulia Silvestrelli
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
| | - Andres Weber
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
| | - Christian Röhrig
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland; (C.R.); (M.S.)
| | - Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland; (C.R.); (M.S.)
| | - Susanne E. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
- Correspondence:
| |
Collapse
|
42
|
Sohrabi M, Sahu B, Kaur H, Hasler WA, Prakash A, Combs CK. Gastrointestinal Changes and Alzheimer's Disease. Curr Alzheimer Res 2022; 19:335-350. [PMID: 35718965 PMCID: PMC10497313 DOI: 10.2174/1567205019666220617121255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/08/2022] [Accepted: 03/06/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND There is a well-described mechanism of communication between the brain and gastrointestinal system in which both organs influence the function of the other. This bi-directional communication suggests that disease in either organ may affect function in the other. OBJECTIVE To assess whether the evidence supports gastrointestinal system inflammatory or degenerative pathophysiology as a characteristic of Alzheimer's disease (AD). METHODS A review of both rodent and human studies implicating gastrointestinal changes in AD was performed. RESULTS Numerous studies indicate that AD changes are not unique to the brain but also occur at various levels of the gastrointestinal tract involving both immune and neuronal changes. In addition, it appears that numerous conditions and diseases affecting regions of the tract may communicate to the brain to influence disease. CONCLUSION Gastrointestinal changes represent an overlooked aspect of AD, representing a more system influence of this disease.
Collapse
Affiliation(s)
- Mona Sohrabi
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Bijayani Sahu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Harpreet Kaur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Wendie A Hasler
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Atish Prakash
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| | - Colin K Combs
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202-9037, USA
| |
Collapse
|
43
|
Kubczak M, Michlewska S, Bryszewska M, Aigner A, Ionov M. Nanoparticles for local delivery of siRNA in lung therapy. Adv Drug Deliv Rev 2021; 179:114038. [PMID: 34742826 DOI: 10.1016/j.addr.2021.114038] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 02/07/2023]
Abstract
An overview of the application of natural and synthetic, non-viral vectors for oligonucleotide delivery into the lung is presented in this review, with a special focus on lung cancer. Due to the specificity of the respiratory tract, its structure and natural barriers, the administration of drugs (especially those based on nucleic acids) is a particular challenge. Among widely tested non-viral drug and oligonucleotides carriers, synthetic polymers seem to be most promising. Unique properties of these nanoparticles allow for essentially unlimited possibilities regarding their design and modification. This gives hope that optimal nanoparticles with ideal nucleic acid carrier properties for lung cancer therapy will eventually emanate.
Collapse
|
44
|
Wei Z, Hao C, Srinivasagan R, Wu H, Chen JK, Fan X. Mitotic Activation Around Wound Edges and Epithelialization Repair in UVB-Induced Capsular Cataracts. Invest Ophthalmol Vis Sci 2021; 62:29. [PMID: 34967856 PMCID: PMC8727316 DOI: 10.1167/iovs.62.15.29] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Ultraviolet B (UVB) has been well documented to induce capsular cataracts; however, the mechanism of the lens epithelial cell-mediated repair process after UVB irradiation is not fully understood. The purpose of this study was to better understand lens epithelial cell repair after UVB-induced epithelium damage. Method C57BL/6J mice were irradiated by various doses of UVB. Lens morphology and lens capsule opacity were monitored by slit lamp, darkfield microscopy, and phase-contrast microscopy. Lens epithelial cell mitotic activation and cell apoptosis were measured by immunohistochemistry. Lens epithelial ultrastructure was analyzed by transmission electron microscopy. Results UVB irradiation above a dose of 2.87 kJ/m2 triggered lens epithelial cell apoptosis and subcapsular cataract formation, with a ring-shaped structure composed of multilayered epithelial cell clusters manifesting a dense ring-shaped capsular cataract. The epithelial cells immediately outside the edge of the ring-shaped aggregates transitioned to mitotically active cells and performed wound healing through the epithelialization process. However, repairs ceased when lens epithelial cells made direct contact, and scar-like tissue in the center of the anterior capsule remained even by 6 months after UVB irradiation. Conclusions Our present study demonstrates that normally quiescent lens epithelial cells can be reactivated for epithelialization repair in response to UV-induced damage.
Collapse
Affiliation(s)
- Zongbo Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Caili Hao
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Ramkumar Srinivasagan
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States
| | - Hongli Wu
- Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas, United States.,North Texas Eye Research Institute, Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Jian-Kang Chen
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Xingjun Fan
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| |
Collapse
|
45
|
Anil A, Apte S, Joseph J, Parthasarathy A, Madhavan S, Banerjee A. Pyruvate Oxidase as a Key Determinant of Pneumococcal Viability during Transcytosis across Brain Endothelium. J Bacteriol 2021; 203:e0043921. [PMID: 34606370 PMCID: PMC8604078 DOI: 10.1128/jb.00439-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/23/2021] [Indexed: 01/23/2023] Open
Abstract
Streptococcus pneumoniae invades a myriad of host tissues following efficient breaching of cellular barriers. However, strategies adopted by pneumococcus for evasion of host intracellular defenses governing successful transcytosis across host cellular barriers remain elusive. In this study, using brain endothelium as a model host barrier, we observed that pneumococcus containing endocytic vacuoles (PCVs), formed following S. pneumoniae internalization into brain microvascular endothelial cells (BMECs), undergo early maturation and acidification, with a major subset acquiring lysosome-like characteristics. Exploration of measures that would preserve pneumococcal viability in the lethal acidic pH of these lysosome-like vacuoles revealed a critical role of the two-component system response regulator, CiaR, which was previously implicated in induction of acid tolerance response. Pyruvate oxidase (SpxB), a key sugar-metabolizing enzyme that catalyzes oxidative decarboxylation of pyruvate to acetyl phosphate, was found to contribute to acid stress tolerance, presumably via acetyl phosphate-mediated phosphorylation and activation of CiaR, independent of its cognate kinase CiaH. Hydrogen peroxide, the by-product of an SpxB-catalyzed reaction, was also found to improve pneumococcal intracellular survival by oxidative inactivation of lysosomal cysteine cathepsins, thus compromising the degradative capacity of the host lysosomes. As expected, a ΔspxB mutant was found to be significantly attenuated in its ability to survive inside the BMEC endocytic vacuoles, reflecting its reduced transcytosis ability. Collectively, our studies establish SpxB as an important virulence determinant facilitating pneumococcal survival inside host cells, ensuring successful trafficking across host cellular barriers. IMPORTANCE Host cellular barriers have innate immune defenses to restrict microbial passage into sterile compartments. Here, by focusing on the blood-brain barrier endothelium, we investigated mechanisms that enable Streptococcus pneumoniae to traverse through host barriers. Pyruvate oxidase, a pneumococcal sugar-metabolizing enzyme, was found to play a crucial role in this via generation of acetyl phosphate and hydrogen peroxide. A two-pronged approach consisting of acetyl phosphate-mediated activation of acid tolerance response and hydrogen peroxide-mediated inactivation of lysosomal enzymes enabled pneumococci to maintain viability inside the degradative vacuoles of the brain endothelium for successful transcytosis across the barrier. Thus, pyruvate oxidase is a key virulence determinant and can potentially serve as a viable candidate for therapeutic interventions for better management of invasive pneumococcal diseases.
Collapse
Affiliation(s)
- Anjali Anil
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Shruti Apte
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Jincy Joseph
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Akhila Parthasarathy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Shilpa Madhavan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| | - Anirban Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, Maharashtra, India
| |
Collapse
|
46
|
Barbara G, Barbaro MR, Fuschi D, Palombo M, Falangone F, Cremon C, Marasco G, Stanghellini V. Corrigendum: Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier. Front Nutr 2021; 8:790387. [PMID: 34790692 PMCID: PMC8591313 DOI: 10.3389/fnut.2021.790387] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/18/2022] Open
Affiliation(s)
- Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Marta Palombo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
47
|
Ferrera-González J, Francés-Soriano L, Galiana-Roselló C, González-Garcia J, González-Béjar M, Fröhlich E, Pérez-Prieto J. Initial Biological Assessment of Upconversion Nanohybrids. Biomedicines 2021; 9:1419. [PMID: 34680536 PMCID: PMC8533627 DOI: 10.3390/biomedicines9101419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
Nanoparticles for medical use should be non-cytotoxic and free of bacterial contamination. Upconversion nanoparticles (UCNPs) coated with cucurbit[7]uril (CB[7]) made by combining UCNPs free of oleic acid, here termed bare UCNPs (UCn), and CB[7], i.e., UC@CB[7] nanohybrids, could be used as photoactive inorganic-organic hybrid scaffolds for biological applications. UCNPs, in general, are not considered to be highly toxic materials, but the release of fluorides and lanthanides upon their dissolution may cause cytotoxicity. To identify potential adverse effects of the nanoparticles, dehydrogenase activity of endothelial cells, exposed to various concentrations of the UCNPs, was determined. Data were verified by measuring lactate dehydrogenase release as the indicator of loss of plasma membrane integrity, which indicates necrotic cell death. This assay, in combination with calcein AM/Ethidium homodimer-1 staining, identified induction of apoptosis as main mode of cell death for both particles. The data showed that the UCNPs are not cytotoxic to endothelial cells, and the samples did not contain endotoxin contamination. Higher cytotoxicity, however, was seen in HeLa and RAW 264.7 cells. This may be explained by differences in lysosome content and particle uptake rate. Internalization of UCn and UC@CB[7] nanohybrids by cells was demonstrated by NIR laser scanning microscopy.
Collapse
Affiliation(s)
- Juan Ferrera-González
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Orgánica, University of Valencia, Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain; (J.F.-G.); (L.F.-S.); (C.G.-R.); (J.G.-G.)
| | - Laura Francés-Soriano
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Orgánica, University of Valencia, Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain; (J.F.-G.); (L.F.-S.); (C.G.-R.); (J.G.-G.)
- nanoFRET.com, Laboratoire COBRA (Chimie Organique, Bioorganique: Réactivité et Analyse), UMR 6014, CNRS, Université de Rouen Normandie, INSA, CEDEX, 76821 Mont-Saint-Aignan, France
| | - Cristina Galiana-Roselló
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Orgánica, University of Valencia, Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain; (J.F.-G.); (L.F.-S.); (C.G.-R.); (J.G.-G.)
| | - Jorge González-Garcia
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Orgánica, University of Valencia, Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain; (J.F.-G.); (L.F.-S.); (C.G.-R.); (J.G.-G.)
| | - María González-Béjar
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Orgánica, University of Valencia, Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain; (J.F.-G.); (L.F.-S.); (C.G.-R.); (J.G.-G.)
| | - Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Stiftingtalstr. 24, 8010 Graz, Austria
| | - Julia Pérez-Prieto
- Instituto de Ciencia Molecular (ICMol), Departamento de Química Orgánica, University of Valencia, Catedrático José Beltrán, 2, Paterna, 46980 Valencia, Spain; (J.F.-G.); (L.F.-S.); (C.G.-R.); (J.G.-G.)
| |
Collapse
|
48
|
Karmakova ТА, Sergeeva NS, Kanukoev КY, Alekseev BY, Kaprin АD. Kidney Injury Molecule 1 (KIM-1): a Multifunctional Glycoprotein and Biological Marker (Review). Sovrem Tekhnologii Med 2021; 13:64-78. [PMID: 34603757 PMCID: PMC8482821 DOI: 10.17691/stm2021.13.3.08] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Indexed: 12/17/2022] Open
Abstract
KIM-1 (kidney injury molecule 1) is a transmembrane glycoprotein also known as HAVcr-1 and TIM-1 belongs to the T-cell immunoglobulin and mucin domain family (TIM) of proteins. TIM glycoproteins are presented on the immune cells and participate in the regulation of immune reactions. KIM-1 differs from other members of its family in that it is expressed not only by immunocompetent cells but epithelial cells as well. Cellular and humoral effects mediated by KIM-1 are involved in a variety of physiological and pathophysiological processes. Current understanding of the mechanisms determining the participation of KIM-1 in viral invasion, the immune response regulation, adaptive reactions of the kidney epithelium to acute ischemic or toxic injury, in progression of chronic renal diseases, and kidney cancer development have been presented in this review. Data of clinical researches demonstrating the association of KIM-1 with viral diseases and immune disorders have also been analyzed. Potential application of KIM-1 as urinary or serological marker in renal and cardiovascular diseases has been considered.
Collapse
Affiliation(s)
- Т А Karmakova
- Leading Researcher, Department of Predicting the Effectiveness of Conservative Therapy; P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 3, 2 Botkinsky Proezd, Moscow, 125284, Russia
| | - N S Sergeeva
- Professor, Head of the Department of Predicting the Effectiveness of Conservative Therapy; P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 3, 2 Botkinsky Proezd, Moscow, 125284, Russia; Professor, Department of Biology; Pirogov Russian National Research Medical University, 1 Ostrovitianova St., Moscow, 117997, Russia
| | - К Yu Kanukoev
- Urologist, Department of Urology with Chemotherapy; P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 3, 2 Botkinsky Proezd, Moscow, 125284, Russia
| | - B Ya Alekseev
- Professor, Deputy General Director for Science; National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 4 Koroleva St., Obninsk, 249036, Russia
| | - А D Kaprin
- Professor, Academician of the Russian Academy of Sciences, General Director; National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 4 Koroleva St., Obninsk, 249036, Russia
| |
Collapse
|
49
|
Elkhatib SK, Alley J, Jepsen M, Smeins L, Barnes A, Naik S, Ackermann MR, Verhoeven D, Kohut ML. Exercise duration modulates upper and lower respiratory fluid cellularity, antiviral activity, and lung gene expression. Physiol Rep 2021; 9:e15075. [PMID: 34676696 PMCID: PMC8531599 DOI: 10.14814/phy2.15075] [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: 09/05/2021] [Accepted: 09/18/2021] [Indexed: 12/02/2022] Open
Abstract
Exercise has substantial health benefits, but the effects of exercise on immune status and susceptibility to respiratory infections are less clear. Furthermore, there is limited research examining the effects of prolonged exercise on local respiratory immunity and antiviral activity. To assess the upper respiratory tract in response to exercise, we collected nasal lavage fluid (NALF) from human subjects (1) at rest, (2) after 45 min of moderate-intensity exercise, and (3) after 180 min of moderate-intensity exercise. To assess immune responses of the lower respiratory tract, we utilized a murine model to examine the effect of exercise duration on bronchoalveolar lavage (BAL) fluid immune cell content and lung gene expression. NALF cell counts did not change after 45 min of exercise, whereas 180 min significantly increased total cells and leukocytes in NALF. Importantly, fold change in NALF leukocytes correlated with the post-exercise fatigue rating in the 180-min exercise condition. The acellular portion of NALF contained strong antiviral activity against Influenza A in both resting and exercise paradigms. In mice undergoing moderate-intensity exercise, BAL total cells and neutrophils decreased in response to 45 or 90 min of exercise. In lung lobes, increased expression of heat shock proteins suggested that cellular stress occurred in response to exercise. However, a broad upregulation of inflammatory genes was not observed, even at 180 min of exercise. This work demonstrates that exercise duration differentially alters the cellularity of respiratory tract fluids, antiviral activity, and gene expression. These changes in local mucosal immunity may influence resistance to respiratory viruses, including influenza or possibly other pathogens in which nasal mucosa plays a protective role, such as rhinovirus or SARS-CoV-2.
Collapse
Affiliation(s)
- Safwan K. Elkhatib
- Department of KinesiologyIowa State UniversityAmesIowaUSA
- Present address:
Cellular & Integrative PhysiologyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNebraskaUSA
| | - Jessica Alley
- Department of KinesiologyIowa State UniversityAmesIowaUSA
- Program of ImmunobiologyIowa State UniversityAmesIowaUSA
- Present address:
Lineberger Comprehensive Cancer Center, School of MedicineUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Michael Jepsen
- Department of KinesiologyIowa State UniversityAmesIowaUSA
- Present address:
College of Osteopathic MedicineCampbell UniversityLillingtonNorth CarolinaUSA
| | - Laurel Smeins
- Department of KinesiologyIowa State UniversityAmesIowaUSA
| | - Andrew Barnes
- Department of KinesiologyIowa State UniversityAmesIowaUSA
- Present address:
Kirksville College of Osteopathic MedicineA.T. Still UniversityKirksvilleMissouriUSA
| | - Shibani Naik
- Program of ImmunobiologyIowa State UniversityAmesIowaUSA
- Present address:
Arisan Therapeutics11189 Sorrento Valley Rd, Suite 104, San DiegoCaliforniaUSA
| | - Mark R. Ackermann
- Department of Veterinary PathologyCollege of Veterinary MedicineIowa State UniversityAmesIowaUSA
- Present address:
Director, Anatomic Veterinary Pathology DiagnosticsZoetisClear LakeIowa50428USA
| | - David Verhoeven
- Department of Veterinary Microbiology and Preventive MedicineCollege of Veterinary MedicineIowa State UniversityAmesIowaUSA
| | - Marian L. Kohut
- Department of KinesiologyIowa State UniversityAmesIowaUSA
- Program of ImmunobiologyIowa State UniversityAmesIowaUSA
| |
Collapse
|
50
|
Barbara G, Barbaro MR, Fuschi D, Palombo M, Falangone F, Cremon C, Marasco G, Stanghellini V. Inflammatory and Microbiota-Related Regulation of the Intestinal Epithelial Barrier. Front Nutr 2021; 8:718356. [PMID: 34589512 PMCID: PMC8475765 DOI: 10.3389/fnut.2021.718356] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
The intestinal epithelial barrier (IEB) is one of the largest interfaces between the environment and the internal milieu of the body. It is essential to limit the passage of harmful antigens and microorganisms and, on the other side, to assure the absorption of nutrients and water. The maintenance of this delicate equilibrium is tightly regulated as it is essential for human homeostasis. Luminal solutes and ions can pass across the IEB via two main routes: the transcellular pathway or the paracellular pathway. Tight junctions (TJs) are a multi-protein complex responsible for the regulation of paracellular permeability. TJs control the passage of antigens through the IEB and have a key role in maintaining barrier integrity. Several factors, including cytokines, gut microbiota, and dietary components are known to regulate intestinal TJs. Gut microbiota participates in several human functions including the modulation of epithelial cells and immune system through the release of several metabolites, such as short-chain fatty acids (SCFAs). Mediators released by immune cells can induce epithelial cell damage and TJs dysfunction. The subsequent disruption of the IEB allows the passage of antigens into the mucosa leading to further inflammation. Growing evidence indicates that dysbiosis, immune activation, and IEB dysfunction have a role in several diseases, including irritable bowel syndrome (IBS), inflammatory bowel disease (IBD), and gluten-related conditions. Here we summarize the interplay between the IEB and gut microbiota and mucosal immune system and their involvement in IBS, IBD, and gluten-related disorders.
Collapse
Affiliation(s)
- Giovanni Barbara
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Maria Raffaella Barbaro
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniele Fuschi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Marta Palombo
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Falangone
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, University Sapienza, Rome, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vincenzo Stanghellini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.,Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|