1
|
Lv M, Zhao Y, Chang S, Gao Z. Identifying signature genes and their associations with immune cell infiltration in spinal cord injury. IBRO Neurosci Rep 2024; 17:320-328. [PMID: 39430218 PMCID: PMC11490871 DOI: 10.1016/j.ibneur.2024.09.002] [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: 04/21/2024] [Accepted: 09/16/2024] [Indexed: 10/22/2024] Open
Abstract
Background Early detection of spinal cord injury (SCI) is conducive to improving patient outcomes. In addition, many studies have revealed the role of immune cells in the progression or treatment of SCI. The objective of this study was to identify the early signature genes and clarify how they are related to immune cell infiltration in SCI. Methods We analysed and identified early signature genes associated with SCI via bioinformatics analysis of the GSE151371 dataset from the GEO database. These genes were subsequently verified in the GSE33886 dataset and qRT-PCR. Finally, the CIBERSORT algorithm was used to examine the immune cell infiltration in SCI and its relationship with signature genes. Results Seven SCI-related signature genes, including ARG1, RETN, BPI, GGH, CCNB1, HIST1H2AC, and HIST1H2BJ, were identified, and their expression was verified via an external validation cohort and qRT-PCR. Moreover, the ROC curves revealed the diagnostic value of these genes. In addition, on the basis of immune cell infiltration analysis, plasma cells, M0 macrophages, activated CD4+ memory T cells, γδ T cells, naive CD4+ T cells, and resting CD4+ memory T cells may participate in the progression of SCI. Conclusion This study identified seven early signature genes of SCI that may serve as biomarkers for the early diagnosis of SCI and contribute to our understanding of immune changes during the pathology of SCI.
Collapse
Affiliation(s)
- Meng Lv
- Department of Orthopaedics, Shaanxi Provincial People's Hospital (Third Affiliated Hospital of Xi’an Jiaotong University), Xi’an, Shaanxi Province 710068, China
| | - Yingjie Zhao
- Department of Orthopaedics, Shaanxi Provincial People's Hospital (Third Affiliated Hospital of Xi’an Jiaotong University), Xi’an, Shaanxi Province 710068, China
| | - Su’e Chang
- Department of Orthoapedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Zhengchao Gao
- Department of Orthoapedic Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| |
Collapse
|
2
|
Melo-Ferraz A, Miller P, Criado MB, Monteiro MC, Coelho C. Exploring the antimicrobial potential of Leukocyte- and Platelet-Rich Fibrin - an in vitro study. APMIS 2024; 132:859-868. [PMID: 39295296 DOI: 10.1111/apm.13468] [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: 12/21/2023] [Accepted: 09/04/2024] [Indexed: 09/21/2024]
Abstract
This study investigates the antimicrobial properties of leukocyte- and platelet-rich fibrin (L-PRF) against Enterococcus faecalis (ATCC 29212), Pseudomonas aeruginosa (ATCC 27853), and Candida albicans (ATCC 90028). Infections can hinder wound healing posing challenges. L-PRF's potential for regeneration and antimicrobial action has been studied. Considering the increasing concern about antibiotic resistance, assessing the antimicrobial properties of L-PRF provides valuable insights into its potential as a therapeutic agent in postoperative infections. Twenty volunteers were enrolled in the study, following ethical guidelines, and obtaining informed consent. Blood samples were collected and L-PRF was prepared. Microbial suspensions were prepared, and susceptibility testing was performed using the Kirby-Bauer agar diffusion method. The study revealed significant heterogeneity in the susceptibility to L-PRF. All L-PRF membrane samples exhibited antimicrobial activity against P. aeruginosa, with inhibition zones of 13 mm ± 3.85 SD. Enterococcus faecalis displayed inhibition diameter of 7.25 mm ± 5.15 SD. Candida albicans susceptibility to L-PRF varied among samples, with both inhibitory and non-inhibitory results. Results showed varying degrees of antimicrobial activity, particularly against P. aeruginosa, and highlight the complexity of the L-PRF-microorganism interaction. Further investigations are needed to elucidate the clinical implications and optimize the use of L-PRF.
Collapse
Affiliation(s)
- António Melo-Ferraz
- UNIPRO - Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
| | - Paulo Miller
- UNIPRO - Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
| | - Maria Begoña Criado
- 1H-TOXRUN - One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
| | - Maria Céu Monteiro
- 1H-TOXRUN - One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
| | - Cristina Coelho
- UNIPRO - Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), CESPU, CRL, Gandra, Portugal
| |
Collapse
|
3
|
Gómez-Oro C, Latorre MC, Arribas-Poza P, Ibáñez-Escribano A, Baca-Cornejo KR, Gallego-Valle J, López-Escobar N, Mondéjar-Palencia M, Pion M, López-Fernández LA, Mercader E, Pérez-Milán F, Relloso M. Progesterone promotes CXCl2-dependent vaginal neutrophil killing by activating cervical resident macrophage-neutrophil crosstalk. JCI Insight 2024; 9:e177899. [PMID: 39298265 PMCID: PMC11529979 DOI: 10.1172/jci.insight.177899] [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: 12/01/2023] [Accepted: 09/05/2024] [Indexed: 09/21/2024] Open
Abstract
Vaginal infections in women of reproductive age represent a clinical dilemma with significant socioeconomic implications. The current understanding of mucosal immunity failure during early pathogenic invasions that allows the pathogen to grow and thrive is far from complete. Neutrophils infiltrate most tissues following circadian patterns as part of normal repair, regulation of microbiota, or immune surveillance and become more numerous after infection. Neutrophils are responsible for maintaining vaginal immunity. Specific to the vagina, neutrophils continuously infiltrate at high levels, although during ovulation, they retreat to avoid sperm damage and permit reproduction. Here we show that, after ovulation, progesterone promotes resident vaginal macrophage-neutrophil crosstalk by upregulating Yolk sac early fetal organs (FOLR2+) macrophage CXCl2 expression, in a TNFA-patrolling monocyte-derived macrophage-mediated (CX3CR1hiMHCIIhi-mediated) manner, to activate the neutrophils' capacity to eliminate sex-transmitted and opportunistic microorganisms. Indeed, progesterone plays an essential role in conciliating the balance between the commensal microbiota, sperm, and the threat of pathogens because progesterone not only promotes a flurry of neutrophils but also increases neutrophilic fury to restore immunity after ovulation to thwart pathogenic invasion after intercourse. Therefore, modest progesterone dysregulations could lead to a suboptimal neutrophilic response, resulting in insufficient mucosal defense and recurrent unresolved infections.
Collapse
Affiliation(s)
- Carla Gómez-Oro
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Maria C. Latorre
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Patricia Arribas-Poza
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alexandra Ibáñez-Escribano
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Katia R. Baca-Cornejo
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | | - Natalia López-Escobar
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Mabel Mondéjar-Palencia
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | - Marjorie Pion
- Laboratorio de InmunoRegulación, IiSGM, Madrid, Spain
| | - Luis A. López-Fernández
- Laboratorio de Farmacogenética, Grupo de Farmacia Hospitalaria y Farmacogenómica, IiSGM, Madrid, Spain
| | - Enrique Mercader
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Unidad Cirugía Endocrino-metabólica, Servicio de Cirugía General y Aparato Digestivo, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Federico Pérez-Milán
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Unidad de Reproducción Asistida, Servicio de Obstetricia y Ginecología, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Miguel Relloso
- Laboratorio de InmunoReproducción, Grupo Fisiopatología de la mujer, del embarazo, parto y puerperio, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| |
Collapse
|
4
|
Wang J, Amoah S, Stafford JL. A leukocyte immune-type receptor specific polyclonal antibody recognizes goldfish kidney leukocytes and activates the MAPK pathway in isolated goldfish kidney neutrophil-like cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105228. [PMID: 38997096 DOI: 10.1016/j.dci.2024.105228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Leukocyte immune-type receptors (LITRs) belong to a large family of teleost immunoregulatory receptors that share phylogenetic and syntenic relationships with mammalian Fc receptor-like molecules (FCRLs). Recently, several putative stimulatory Carassius auratus (Ca)-LITR transcripts, including CaLITR3, have been identified in goldfish. CaLITR3 has four extracellular immunoglobulin-like (Ig-like) domains, a transmembrane domain containing a positively charged histidine residue, and a short cytoplasmic tail region. Additionally, the calitr3 transcript is highly expressed by goldfish primary kidney neutrophils (PKNs) and macrophages (PKMs). To further investigate the immunoregulatory potential of CaLITR3 in goldfish myeloid cells, we developed and characterized a CaLITR3-epitope-specific polyclonal antibody (anti-CaL3.D1 pAb). We show that the anti-CaL3.D1 pAb stains various hematopoietic cell types within the goldfish kidney, as well as in PKNs and PKMs. Moreover, cross-linking of the anti-CaL3.D1-pAb on PKN membranes induces phosphorylation of p38 and ERK1/2, critical components of the MAPK pathway involved in controlling a wide variety of innate immune effector responses such as NETosis, respiratory burst, and cytokine release. These findings support the stimulatory potential of CaLITR3 proteins as activators of fish granulocytes and pave the way for a more in-depth examination of the immunoregulatory functions of CaLITRs in goldfish myeloid cells.
Collapse
Affiliation(s)
- Jiahui Wang
- Department of Biological Sciences, University of Alberta, Alberta, Canada
| | - Samuel Amoah
- Department of Biological Sciences, University of Alberta, Alberta, Canada
| | - James L Stafford
- Department of Biological Sciences, University of Alberta, Alberta, Canada.
| |
Collapse
|
5
|
Todorova VK, Azhar G, Stone A, Malapati SJ, Che Y, Zhang W, Makhoul I, Wei JY. Neutrophil Biomarkers Can Predict Cardiotoxicity of Anthracyclines in Breast Cancer. Int J Mol Sci 2024; 25:9735. [PMID: 39273682 PMCID: PMC11395913 DOI: 10.3390/ijms25179735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/29/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
Doxorubicin (DOX), a commonly used anticancer agent, causes cardiotoxicity that begins with the first dose and may progress to heart failure years after treatment. An inflammatory response associated with neutrophil recruitment has been recognized as a mechanism of DOX-induced cardiotoxicity. This study aimed to validate mRNA expression of the previously identified biomarkers of DOX-induced cardiotoxicity, PGLYRP1, CAMP, MMP9, and CEACAM8, and to assay their protein expression in the peripheral blood of breast cancer patients. Blood samples from 40 breast cancer patients treated with DOX-based chemotherapy were collected before and after the first chemotherapy cycle and > 2 years after treatment. The protein and gene expression of PGLYRP1/Tag7, CAMP/LL37, MMP9/gelatinase B, and CEACAM8/CD66b were determined using ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of each candidate biomarker. Patients with cardiotoxicity (n = 20) had significantly elevated levels of PGLYRP1, CAMP, MMP9, and CEACAM8 at baseline, after the first dose of DOX-based chemotherapy, and at > 2 years after treatment relative to patients without cardiotoxicity (n = 20). The first dose of DOX induced significantly higher levels of all examined biomarkers in both groups of patients. At > 2 years post treatment, the levels of all but MMP9 dropped below the baseline. There was a good correlation between the expression of mRNA and the target proteins. We demonstrate that circulating levels of PGLYRP1, CAMP, MMP9, and CEACAM8 can predict the cardiotoxicity of DOX. This novel finding may be of value in the early identification of patients at risk for cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Annjanette Stone
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Sindhu J Malapati
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Yingni Che
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Zhang
- Department of Mathematics and Statistics, University of Arkansas at Little Rock, Little Rock, AR 72205, USA
| | - Issam Makhoul
- Division of Hematology/Oncology, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
6
|
Manole CG, Voiculescu VM, Soare C, Ceafalan LC, Gherghiceanu M, Hinescu ME. Skin Telocytes Could Fundament the Cellular Mechanisms of Wound Healing in Platelet-Rich Plasma Administration. Cells 2024; 13:1321. [PMID: 39195210 DOI: 10.3390/cells13161321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
For more than 40 years, autologous platelet concentrates have been used in clinical medicine. Since the first formula used, namely platelet-rich plasma (PRP), other platelet concentrates have been experimented with, including platelet-rich fibrin and concentrated growth factor. Platelet concentrates have three standard characteristics: they act as scaffolds, they serve as a source of growth factors and cytokines, and they contain live cells. PRP has become extensively used in regenerative medicine for the successful treatment of a variety of clinical (non-)dermatological conditions like alopecies, acne scars, skin burns, skin ulcers, muscle, cartilage, and bone repair, and as an adjuvant in post-surgery wound healing, with obvious benefits in terms of functionality and aesthetic recovery of affected tissues/organs. These indications were well documented, and a large amount of evidence has already been published supporting the efficacy of this method. The primordial principle behind minimally invasive PRP treatments is the usage of the patient's own platelets. The benefits of the autologous transplantation of thrombocytes are significant, representing a fast and economic method that requires only basic equipment and training, and it is biocompatible, thus being a low risk for the patient (infection and immunological reactions can be virtually disregarded). Usually, the structural benefits of applying PRP are attributed to fibroblasts only, as they are considered the most numerous cell population within the interstitium. However, this apparent simplistic explanation is still eluding those different types of interstitial cells (distinct from fibroblasts) that are residing within stromal tissue, e.g., telocytes (TCs). Moreover, dermal TCs have an already documented potential in angiogenesis (extra-cutaneous, but also within skin), and their implication in skin recovery in a few dermatological conditions was attested and described ultrastructurally and immunophenotypically. Interestingly, PRP biochemically consists of a series of growth factors, cytokines, and other molecules, to which TCs have also proven to have a positive expression. Thus, it is attractive to hypothesize and to document any tissular collaboration between cutaneous administered PRP and local dermal TCs in skin recovery/repair/regeneration. Therefore, TCs could be perceived as the missing link necessary to provide a solid explanation of the good results achieved by administering PRP in skin-repairing processes.
Collapse
Affiliation(s)
- Catalin G Manole
- Department of Cellular and Molecular Biology and Histology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ultrastructural Pathology Laboratory, "Victor Babeș" National Institute of Pathology, 050096 Bucharest, Romania
| | - Vlad M Voiculescu
- Department of Oncological Dermatology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cristina Soare
- Department of Oncological Dermatology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Department of Cellular and Molecular Biology and Histology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Cell Biology, Neurosciences and Experimental Myology Laboratory, "Victor Babeș" National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihaela Gherghiceanu
- Department of Cellular and Molecular Biology and Histology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Ultrastructural Pathology Laboratory, "Victor Babeș" National Institute of Pathology, 050096 Bucharest, Romania
| | - Mihail E Hinescu
- Department of Cellular and Molecular Biology and Histology, "Carol Davila" University of Medicine and Pharmacy, 020021 Bucharest, Romania
- "Victor Babeș" National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
7
|
Barrera K, Rodellar J, Alférez S, Merino A. A deep learning approach for automatic recognition of abnormalities in the cytoplasm of neutrophils. Comput Biol Med 2024; 178:108691. [PMID: 38905894 DOI: 10.1016/j.compbiomed.2024.108691] [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: 04/03/2024] [Revised: 05/12/2024] [Accepted: 06/01/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND AND OBJECTIVES This study aims to develop and evaluate NeuNN, a system based on convolutional neural networks (CNN) and generative adversarial networks (GAN) for the automatic identification of normal neutrophils and those containing several types of inclusions or showing hypogranulation. METHODS From peripheral blood smears, a set of 5605 digital images was obtained with neutrophils belonging to seven categories: Normal neutrophils (NEU), Hypogranulated (HYP) or containing cryoglobulins (CRY), Döhle bodies (DB), Howell-Jolly body-like inclusions (HJBLI), Green-blue inclusions of death (GBI) and phagocytosed bacteria (BAC). The dataset utilized in this study has been made publicly available. The class of GBI was augmented using synthetic images generated by GAN. The NeuNN classification model is based on an EfficientNet-B7 architecture trained from scratch. RESULTS NeuNN achieved an overall performance of 94.3% accuracy on the test data set. Performance metrics, including sensitivity, specificity, precision, F1-Score, Jaccard index, and Matthews correlation coefficient indicated overall values of 94%, 99.1%, 94.3%, 94.3%, 89.6%, and 93.6%, respectively. CONCLUSIONS The proposed approach, combining data augmentation and classification techniques, allows for automated identification of morphological findings in neutrophils, such us inclusions or hypogranulation. The system can be used as a support tool for clinical pathologists to detect these specific abnormalities with clinical relevance.
Collapse
Affiliation(s)
- Kevin Barrera
- Technical University of Catalonia, Barcelona East Engineering School, Department of Mathematics, Barcelona, Spain.
| | - José Rodellar
- Technical University of Catalonia, Barcelona East Engineering School, Department of Mathematics, Barcelona, Spain.
| | - Santiago Alférez
- Technical University of Catalonia, Barcelona East Engineering School, Department of Mathematics, Barcelona, Spain.
| | - Anna Merino
- Hospital Clínic of Barcelona-IDIBAPS, Biochemistry and Molecular Genetics Department, CORE Laboratory, Biomedical Diagnostic, Barcelona, Spain.
| |
Collapse
|
8
|
Ranson T, Rourick H, Sooch R, Ford N, Beyersdorfer N, Johnson K, Paulson J. An Investigation of Mortality Associated With Comorbid Pneumonia and Thrombocytopenia in a Rural Southwest Missouri Hospital System. Cureus 2024; 16:e67330. [PMID: 39170646 PMCID: PMC11338473 DOI: 10.7759/cureus.67330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Pneumonia places a significant burden on individuals and society, contributing to a substantial number of hospital admissions, emergency department visits, deaths, and healthcare costs each year. Comorbidities can greatly increase the risk of poor outcomes when associated with pneumonia. One comorbidity that has yet to be thoroughly researched is thrombocytopenia, which is known to play an important role in activating the immune response to infections. A decrease in platelet count may limit the immune response and consequently increase mortality in patients with pneumonia. The purpose of this study was to investigate whether comorbid thrombocytopenia and pneumonia are associated with poor outcomes. METHODS This study was a retrospective cohort analysis comparing mortality rates among patients with comorbid thrombocytopenia and pneumonia, pneumonia without thrombocytopenia, and thrombocytopenia without pneumonia. Data were collected from Freeman Health System using International Classification of Diseases, Tenth Revision (ICD-10) codes from January 1, 2019, to December 31, 2021. ICD-10 codes for pneumonia and thrombocytopenia were extracted and stratified into three groups: those with both pneumonia and thrombocytopenia, those with pneumonia without thrombocytopenia, and those with thrombocytopenia without pneumonia. Mortality rates were then compared across the three groups. RESULTS There were 4,414 patients admitted with pneumonia and 1,157 admissions for thrombocytopenia without pneumonia. Among the 4,414 patients admitted with pneumonia, 3,902 did not have thrombocytopenia, while 512 had thrombocytopenia. Of the patients without thrombocytopenia, 14% (3,902) expired. Among the 512 patients with thrombocytopenia, 43% expired. In the thrombocytopenia without pneumonia group, 11% (1,157) expired. CONCLUSION These results indicate a significant increase in mortality in patients with both pneumonia and thrombocytopenia compared to those with pneumonia without thrombocytopenia (an increase in mortality of 28.93% with a 95% CI: 24.50-33.36%, P < 0.0001). While pneumonia itself increases mortality compared to the general population, patients with both pneumonia and thrombocytopenia exhibit even higher mortality rates.
Collapse
Affiliation(s)
- Tabitha Ranson
- College of Medicine, Kansas City University, Joplin, USA
| | - Hannah Rourick
- College of Medicine, Kansas City University, Joplin, USA
| | - Rajbir Sooch
- College of Medicine, Kansas City University, Joplin, USA
| | - Nicole Ford
- College of Medicine, Kansas City University, Joplin, USA
| | - Nova Beyersdorfer
- Primary Care, College of Medicine, Kansas City University, Joplin, USA
| | - Kerry Johnson
- Mathematics, Missouri Southern State University, Joplin, USA
| | - John Paulson
- College of Medicine, Kansas City University, Joplin, USA
| |
Collapse
|
9
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [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: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
10
|
Ledderose C, Valsami EA, Elevado M, Junger WG. Adenosine Triphosphate Release From Influenza-Infected Lungs Enhances Neutrophil Activation and Promotes Disease Progression. J Infect Dis 2024; 230:120-130. [PMID: 39052721 PMCID: PMC11272046 DOI: 10.1093/infdis/jiad442] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 10/08/2023] Open
Abstract
BACKGROUND Adenosine triphosphate (ATP) enhances neutrophil responses, but little is known about the role of ATP in influenza infections. METHODS We used a mouse influenza model to study if ATP release is associated with neutrophil activation and disease progression. RESULTS Influenza infection increased pulmonary ATP levels 5-fold and plasma ATP levels 3-fold vs healthy mice. Adding ATP at those concentrations to blood from healthy mice primed neutrophils and enhanced CD11b and CD63 expression, CD62L shedding, and reactive oxygen species production in response to formyl peptide receptor stimulation. Influenza infection also primed neutrophils in vivo, resulting in formyl peptide receptor-induced CD11b expression and CD62L shedding up to 3 times higher than that of uninfected mice. In infected mice, large numbers of neutrophils entered the lungs. These cells were significantly more activated than the peripheral neutrophils of infected mice and pulmonary neutrophils of healthy mice. Plasma ATP levels of infected mice and influenza disease progression corresponded with the numbers and activation level of their pulmonary neutrophils. CONCLUSIONS Findings suggest that ATP release from the lungs of infected mice promotes influenza disease progression by priming peripheral neutrophils, which become strongly activated and cause pulmonary tissue damage after their recruitment to the lungs.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, University of California, San Diego Health
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | | | - Mark Elevado
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Wolfgang G Junger
- Department of Surgery, University of California, San Diego Health
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
11
|
Ho MY, Liu S, Xing B. Bacteria extracellular vesicle as nanopharmaceuticals for versatile biomedical potential. NANO CONVERGENCE 2024; 11:28. [PMID: 38990415 PMCID: PMC11239649 DOI: 10.1186/s40580-024-00434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/20/2024] [Indexed: 07/12/2024]
Abstract
Bacteria extracellular vesicles (BEVs), characterized as the lipid bilayer membrane-surrounded nanoparticles filled with molecular cargo from parent cells, play fundamental roles in the bacteria growth and pathogenesis, as well as facilitating essential interaction between bacteria and host systems. Notably, benefiting from their unique biological functions, BEVs hold great promise as novel nanopharmaceuticals for diverse biomedical potential, attracting significant interest from both industry and academia. Typically, BEVs are evaluated as promising drug delivery platforms, on account of their intrinsic cell-targeting capability, ease of versatile cargo engineering, and capability to penetrate physiological barriers. Moreover, attributing to considerable intrinsic immunogenicity, BEVs are able to interact with the host immune system to boost immunotherapy as the novel nanovaccine against a wide range of diseases. Towards these significant directions, in this review, we elucidate the nature of BEVs and their role in activating host immune response for a better understanding of BEV-based nanopharmaceuticals' development. Additionally, we also systematically summarize recent advances in BEVs for achieving the target delivery of genetic material, therapeutic agents, and functional materials. Furthermore, vaccination strategies using BEVs are carefully covered, illustrating their flexible therapeutic potential in combating bacterial infections, viral infections, and cancer. Finally, the current hurdles and further outlook of these BEV-based nanopharmaceuticals will also be provided.
Collapse
Affiliation(s)
- Ming Yao Ho
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore
| | - Songhan Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore
| | - Bengang Xing
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore, S637371, Singapore.
| |
Collapse
|
12
|
Ledderose C, Valsami EA, Elevado M, Liu Q, Giva B, Curatolo J, Delfin J, Abutabikh R, Junger WG. Impaired ATP hydrolysis in blood plasma contributes to age-related neutrophil dysfunction. Immun Ageing 2024; 21:45. [PMID: 38961477 PMCID: PMC11221114 DOI: 10.1186/s12979-024-00441-4] [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: 02/29/2024] [Accepted: 05/29/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND The function of polymorphonuclear neutrophils (PMNs) decreases with age, which results in infectious and inflammatory complications in older individuals. The underlying causes are not fully understood. ATP release and autocrine stimulation of purinergic receptors help PMNs combat microbial invaders. Excessive extracellular ATP interferes with these mechanisms and promotes inflammatory PMN responses. Here, we studied whether dysregulated purinergic signaling in PMNs contributes to their dysfunction in older individuals. RESULTS Bacterial infection of C57BL/6 mice resulted in exaggerated PMN activation that was significantly greater in old mice (64 weeks) than in young animals (10 weeks). In contrast to young animals, old mice were unable to prevent the systemic spread of bacteria, resulting in lethal sepsis and significantly greater mortality in old mice than in their younger counterparts. We found that the ATP levels in the plasma of mice increased with age and that, along with the extracellular accumulation of ATP, the PMNs of old mice became increasingly primed. Stimulation of the formyl peptide receptors of those primed PMNs triggered inflammatory responses that were significantly more pronounced in old mice than in young animals. However, bacterial phagocytosis and killing by PMNs of old mice were significantly lower than that of young mice. These age-dependent PMN dysfunctions correlated with a decrease in the enzymatic activity of plasma ATPases that convert extracellular ATP to adenosine. ATPases depend on divalent metal ions, including Ca2+, Mg2+, and Zn2+, and we found that depletion of these ions blocked the hydrolysis of ATP and the formation of adenosine in human blood, resulting in ATP accumulation and dysregulation of PMN functions equivalent to those observed in response to aging. CONCLUSIONS Our findings suggest that impaired hydrolysis of plasma ATP dysregulates PMN function in older individuals. We conclude that strategies aimed at restoring plasma ATPase activity may offer novel therapeutic opportunities to reduce immune dysfunction, inflammation, and infectious complications in older patients.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Mark Elevado
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Qing Liu
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Brennan Giva
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Julian Curatolo
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Joshua Delfin
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Reem Abutabikh
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA
| | - Wolfgang G Junger
- Department of Surgery, University of California, San Diego Health, 9452 Medical Ctr Dr, La Jolla, San Diego, CA, 92037, USA.
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Jandus C, Jandus P. Effects of Intravenous Immunoglobulins on Human Innate Immune Cells: Collegium Internationale Allergologicum Update 2024. Int Arch Allergy Immunol 2024; 185:975-996. [PMID: 38852585 DOI: 10.1159/000539069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/22/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Intravenous immunoglobulin (IVIg) has been used for almost 40 years in the treatment of autoimmune and systemic inflammatory diseases. Numerous cells are involved in the innate immune response, including monocytes/macrophages, neutrophils, dendritic cells, mast cells, basophils, eosinophils, natural killer cells, and innate lymphoid cells. Many studies have investigated the mechanisms by which IVIg down-modulates inflammatory and autoimmune processes of innate immune cells. However, questions remain regarding the precise mechanism of action in autoimmune or inflammatory conditions. The aim of this work was to review the immunomodulatory effect of IVIg on only human innate immune cells. A narrative review approach was chosen to summarize key evidence on the immunomodulatory effects of commercially available and unmodified IVIg on human innate immune cells. SUMMARY Numerous different immunomodulatory effects of IVIg have been reported, with some very different effects depending on the immune cell type and disease. Several limitations of the different studies were identified. Of the 77 studies identified and reviewed, 29 (37.7%) dealt with autoimmune or inflammatory diseases. Otherwise, the immunomodulatory effects of IVIg were studied only in healthy donors using an in vitro experimental approach. Some of the documented effects showed disease-specific effects, such as in Kawasaki disease. Various methodological limitations have also been identified that may reduce the validity of some studies. KEY MESSAGE As further insights have been gained into the various inflammatory cascades activated in immunological diseases, interesting insights have also been gained into the mechanism of action of IVIg. We are still far from discovering all the immunomodulatory mechanisms of IVIg.
Collapse
Affiliation(s)
- Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Centre in Onco-Haematology (CRTOH), Geneva, Switzerland
| | - Peter Jandus
- Division of Immunology and Allergology, University Hospitals and Medical Faculty, Geneva, Switzerland
| |
Collapse
|
14
|
Stream A, Corriden R, Döhrmann S, Gallo RL, Nizet V, Anderson EL. The Effect of Retinoic Acid on Neutrophil Innate Immune Interactions With Cutaneous Bacterial Pathogens. INFECTIOUS MICROBES & DISEASES 2024; 6:65-73. [PMID: 38952747 PMCID: PMC11216695 DOI: 10.1097/im9.0000000000000145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Vitamin A and its biologically active derivative, retinoic acid (RA), are important for many immune processes. RA, in particular, is essential for the development of immune cells, including neutrophils, which serve as a front-line defense against infection. While vitamin A deficiency has been linked to higher susceptibility to infections, the precise role of vitamin A/RA in host-pathogen interactions remains poorly understood. Here, we provided evidence that RA boosts neutrophil killing of methicillin-resistant Staphylococcus aureus (MRSA). RA treatment stimulated primary human neutrophils to produce reactive oxygen species, neutrophil extracellular traps, and the antimicrobial peptide cathelicidin (LL-37). Because RA treatment was insufficient to reduce MRSA burden in an in vivo murine model of skin infection, we expanded our analysis to other infectious agents. RA did not affect the growth of a number of common bacterial pathogens, including MRSA, Escherichia coli K1 and Pseudomonas aeruginosa; however, RA directly inhibited the growth of group A Streptococcus (GAS). This antimicrobial effect, likely in combination with RA-mediated neutrophil boosting, resulted in substantial GAS killing in neutrophil killing assays conducted in the presence of RA. Furthermore, in a murine model of GAS skin infection, topical RA treatment showed therapeutic potential by reducing both skin lesion size and bacterial burden. These findings suggest that RA may hold promise as a therapeutic agent against GAS and perhaps other clinically significant human pathogens.
Collapse
Affiliation(s)
- Alexandra Stream
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Ross Corriden
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Simon Döhrmann
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Richard L. Gallo
- Department of Dermatology, University of California San Diego School of Medicine, La Jolla, California, USA
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 3, USA
| | - Ericka L. Anderson
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California, USA
| |
Collapse
|
15
|
Viola H, Chen LH, Jo S, Washington K, Selva C, Li A, Feng D, Giacalone V, Stephenson ST, Cottrill K, Mohammed A, Williams E, Qu X, Lam W, Ng NL, Fitzpatrick A, Grunwell J, Tirouvanziam R, Takayama S. HIGH THROUGHPUT QUANTITATION OF HUMAN NEUTROPHIL RECRUITMENT AND FUNCTIONAL RESPONSES IN AN AIR-BLOOD BARRIER ARRAY. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593624. [PMID: 38798413 PMCID: PMC11118313 DOI: 10.1101/2024.05.10.593624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Dysregulated neutrophil recruitment drives many pulmonary diseases, but most preclinical screening methods are unsuited to evaluate pulmonary neutrophilia, limiting progress towards therapeutics. Namely, high throughput therapeutic screening systems typically exclude critical neutrophilic pathophysiology, including blood-to-lung recruitment, dysfunctional activation, and resulting impacts on the air-blood barrier. To meet the conflicting demands of physiological complexity and high throughput, we developed an assay of 96-well Leukocyte recruitment in an Air-Blood Barrier Array (L-ABBA-96) that enables in vivo -like neutrophil recruitment compatible with downstream phenotyping by automated flow cytometry. We modeled acute respiratory distress syndrome (ARDS) with neutrophil recruitment to 20 ng/mL epithelial-side interleukin 8 (IL-8) and found a dose dependent reduction in recruitment with physiologic doses of baricitinib, a JAK1/2 inhibitor recently FDA-approved for severe COVID-19 ARDS. Additionally, neutrophil recruitment to patient-derived cystic fibrosis sputum supernatant induced disease-mimetic recruitment and activation of healthy donor neutrophils and upregulated endothelial e-selectin. Compared to 24-well assays, the L-ABBA-96 reduces required patient sample volumes by 25 times per well and quadruples throughput per plate. Compared to microfluidic assays, the L-ABBA-96 recruits two orders of magnitude more neutrophils per well, enabling downstream flow cytometry and other standard biochemical assays. This novel pairing of high-throughput in vitro modeling of organ-level lung function with parallel high-throughput leukocyte phenotyping substantially advances opportunities for pathophysiological studies, personalized medicine, and drug testing applications.
Collapse
|
16
|
Choi UY, Choi YJ, Lee SA, Yoo JS. Cisd2 deficiency impairs neutrophil function by regulating calcium homeostasis via Calnexin and SERCA. BMB Rep 2024; 57:256-261. [PMID: 38627949 PMCID: PMC11139677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/16/2024] [Accepted: 04/05/2024] [Indexed: 05/25/2024] Open
Abstract
In the context of aging, the susceptibility to infectious diseases increases, leading to heightened morbidity and mortality. This phenomenon, termed immunosenescence, is characterized by dysregulation in the aging immune system, including abnormal alterations in lymphocyte composition, elevated basal inflammation, and the accumulation of senescent T cells. Such changes contribute to increased autoimmune diseases, enhanced infection severity, and reduced responsiveness to vaccines. Utilizing aging animal models becomes imperative for a comprehensive understanding of immunosenescence, given the complexity of aging as a physiological process in living organisms. Our investigation focuses on Cisd2, a causative gene for Wolfram syndrome, to elucidate on immunosenescence. Cisd2 knockout (KO) mice, serving as a model for premature aging, exhibit a shortened lifespan with early onset of aging-related features, such as decreased bone density, hair loss, depigmentation, and optic nerve degeneration. Intriguingly, we found that the Cisd2 KO mice present a higher number of neutrophils in the blood; however, isolated neutrophils from these mice display functional defects. Through mass spectrometry analysis, we identified an interaction between Cisd2 and Calnexin, a protein known for its role in protein quality control. Beyond this function, Calnexin also regulates calcium homeostasis through interaction with sarcoendoplasmic reticulum calcium transport ATPase (SERCA). Our study proposes that Cisd2 modulates calcium homeostasis via its interaction with Calnexin and SERCA, consequently influencing neutrophil functions. [BMB Reports 2024; 57(5): 256-261].
Collapse
Affiliation(s)
- Un Yung Choi
- Department of Microbiology, Konkuk University School of Medicine, Chungju 07478, Korea
- KU Open Innovation Center, Research Institute of Medical Science, Konkuk University School of Medicine, Chungju 07478, Korea, Daegu 41566, Korea
| | - Youn Jung Choi
- Kao Autoimmunity Institute and Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, Korea
| | - Shin-Ae Lee
- Department of Cancer Biology, Infection Biology Program, Global Center for Pathogen and Human Health Research, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA, Daegu 41566, Korea
| | - Ji-Seung Yoo
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
17
|
Guo H, Guo L, Li L, Li N, Lin X, Wang Y. Identification of key genes and molecular mechanisms of chronic urticaria based on bioinformatics. Skin Res Technol 2024; 30:e13624. [PMID: 38558219 PMCID: PMC10982677 DOI: 10.1111/srt.13624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 04/04/2024]
Abstract
Chronic urticaria (CU) is characterized by persistent skin hives, redness, and itching, enhanced by immune dysregulation and inflammation. Our main objective is identifying key genes and molecular mechanisms of chronic urticaria based on bioinformatics. We used the Gene Expression Omnibus (GEO) database and retrieved two GEO datasets, GSE57178 and GSE72540. The raw data were extracted, pre-processed, and analyzed using the GEO2R tool to identify the differentially expressed genes (DEGs). The samples were divided into two groups: healthy samples and CU samples. We defined cut-off values of log2 fold change ≥1 and p < .05. Analyses were performed in the Kyoto Encyclopaedia of Genes and Genomes (KEGG), the Database for Annotation, Visualization and Integrated Discovery (DAVID), Metascape, Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and CIBERSOFT databases. We obtained 1613 differentially expressed genes. There were 114 overlapping genes in both datasets, out of which 102 genes were up-regulated while 12 were down-regulated. The biological processes included activation of myeloid leukocytes, response to inflammations, and response to organic substances. Moreover, the KEGG pathways of CU were enriched in the Nuclear Factor-Kappa B (NF-kB) signaling pathway, Tumor Necrosis Factor (TNF) signaling pathway, and Janus kinase/signal transducers and activators of transcription (JAK-STAT) signaling pathway. We identified 27 hub genes that were implicated in the pathogenesis of CU, such as interleukin-6 (IL-6), Prostaglandin-endoperoxide synthase 2 (PTGS2), and intercellular adhesion molecule-1 (ICAM1). The complex interplay between immune responses, inflammatory pathways, cytokine networks, and specific genes enhances CU. Understanding these mechanisms paves the way for potential interventions to mitigate symptoms and improve the quality of life of CU patients.
Collapse
Affiliation(s)
- Haichao Guo
- Department of Acupuncture and MoxibustionThe First Affiliated Hospital of Hebei University of Chinese MedicineShijiazhuangHebeiChina
- Department of DermatologyXingtai Hospital of Traditional Chinese MedicineXingtaiHebeiChina
| | - Lifang Guo
- Department of DermatologyXingtai Hospital of Traditional Chinese MedicineXingtaiHebeiChina
| | - Li Li
- Department of DermatologyXingtai Hospital of Traditional Chinese MedicineXingtaiHebeiChina
| | - Na Li
- Department of PsychiatryThe First Affiliated Hospital of Hebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Xiaoyun Lin
- Department of Acupuncture and MoxibustionThe First Affiliated Hospital of Hebei University of Chinese MedicineShijiazhuangHebeiChina
| | - Yanjun Wang
- Department of Acupuncture and MoxibustionThe First Affiliated Hospital of Hebei University of Chinese MedicineShijiazhuangHebeiChina
| |
Collapse
|
18
|
Zaid A, Ariel A. Harnessing anti-inflammatory pathways and macrophage nano delivery to treat inflammatory and fibrotic disorders. Adv Drug Deliv Rev 2024; 207:115204. [PMID: 38342241 DOI: 10.1016/j.addr.2024.115204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Targeting specific organs and cell types using nanotechnology and sophisticated delivery methods has been at the forefront of applicative biomedical sciences lately. Macrophages are an appealing target for immunomodulation by nanodelivery as they are heavily involved in various aspects of many diseases and are highly plastic in their nature. Their continuum of functional "polarization" states has been a research focus for many years yielding a profound understanding of various aspects of these cells. The ability of monocyte-derived macrophages to metamorphose from pro-inflammatory to reparative and consequently to pro-resolving effectors has raised significant interest in its therapeutic potential. Here, we briefly survey macrophages' ontogeny and various polarization phenotypes, highlighting their function in the inflammation-resolution shift. We review their inducing mediators, signaling pathways, and biological programs with emphasis on the nucleic acid sensing-IFN-I axis. We also portray the polarization spectrum of macrophages and the characteristics of their transition between different subtypes. Finally, we highlighted different current drug delivery methods for targeting macrophages with emphasis on nanotargeting that might lead to breakthroughs in the treatment of wound healing, bone regeneration, autoimmune, and fibrotic diseases.
Collapse
Affiliation(s)
- Ahmad Zaid
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel
| | - Amiram Ariel
- Department of Biology and Human Biology, University of Haifa, Haifa, 3498838 Israel.
| |
Collapse
|
19
|
Bharatha M, Nandana MB, Praveen R, Nayaka S, Velmurugan D, Vishwanath BS, Rajaiah R. Unconjugated bilirubin and its derivative ameliorate IMQ-induced psoriasis-like skin inflammation in mice by inhibiting MMP9 and MAPK pathway. Int Immunopharmacol 2024; 130:111679. [PMID: 38377853 DOI: 10.1016/j.intimp.2024.111679] [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: 12/16/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/22/2024]
Abstract
Psoriasis is a chronic immune-mediated inflammatory skin disease that involves dysregulated proliferation of keratinocytes. Psoriatic skin lesions are characterized by redness, thickness, and scaling. The interleukin axis of IL-23/IL-17 is critically involved in the development of human psoriasis. Imiquimod (IMQ), an agonist of TLR7 is known to induce psoriatic-like skin inflammation in mice. The topical application of IMQ induces systemic inflammation with increased proinflammatory cytokines in serum and secondary lymphoid organs. Further, matrix metalloproteases (MMPs) have been implicated in the pathophysiology of psoriatic-like skin inflammation. The increased MMP9 activity and gene expression of proinflammatory cytokines in IMQ-induced psoriatic skin is mediated by the activation of the MAPK pathway. Moreover, the increased expression of neutrophil-specific chemokines confirmed the infiltration of neutrophils at the site of psoriatic skin inflammation. In contrast, expression of IL-10, an anti-inflammatory cytokine gene expression is reduced in IMQ-treated mice skin. Topical application of unconjugated bilirubin (UCB) and its derivative dimethyl ester of bilirubin (BD1) on IMQ-induced psoriatic mice skin significantly mitigated the symptoms of psoriasis by inhibiting the activity of MMP9. Further, UCB and BD1 reduced neutrophil infiltration as evidenced by decreased myeloperoxidase (MPO) activity and reduced gene expression of proinflammatory cytokines, and neutrophil-specific chemokines. Apart from these modulations UCB and BD1 reduced MAPK phosphorylation and upregulated anti-inflammatory cytokines. To conclude, UCB and BD1 immunomodulated the psoriatic skin inflammation induced by IMQ in mice by inhibiting neutrophil mediated MMP9, decreased proinflammatory cytokines gene expression and modulating the MAPK pathway.
Collapse
Affiliation(s)
- Madeva Bharatha
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India
| | - Manuganahalli B Nandana
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India
| | - Raju Praveen
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India
| | - Spandan Nayaka
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India
| | - Devadasan Velmurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, India
| | - Bannikuppe S Vishwanath
- Department of Studies in Biochemistry, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India.
| | - Rajesh Rajaiah
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India.
| |
Collapse
|
20
|
Jatczak-Pawlik I, Ewiak-Paszyńska A, Domowicz M, Jurewicz A, Stasiołek M. Intracellular Accumulation and Secretion of YKL-40 (CHI3L1) in the Course of DMSO-Induced HL-60 Cell Differentiation. Pharmaceuticals (Basel) 2024; 17:443. [PMID: 38675403 PMCID: PMC11053806 DOI: 10.3390/ph17040443] [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: 01/19/2024] [Revised: 03/04/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
YKL-40 (CHI3L1) is a matrix glycoprotein stored in human neutrophil-specific granules and released upon activation. While it is implicated in inflammation, cancer progression, and cell differentiation, its exact physiological role remains unclear. This study investigated the intracellular expression and secretion of YKL-40 by untreated and DMSO-treated HL-60 cells in association with surface expression of CD11b and CD66b throughout the differentiation process (up to 120 h). Secreted YKL-40 protein and mRNA levels of YKL-40, CD66b, and CD11b were measured by ELISA and quantitative RT-PCR, respectively. The intracellular YKL-40 and surface CD11b and CD66b expression were assessed by flow cytometry. A significant increase in CD11b expression confirmed DMSO-induced differentiation of HL-60 cells. Upon DMSO stimulation, YKL-40 mRNA expression increased in a time-dependent manner, unlike CD66b. The lack of CD66b (a granulocyte maturation and activation marker) on the surface of HL-60 cells might suggest that DMSO treatment did not induce full maturation or activation. The intracellular YKL-40 protein expression was increasing up to 96 h of DMSO treatment and then declined. YKL-40 secretion into the culture medium was detectable only at later time points (96 and 120 h), which was correlated with a decreased proliferation of DMSO-treated HL-60 cells. These findings suggest sequential changes in YKL-40 production and secretion during DMSO-induced differentiation of HL-60 cells and might contribute to a better understanding of YKL-40's involvement in both physiological processes and disease development, including multiple sclerosis.
Collapse
Affiliation(s)
| | | | | | | | - Mariusz Stasiołek
- Department of Neurology, Medical University of Lodz, Kosciuszki Street 4, 90-419 Lodz, Poland; (I.J.-P.); (A.E.-P.); (M.D.); (A.J.)
| |
Collapse
|
21
|
Schoberleitner I, Faserl K, Tripp CH, Pechriggl EJ, Sigl S, Brunner A, Zelger B, Hermann-Kleiter N, Baier L, Steinkellner T, Sarg B, Egle D, Brunner C, Wolfram D. Silicone implant surface microtopography modulates inflammation and tissue repair in capsular fibrosis. Front Immunol 2024; 15:1342895. [PMID: 38566997 PMCID: PMC10985323 DOI: 10.3389/fimmu.2024.1342895] [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: 11/28/2023] [Accepted: 02/29/2024] [Indexed: 04/04/2024] Open
Abstract
Excessive fibrous capsule formation around silicone mammary implants (SMI) involves immune reactions to silicone. Capsular fibrosis, a common SMI complication linked to host responses, worsens with specific implant topographies. Our study with 10 patients investigated intra- and inter-individually, reduced surface roughness effects on disease progression, wound responses, chronic inflammation, and capsular composition. The results illuminate the significant impact of surface roughness on acute inflammatory responses, fibrinogen accumulation, and the subsequent fibrotic cascade. The reduction of surface roughness to an average roughness of 4 μm emerges as a promising approach for mitigating detrimental immune reactions, promoting healthy wound healing, and curbing excessive fibrosis. The identified proteins adhering to rougher surfaces shed light on potential mediators of pro-inflammatory and pro-fibrotic processes, further emphasizing the need for meticulous consideration of surface design. The composition of the implant capsule and the discovery of intracapsular HSP60 expression highlight the intricate web of stress responses and immune activation that can impact long-term tissue outcomes.
Collapse
Affiliation(s)
- Ines Schoberleitner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Faserl
- Protein Core Facility, Institute of Medical Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph H. Tripp
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elisabeth Judith Pechriggl
- Department of Anatomy, Histology and Embryology, Institute of Clinical and Functional Anatomy, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Andrea Brunner
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
- INNPATH GmbH, Tirol Kliniken, Innsbruck, Austria
| | - Bettina Zelger
- Institute of Pathology, Neuropathology and Molecular Pathology, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Institute of Cell Genetics, Department for Genetics and Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Leoni Baier
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Theresia Steinkellner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Bettina Sarg
- Protein Core Facility, Institute of Medical Chemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Egle
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christine Brunner
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Innsbruck, Austria
| | - Dolores Wolfram
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Martin KR, Gamell C, Tai TY, Bonelli R, Hansen J, Tatoulis J, Alhamdoosh M, Wilson N, Wicks I. Whole blood transcriptomics reveals granulocyte colony-stimulating factor as a mediator of cardiopulmonary bypass-induced systemic inflammatory response syndrome. Clin Transl Immunology 2024; 13:e1490. [PMID: 38375330 PMCID: PMC10875393 DOI: 10.1002/cti2.1490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 12/20/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Objectives Systemic inflammatory response syndrome (SIRS) is a frequent complication of cardiopulmonary bypass (CPB). SIRS is associated with significant morbidity and mortality, but its pathogenesis remains incompletely understood, and as a result, biomarkers are lacking and treatment remains expectant and supportive. This study aimed to understand the pathophysiological mechanisms driving SIRS induced by CPB and identify novel therapeutic targets that might reduce systemic inflammation and improve patient outcomes. Methods Twenty-one patients undergoing cardiac surgery and CPB were recruited, and blood was sampled before, during and after surgery. SIRS was defined using the American College of Chest Physicians/Society of Critical Care Medicine criteria. We performed immune cell profiling and whole blood transcriptomics and measured individual mediators in plasma/serum to characterise SIRS induced by CPB. Results Nineteen patients fulfilled criteria for SIRS, with a mean duration of 2.7 days. Neutrophil numbers rose rapidly with CPB and remained elevated for at least 48 h afterwards. Transcriptional signatures associated with neutrophil activation and degranulation were enriched during CPB. We identified a network of cytokines governing these transcriptional changes, including granulocyte colony-stimulating factor (G-CSF), a regulator of neutrophil production and function. Conclusions We identified neutrophils and G-CSF as major regulators of CPB-induced systemic inflammation. Short-term targeting of G-CSF could provide a novel therapeutic strategy to limit neutrophil-mediated inflammation and tissue damage in SIRS induced by CPB.
Collapse
Affiliation(s)
- Katherine R Martin
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
| | | | - Tsin Yee Tai
- WEHIParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | - Roberto Bonelli
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- CSL Innovation, Bio21 InstituteParkvilleVICAustralia
| | | | - James Tatoulis
- Cardiothoracic SurgeryRoyal Melbourne HospitalParkvilleVICAustralia
- Department of SurgeryUniversity of MelbourneParkvilleVICAustralia
| | | | | | - Ian Wicks
- WEHIParkvilleVICAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVICAustralia
- Department of RheumatologyRoyal Melbourne HospitalParkvilleVICAustralia
| |
Collapse
|
23
|
Ouyang J, Hong Y, Wan Y, He X, Geng B, Yang X, Xiang J, Cai J, Zeng Z, Liu Z, Peng N, Jiang Y, Liu J. PVB exerts anti-inflammatory effects by inhibiting the activation of MAPK and NF-κB signaling pathways and ROS generation in neutrophils. Int Immunopharmacol 2024; 126:111271. [PMID: 38006749 DOI: 10.1016/j.intimp.2023.111271] [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: 09/12/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 11/27/2023]
Abstract
Pinaverium bromide (PVB) has been shown to protect mice against sepsis, which is predominantly attributed to PVB-mediated anti-inflammatory effects by inhibiting primed neutrophils to produce proinflammatory cytokines. However, the underlying mechanism(s) by which PVB affects neutrophils remains unknown. In this study, we report that treatment with PVB either before or after LPS stimulation attenuated IL-1β and TNF-α expression at both mRNA and protein levels in LPS-activated murine neutrophils. Further experiments revealed that PVB inhibited the phosphorylation of ERK, JNK, and IκBα in LPS-stimulated murine neutrophils. Moreover, PVB reduced reactive oxygen species (ROS) levels via regulating NADPH oxidase 2 (NOX2) activity, as represented by inhibiting p47phox translocation from the cytoplasm to the cellular membrane. Importantly, PVB significantly attenuated IL-1β, TNF-α, IL-6, CXCL1 production in both LPS-stimulated low density neutrophils (LDNs) and normal density neutrophils (NDNs) isolated from septic patients. Collectively, we demonstrated that PVB exerts anti-inflammatory effect by attenuating ROS generation and suppressing the activation of MAPK and NF-κB signaling pathways, suggesting that PVB may act as a potential therapeutic agent for sepsis by inhibiting neutrophil priming and activation.
Collapse
Affiliation(s)
- Jiafu Ouyang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yinghao Hong
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangyi He
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Bingxuan Geng
- School of the First Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinxing Yang
- School of the First Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Xiang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Junwei Cai
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhifeng Liu
- Department of Critical Care Medicine, General Hospital of Southern Theater Command, Guangzhou, Guangdong, China
| | - Na Peng
- Department of Emergency Medicine, General Hospital of Southern Theater Command, Guangzhou, Guangdong, China.
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
24
|
Ayadi I, Lamrabet O, Munoz-Ruiz R, Jauslin T, Guilhen C, Cosson P. Extracellular and intracellular destruction of Pseudomonas aeruginosa by Dictyostelium discoideum phagocytes mobilize different antibacterial mechanisms. Mol Microbiol 2024; 121:69-84. [PMID: 38017607 DOI: 10.1111/mmi.15197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
Ingestion and killing of bacteria by phagocytic cells are critical processes to protect the human body from bacterial infections. In addition, some immune cells (neutrophils, NK cells) can release microbicidal molecules in the extracellular medium to eliminate non-ingested microorganism. Molecular mechanisms involved in the resulting intracellular and extracellular killing are still poorly understood. In this study, we used the amoeba Dictyostelium discoideum as a model phagocyte to investigate the mechanisms allowing intracellular and extracellular killing of Pseudomonas aeruginosa. When a D. discoideum cell establishes a close contact with a P. aeruginosa bacterium, it can either ingest it and kill it in phagosomes, or kill it extracellularly, allowing a direct side-by-side comparison of these two killing modalities. Efficient intracellular destruction of P. aeruginosa requires the presence of the Kil2 pump in the phagosomal membrane. On the contrary, extracellular lysis is independent on Kil2 but requires the expression of the superoxide-producing protein NoxA, and the extracellular release of the AplA bacteriolytic protein. These results shed new light on the molecular mechanisms allowing elimination of P. aeruginosa bacteria by phagocytic cells.
Collapse
Affiliation(s)
- Imen Ayadi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Otmane Lamrabet
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Raphael Munoz-Ruiz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Tania Jauslin
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Cyril Guilhen
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pierre Cosson
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
25
|
Fitzpatrick AM, Mohammad AF, Huang M, Stephenson ST, Patrignani J, Kamaleswaran R, Grunwell JR. Functional immunophenotyping of blood neutrophils identifies novel endotypes of viral response in preschool children with recurrent wheezing. J Allergy Clin Immunol 2023; 152:1433-1443. [PMID: 37604313 PMCID: PMC10841272 DOI: 10.1016/j.jaci.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Preschool children with recurrent wheezing are heterogeneous, with differing responses to respiratory viral infections. Although neutrophils are crucial for host defense, their function has not been studied in this population. OBJECTIVE We performed functional immunophenotyping on isolated blood neutrophils from 52 preschool children with recurrent wheezing (aeroallergen sensitization, n = 16; no sensitization, n = 36). METHODS Blood neutrophils were purified and cultured overnight with polyinosinic:polycytidylic acid [poly(I:C)] as a viral analog stimulus. Neutrophils underwent next-generation sequencing with Reactome pathway analysis and were analyzed for cytokine secretion, apoptosis, myeloperoxidase, and extracellular DNA release. CD14+ monocytes were also exposed to neutrophil culture supernatant and analyzed for markers of M1 and M2 activation. RESULTS A total of 495 genes, related largely to the innate immune system and neutrophil degranulation, were differently expressed in children with versus without aeroallergen sensitization. Functional experiments identified more neutrophil degranulation and extracellular trap formation (ie, more myeloperoxidase and extracellular DNA) and less neutrophil proinflammatory cytokine secretion in children with aeroallergen sensitization. Neutrophils also shifted CD14+ monocytes to a more anti-inflammatory (ie, M2) phenotype in sensitized children and a more proinflammatory (ie, M1) phenotype in nonsensitized children. Although both groups experienced viral exacerbations, annualized exacerbation rates prompting unscheduled health care were also higher in children without aeroallergen sensitization after enrollment. CONCLUSIONS Systemic neutrophil responses to viral infection differ by allergic phenotype and may be less effective in preschool children without allergic inflammation. Further studies of neutrophil function are needed in this population, which often has less favorable therapeutic responses to inhaled corticosteroids and other therapies directed at type 2-high inflammation.
Collapse
Affiliation(s)
- Anne M Fitzpatrick
- Department of Pediatrics, Emory University, Atlanta, Ga; Division of Pulmonary Medicine, Children's Healthcare of Atlanta, Atlanta, Ga.
| | | | - Min Huang
- Department of Biomedical Informatics, Emory University, Atlanta, Ga
| | | | | | | | - Jocelyn R Grunwell
- Department of Pediatrics, Emory University, Atlanta, Ga; Division of Critical Care Medicine, Children's Healthcare of Atlanta, Atlanta, Ga
| |
Collapse
|
26
|
Riaz B, Sohn S. Neutrophils in Inflammatory Diseases: Unraveling the Impact of Their Derived Molecules and Heterogeneity. Cells 2023; 12:2621. [PMID: 37998356 PMCID: PMC10670008 DOI: 10.3390/cells12222621] [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: 10/27/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Inflammatory diseases involve numerous disorders and medical conditions defined by an insufficient level of self-tolerance. These diseases evolve over the course of a multi-step process through which environmental variables play a crucial role in the emergence of aberrant innate and adaptive immunological responses. According to experimental data accumulated over the past decade, neutrophils play a significant role as effector cells in innate immunity. However, neutrophils are also involved in the progression of numerous diseases through participation in the onset and maintenance of immune-mediated dysregulation by releasing neutrophil-derived molecules and forming neutrophil extracellular traps, ultimately causing destruction of tissues. Additionally, neutrophils have a wide variety of functional heterogeneity with adverse effects on inflammatory diseases. However, the complicated role of neutrophil biology and its heterogeneity in inflammatory diseases remains unclear. Moreover, neutrophils are considered an intriguing target of interventional therapies due to their multifaceted role in a number of diseases. Several approaches have been developed to therapeutically target neutrophils, involving strategies to improve neutrophil function, with various compounds and inhibitors currently undergoing clinical trials, although challenges and contradictions in the field persist. This review outlines the current literature on roles of neutrophils, neutrophil-derived molecules, and neutrophil heterogeneity in the pathogenesis of autoimmune and inflammatory diseases with potential future therapeutic strategies.
Collapse
Affiliation(s)
- Bushra Riaz
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Seonghyang Sohn
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
27
|
Huldani H, Abdul-Jabbar Ali S, Al-Dolaimy F, Hjazi A, Denis Andreevich N, Oudaha KH, Almulla AF, Alsaalamy A, Kareem Oudah S, Mustafa YF. The potential role of interleukins and interferons in ovarian cancer. Cytokine 2023; 171:156379. [PMID: 37757536 DOI: 10.1016/j.cyto.2023.156379] [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: 09/03/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
Ovarian cancer poses significant challenges and remains a highly lethal disease with limited treatment options. In the context of ovarian cancer, interleukins (ILs) and interferons (IFNs), important cytokines that play crucial roles in regulating the immune system, have emerged as significant factors influencing its development. This article provides a comprehensive review of the involvement of various ILs, including those from the IL-1 family, IL-2 family, IL-6 family, IL-8 family, IL-10 family, and IL-17 family, in ovarian cancer. The focus is on their impact on tumor growth, metastasis, and their role in evading immune responses within the tumor microenvironment. Additionally, the article conducts an in-depth examination of the oncogenic or antitumor roles of each IL in the context of ovarian cancer pathogenesis and progression. Besides, we elucidated the enhancements in the treatment of ovarian cancer through the utilization of type-I IFN and type-II IFN. Recent research has shed light on the intricate mechanisms through which specific ILs and IFNs contribute to the advancement of the disease. By incorporating recent findings, this review also seeks to inspire further investigations into unexplored mechanisms, fostering ongoing research to develop more effective therapeutic strategies for ovarian cancer. Moreover, through an in-depth analysis of IL- and IFN-associated clinical trials, we have highlighted their promising potential of in the treatment of ovarian cancer. These clinical trials serve to reinforce the significant outlook for utilizing ILs and IFNs as therapeutic agents in combating this disease.
Collapse
Affiliation(s)
- Huldani Huldani
- Department of Physiology, Faculty of Medicine, Lambung Mangkurat University, Banjarmasin, South Kalimantan, Indonesia
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Khulood H Oudaha
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Abbas F Almulla
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Shamam Kareem Oudah
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul 41001, Iraq
| |
Collapse
|
28
|
McGrouther DA. Hand infection: a management approach based on a new understanding of combined bacterial and neutrophil mediated tissue damage. J Hand Surg Eur Vol 2023; 48:838-848. [PMID: 37218740 DOI: 10.1177/17531934231174819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Concepts of tissue damage from sepsis are rooted in the works of Pasteur regarding colonization by microorganisms, and Lister's observation of avoiding suppuration by their exclusion. The reactive inflammation has been considered a beneficial defence mechanism. A more complex biology is now unfolding of pathogenic mechanisms with toxins produced by the organisms now being placed in a broad category of virulence factors. Neutrophils are key cells in providing innate immunity and their trafficking to sites of infection results in entry to the extracellular space where they attack pathogens by release of the contents of neutrophil granules and neutrophil extracellular traps. There is now considerable evidence that much of the tissue damage in infection is due to excessive host innate immunological reaction; a hyperinflammatory response, whether localized or systemic. In addition to traditional surgical methods of drainage and decompression there is now a focus on dilution of inflammatory mediators. This emerging knowledge can potentially alter the way we approach hand infections.
Collapse
|
29
|
Ribeiro Vitorino T, Ferraz do Prado A, Bruno de Assis Cau S, Rizzi E. MMP-2 and its implications on cardiac function and structure: Interplay with inflammation in hypertension. Biochem Pharmacol 2023; 215:115684. [PMID: 37459959 DOI: 10.1016/j.bcp.2023.115684] [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: 05/27/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023]
Abstract
Hypertension is one of the leading risk factors for the development of heart failure. Despite being a multifactorial disease, in recent years, preclinical and clinical studies suggest strong evidence of the pivotal role of inflammatory cells and cytokines in the remodeling process and cardiac dysfunction. During the heart remodeling, activation of extracellular matrix metalloproteinases (MMPs) occurs, with MMP-2 being one of the main proteases secreted by cardiomyocytes, fibroblasts, endothelial and inflammatory cells in cardiac tissue. In this review, we will address the process of cardiac remodeling and injury induced by the increase in MMP-2 and the main signaling pathways involving cytokines and inflammatory cells in the process of transcriptional, secretion and activation of MMP-2. In addition, an interaction and coordinated action between MMP-2 and inflammation are explored and significant in maintaining the cardiac cycle. These observations suggest that new therapeutic opportunities targeting MMP-2 could be used to reduce inflammatory biomarkers and reduce cardiac damage in hypertension.
Collapse
Affiliation(s)
- Thaís Ribeiro Vitorino
- Unit of Biotechnology, University of Ribeirao Preto, UNAERP, Brazil; Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, UNICAMP, Brazil
| | - Alejandro Ferraz do Prado
- Cardiovascular System Pharmacology and Toxicology, Institute of Biological Sciences, Federal University of Para, UFPA, Brazil
| | - Stefany Bruno de Assis Cau
- Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, UFMG, Brazil.
| | - Elen Rizzi
- Unit of Biotechnology, University of Ribeirao Preto, UNAERP, Brazil.
| |
Collapse
|
30
|
Huber A, Jose S, Kassam A, Weghorn KN, Powers-Fletcher M, Sharma D, Mukherjee A, Mathew A, Kulkarni N, Chandramouli S, Alder MN, Madan R. Olfactomedin-4 + neutrophils exacerbate intestinal epithelial damage and worsen host survival after Clostridioides difficile infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.553751. [PMID: 37662327 PMCID: PMC10473617 DOI: 10.1101/2023.08.21.553751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Neutrophils are key first responders to Clostridioides difficile infection (CDI). Excessive tissue and blood neutrophils are associated with worse histopathology and adverse outcomes, however their functional role during CDI remains poorly defined. Utilizing intestinal epithelial cell (IEC)-neutrophil co-cultures and a pre-clinical animal model of CDI, we show that neutrophils exacerbate C. difficile -induced IEC injury. We utilized cutting-edge single-cell transcriptomics to illuminate neutrophil subtypes and biological pathways that could exacerbate CDI-associated IEC damage. As such, we have established the first transcriptomics atlas of bone marrow (BM), blood, and colonic neutrophils after CDI. We found that CDI altered the developmental trajectory of BM and blood neutrophils towards populations that exhibit gene signatures associated with pro-inflammatory responses and neutrophil-mediated tissue damage. Similarly, the transcriptomic signature of colonic neutrophils was consistent with hyper-inflammatory and highly differentiated cells that had amplified expression of cytokine-mediated signaling and degranulation priming genes. One of the top 10 variable features in colonic neutrophils was the gene for neutrophil glycoprotein, Olfactomedin 4 (OLFM4). CDI enhanced OLFM4 mRNA and protein expression in neutrophils, and OLFM4 + cells aggregated to areas of severe IEC damage. Compared to uninfected controls, both humans and mice with CDI had higher concentrations of circulating OLFM4; and in mice, OLFM4 deficiency resulted in faster recovery and better survival after infection. Collectively, these studies provide novel insights into neutrophil-mediated pathology after CDI and highlight the pathogenic role of OLFM4 + neutrophils in regulating CDI-induced IEC damage. One Sentence Summary Utilizing single-cell transcriptomics, IEC-epithelial co-cultures, and pre-clinical models of CDI, we have identified a subset of neutrophils that are marked by OLFM4 expression as pathogenic determinants of IEC barrier damage after CDI.
Collapse
|
31
|
Libreros S, Nshimiyimana R, Lee B, Serhan CN. Infectious neutrophil deployment is regulated by resolvin D4. Blood 2023; 142:589-606. [PMID: 37295018 PMCID: PMC10447623 DOI: 10.1182/blood.2022019145] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 06/11/2023] Open
Abstract
Neutrophils reside in the bone marrow (BM), ready for deployment to sites of injury/infection, initiating inflammation and its resolution. Here, we report that distal infections signal to the BM via resolvins to regulate granulopoiesis and BM neutrophil deployment. Emergency granulopoiesis during peritonitis evoked changes in BM resolvin D1 (RvD1) and BM RvD4. We found that leukotriene B4 stimulates neutrophil deployment. RvD1 and RvD4 each limited neutrophilic infiltration to infections, and differently regulated BM myeloid populations: RvD1 increased reparative monocytes, and RvD4 regulated granulocytes. RvD4 disengaged emergency granulopoiesis, prevented excess BM neutrophil deployment, and acted on granulocyte progenitors. RvD4 also stimulated exudate neutrophil, monocyte, and macrophage phagocytosis, and enhanced bacterial clearance. This mediator accelerated both neutrophil apoptosis and clearance by macrophages, thus expediting the resolution phase of inflammation. RvD4 stimulated phosphorylation of ERK1/2 and STAT3 in human BM-aspirate-derived granulocytes. RvD4 in the 1 to 100 nM range stimulated whole-blood neutrophil phagocytosis of Escherichia coli. RvD4 increased BM macrophage efferocytosis of neutrophils. Together, these results demonstrate the novel functions of resolvins in granulopoiesis and neutrophil deployment, contributing to the resolution of infectious inflammation.
Collapse
Affiliation(s)
- Stephania Libreros
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Robert Nshimiyimana
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Brendon Lee
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
32
|
Schuurman AR, Butler JM, Michels EH, Otto NA, Brands X, Haak BW, Uhel F, Klarenbeek AM, Faber DR, Schomakers BV, van Weeghel M, de Vos AF, Scicluna BP, Houtkooper RH, Wiersinga WJ, van der Poll T. Inflammatory and glycolytic programs underpin a primed blood neutrophil state in patients with pneumonia. iScience 2023; 26:107181. [PMID: 37496676 PMCID: PMC10366455 DOI: 10.1016/j.isci.2023.107181] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/21/2023] [Accepted: 06/15/2023] [Indexed: 07/28/2023] Open
Abstract
Neutrophils are potent immune cells with key antimicrobial functions. Previous in vitro work has shown that neutrophil effector functions are mainly fueled by intracellular glycolysis. Little is known about the state of neutrophils still in the circulation in patients during infection. Here, we combined flow cytometry, stimulation assays, transcriptomics, and metabolomics to investigate the link between inflammatory and metabolic pathways in blood neutrophils of patients with community-acquired pneumonia. Patients' neutrophils, relative to neutrophils from age- and sex- matched controls, showed increased degranulation upon ex vivo stimulation, and portrayed distinct upregulation of inflammatory transcriptional programs. This neutrophil phenotype was accompanied by a high-energy state with increased intracellular ATP content, and transcriptomic and metabolic upregulation of glycolysis and glycogenolysis. One month after hospital admission, these metabolic and transcriptomic changes were largely normalized. These data elucidate the molecular programs that underpin a balanced, yet primed state of blood neutrophils during pneumonia.
Collapse
Affiliation(s)
- Alex R. Schuurman
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Joe M. Butler
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Erik H.A. Michels
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Natasja A. Otto
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Xanthe Brands
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Bastiaan W. Haak
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Fabrice Uhel
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Augustijn M. Klarenbeek
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Daniël R. Faber
- BovenIJ Hospital, Statenjachtstraat 1, 1034 CS Amsterdam, the Netherlands
| | - Bauke V. Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC, 1105 AZ Amsterdam, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, 1105 AZ Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC, 1105 AZ Amsterdam, the Netherlands
| | - Alex F. de Vos
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Brendon P. Scicluna
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta
| | - Riekelt H. Houtkooper
- Amsterdam Gastroenterology Endocrinology and Metabolism Institute, 1105 AZ Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences Institute, 1105 AZ Amsterdam, the Netherlands
| | - W. Joost Wiersinga
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers - Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| |
Collapse
|
33
|
Zhang C, Cao J, Xu M, Wu D, Li W, Chang Y. The role of neutrophils in chorioamnionitis. Front Immunol 2023; 14:1198831. [PMID: 37475854 PMCID: PMC10354368 DOI: 10.3389/fimmu.2023.1198831] [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: 04/02/2023] [Accepted: 06/16/2023] [Indexed: 07/22/2023] Open
Abstract
Chorioamnionitis, commonly referred to as intrauterine infection or inflammation, is pathologically defined by neutrophil infiltration and inflammation at the maternal-fetal interface. Chorioamnionitis is the common complication during late pregnancy, which lead to a series of serious consequences, such as preterm labor, preterm premature rupture of the fetal membranes, and fetal inflammatory response syndrome. During infection, a large number of neutrophils migrate to the chorio-decidua in response to chemokines. Although neutrophils, a crucial part of innate immune cells, have strong anti-inflammatory properties, over-activating them can harm the body while also eliminating pathogens. This review concentrated on the latest studies on chorioamnionitis-related consequences as well as the function and malfunction of neutrophils. The release of neutrophil extracellular traps, production of reactive oxygen species, and degranulation from neutrophils during intrauterine infection, as well as their pathological roles in complications related to chorioamnionitis, were discussed in detail, offering fresh perspectives on the treatment of chorioamnionitis.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Chang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin central hospital of Gynecology Obstetrics, Tianjin, China
| |
Collapse
|
34
|
Mariod AA, Jabbar AA, Alamri ZZ, Salim Al Rashdi A, Abdulla MA. Gastroprotective effects of Polygonatum odoratum in rodents by regulation of apoptotic proteins and inflammatory cytokines. Saudi J Biol Sci 2023; 30:103678. [PMID: 37266408 PMCID: PMC10230262 DOI: 10.1016/j.sjbs.2023.103678] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 06/03/2023] Open
Abstract
In an increasing interest in natural antiulcer compounds that may have gastric healing effects and possibly prevent ulcer recurrence, Polygonatum odoratum appears as a strong candidate. The gastroprotective potentials of P. odoratum rhizome extract (PORE) were explored on ethanol-induced gastric ulceration in rats. Sprague Dawley rats were caged in 5 groups, normal and ulcer control rats received CMC (1% carboxymethyl cellulose). Omeprazole (20 mg/kg) was given to reference Rats. Experimental rats were treated with 250 mg/kg and 500 mg/kg PORE, respectively. After an hour, the normal control rats received 1% CMC, whereas rat groups 2-5 were given absolute ethanol by oral gavage. After 60 min, rats received anesthesia and were sacrificed. Dissected gastric tissue was analyzed by histopathological and immunohistochemical techniques. PORE treatment significantly lowered the ethanol-induced gastric injury, as shown by up-surging gastric pH and mucus content, reduced leukocyte infiltration, lower ulcerative areas in mucosal layers, and increased antioxidants (SOD and CAT) and (MDA) levels. Furthermore, PORE pre-treated rats showed significantly increased expression of the Periodic acid-Schiff (PAS), HSP-70 protein, and decreased Bax protein in their gastric epithelial layers. PORE treatment showed an important regulation of inflammatory cytokines shown by decreasing the TNF-a, and IL-6 and increasing the IL-10 values. The detected biological activity of PORE is encouraging and presents the scientific evidence for its traditional use as a gastroprotection agent however further studies are required to determine the exact phytochemicals and mechanism pathway responsible for this bioactivity.
Collapse
Affiliation(s)
- Abdalbasit A. Mariod
- College of Science and Arts, Alkamil Branch, University of Jeddah, Alkamil 21931, Saudi Arabia
- Indigenous Knowledge and Heritage Centre, Ghibaish College of Science and Technology, 110 Ghibaish, Sudan
| | - Ahmed A.J. Jabbar
- Department of Medical Laboratory Technology, Erbil Technical Health and Medical College, Erbil Polytechnic University, Erbil 44001, Iraq
| | - Zaenah Zuhair Alamri
- Department of Biological Sciences, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | | | - Mahmood Ameen Abdulla
- Department of Medical Microbiology, College of Science, Cihan University-Erbil, Erbil 44001, Kurdistan Region, Iraq
| |
Collapse
|
35
|
Wu Z, Huang W, He X, Dutta S, Paul C, Fan GC, Kanisicak O, Xu M, Liang J, Wang Y. Myocardial IGF2R is a critical mediator of inflammation and fibrosis after ischemia-reperfusion injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537835. [PMID: 37131709 PMCID: PMC10153233 DOI: 10.1101/2023.04.21.537835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Ischemia-reperfusion (I/R) injury is a common occurrence in various surgical procedures used to treat heart diseases. However, the role of insulin-like growth factor 2 receptor (IGF2R) during the process of myocardial I/R remains unclear. Therefore, this study aims to investigate the expression, distribution, and functionality of IGF2R in various I/R-associated models (such as reoxygenation, revascularization, and heart transplant). Loss-of-function studies (including myocardial conditional knockout and CRISPR interference) were performed to clarify the role of IGF2R in I/R injuries. Following hypoxia, IGF2R expression increased, but this effect was reversed upon restoration of oxygen levels. Loss of myocardial IGF2R was found to enhance the cardiac contractile functions, and reduced cell infiltration or cardiac fibrosis of I/R mouse models compared to the genotype control. CRISPR-inhibition of IGF2R decreased cell apoptotic death under hypoxia. RNA sequencing analysis indicated that myocardial IGF2R played a critical role in regulating the inflammatory response, innate immune response, and apoptotic process following I/R. Integrated analysis of the mRNA profiling, pulldown assays, and mass spectrometry identified granulocyte-specific factors as potential targets of myocardial IGF2R in the injured heart. In conclusion, myocardial IGF2R emerges as a promising therapeutic target to ameliorate inflammation or fibrosis following I/R injuries.
Collapse
|
36
|
Sorobetea D, Matsuda R, Peterson ST, Grayczyk JP, Rao I, Krespan E, Lanza M, Assenmacher CA, Mack M, Beiting DP, Radaelli E, Brodsky IE. Inflammatory monocytes promote granuloma control of Yersinia infection. Nat Microbiol 2023; 8:666-678. [PMID: 36879169 PMCID: PMC10653359 DOI: 10.1038/s41564-023-01338-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/09/2023] [Indexed: 03/08/2023]
Abstract
Granulomas are organized immune cell aggregates formed in response to chronic infection or antigen persistence. The bacterial pathogen Yersinia pseudotuberculosis (Yp) blocks innate inflammatory signalling and immune defence, inducing neutrophil-rich pyogranulomas (PGs) within lymphoid tissues. Here we uncover that Yp also triggers PG formation within the murine intestinal mucosa. Mice lacking circulating monocytes fail to form defined PGs, have defects in neutrophil activation and succumb to Yp infection. Yersinia lacking virulence factors that target actin polymerization to block phagocytosis and reactive oxygen burst do not induce PGs, indicating that intestinal PGs form in response to Yp disruption of cytoskeletal dynamics. Notably, mutation of the virulence factor YopH restores PG formation and control of Yp in mice lacking circulating monocytes, demonstrating that monocytes override YopH-dependent blockade of innate immune defence. This work reveals an unappreciated site of Yersinia intestinal invasion and defines host and pathogen drivers of intestinal granuloma formation.
Collapse
Affiliation(s)
- Daniel Sorobetea
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rina Matsuda
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefan T Peterson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James P Grayczyk
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Indira Rao
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elise Krespan
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Matthias Mack
- Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Daniel P Beiting
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Igor E Brodsky
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Contini C, Serrao S, Manconi B, Olianas A, Iavarone F, Guadalupi G, Messana I, Castagnola M, Masullo C, Bizzarro A, Turck CW, Maccarrone G, Cabras T. Characterization of Cystatin B Interactome in Saliva from Healthy Elderly and Alzheimer’s Disease Patients. Life (Basel) 2023; 13:life13030748. [PMID: 36983903 PMCID: PMC10054399 DOI: 10.3390/life13030748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Cystatin B is a small, multifunctional protein involved in the regulation of inflammation, innate immune response, and neuronal protection and found highly abundant in the brains of patients with Alzheimer’s disease (AD). Recently, our study demonstrated a significant association between the level of salivary cystatin B and AD. Since the protein is able to establish protein-protein interaction (PPI) in different contexts and aggregation-prone proteins and the PPI networks are relevant for AD pathogenesis, and due to the relevance of finding new AD markers in peripheral biofluids, we thought it was interesting to study the possible involvement of cystatin B in PPIs in saliva and to evaluate differences and similarities between AD and age-matched elderly healthy controls (HC). For this purpose, we applied a co-immunoprecipitation procedure and a bottom-up proteomics analysis to purify, identify, and quantify cystatin B interactors. Results demonstrated for the first time the existence of a salivary cystatin B-linked multi-protein complex composed by 82 interactors and largely expressed in the body. Interactors are involved in neutrophil activation, antimicrobial activity, modulation of the cytoskeleton and extra-cellular matrix (ECM), and glucose metabolism. Preliminary quantitative data showed significantly lower levels of triosophosphate isomerase 1 and higher levels of mucin 7, BPI, and matrix Gla protein in AD with respect to HC, suggesting implications associated with AD of altered glucose metabolism, antibacterial activities, and calcification-associated processes. Data are available via ProteomeXchange with identifiers PXD039286 and PXD030679.
Collapse
Affiliation(s)
- Cristina Contini
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Simone Serrao
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
- Correspondence:
| | - Alessandra Olianas
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Federica Iavarone
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Policlinico Universitario “A. Gemelli” Foundation IRCCS, 00168 Rome, Italy
| | - Giulia Guadalupi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Irene Messana
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy
| | - Massimo Castagnola
- Proteomics Laboratory, European Center for Brain Research, (IRCCS) Santa Lucia Foundation, 00168 Rome, Italy
| | - Carlo Masullo
- Department of Neuroscience, Neurology Section, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Christoph W. Turck
- Proteomics and Biomarkers, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Giuseppina Maccarrone
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Tiziana Cabras
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy
| |
Collapse
|
38
|
Abstract
Neutrophils or polymorphonuclear neutrophils (PMNs) are an important component of innate host defense. These phagocytic leukocytes are recruited to infected tissues and kill invading microbes. There are several general characteristics of neutrophils that make them highly effective as antimicrobial cells. First, there is tremendous daily production and turnover of granulocytes in healthy adults-typically 1011 per day. The vast majority (~95%) of these cells are neutrophils. In addition, neutrophils are mobilized rapidly in response to chemotactic factors and are among the first leukocytes recruited to infected tissues. Most notably, neutrophils contain and/or produce an abundance of antimicrobial molecules. Many of these antimicrobial molecules are toxic to host cells and can destroy host tissues. Thus, neutrophil activation and turnover are highly regulated processes. To that end, aged neutrophils undergo apoptosis constitutively, a process that contains antimicrobial function and proinflammatory capacity. Importantly, apoptosis facilitates nonphlogistic turnover of neutrophils and removal by macrophages. This homeostatic process is altered by interaction with microbes and their products, as well as host proinflammatory molecules. Microbial pathogens can delay neutrophil apoptosis, accelerate apoptosis following phagocytosis, or cause neutrophil cytolysis. Here, we review these processes and provide perspective on recent studies that have potential to impact this paradigm.
Collapse
Affiliation(s)
- Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, USA
| | - Mark T Quinn
- Department of Microbiology & Cell Biology, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
39
|
Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil Extracellular Traps in Cerebral Ischemia/Reperfusion Injury: Friend and Foe. Curr Neuropharmacol 2023; 21:2079-2096. [PMID: 36892020 PMCID: PMC10556361 DOI: 10.2174/1570159x21666230308090351] [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: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 03/10/2023] Open
Abstract
Cerebral ischemic injury, one of the leading causes of morbidity and mortality worldwide, triggers various central nervous system (CNS) diseases, including acute ischemic stroke (AIS) and chronic ischemia-induced Alzheimer's disease (AD). Currently, targeted therapies are urgently needed to address neurological disorders caused by cerebral ischemia/reperfusion injury (CI/RI), and the emergence of neutrophil extracellular traps (NETs) may be able to relieve the pressure. Neutrophils are precursors to brain injury following ischemic stroke and exert complicated functions. NETs extracellularly release reticular complexes of neutrophils, i.e., double-stranded DNA (dsDNA), histones, and granulins. Paradoxically, NETs play a dual role, friend and foe, under different conditions, for example, physiological circumstances, infection, neurodegeneration, and ischemia/reperfusion. Increasing evidence indicates that NETs exert anti-inflammatory effects by degrading cytokines and chemokines through protease at a relatively stable and moderate level under physiological conditions, while excessive amounts of NETs release (NETosis) irritated by CI/RI exacerbate the inflammatory response and aggravate thrombosis, disrupt the blood-brain barrier (BBB), and initiates sequential neuron injury and tissue damage. This review provides a comprehensive overview of the machinery of NETs formation and the role of an abnormal cascade of NETs in CI/RI, as well as other ischemia-induced neurological diseases. Herein, we highlight the potential of NETs as a therapeutic target against ischemic stroke that may inspire translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Rui Fang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| |
Collapse
|
40
|
Vakhrusheva TV, Sokolov AV, Moroz GD, Kostevich VA, Gorbunov NP, Smirnov IP, Grafskaia EN, Latsis IA, Panasenko OM, Lazarev VN. Effects of Synthetic Short Cationic Antimicrobial Peptides on the Catalytic Activity of Myeloperoxidase, Reducing Its Oxidative Capacity. Antioxidants (Basel) 2022; 11:antiox11122419. [PMID: 36552626 PMCID: PMC9774438 DOI: 10.3390/antiox11122419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Cationic antimicrobial peptides (CAMPs) have gained attention as promising antimicrobial therapeutics causing lower or no bacterial resistance. Considerable achievements have been made in designing new CAMPs that are highly active as antimicrobials. However, there is a lack of research on their interaction with biologically important proteins. This study focused on CAMPs' effects on myeloperoxidase (MPO), an enzyme which is microbicidal and concomitantly damaging to host biomolecules and cells due to its ability to produce reactive oxygen and halogen species (ROS/RHS). Four CAMPs designed by us were employed. MPO catalytic activity was assessed by an absorbance spectra analysis and by measuring enzymatic activity using Amplex Red- and Celestine Blue B-based assays. The peptide Hm-AMP2 accelerated MPO turnover. Pept_1545 and Hm-AMP8 inhibited both the MPO chlorinating and peroxidase activities, with components of different inhibition types. Hm-AMP8 was a stronger inhibitor. Its Ki towards H2O2 and Cl- was 0.3-0.4 μM vs. 11-20 μM for pept_1545. Peptide tyrosine and cysteine residues were involved in the mechanisms of the observed effects. The results propose a possible dual role of CAMPs as both antimicrobial agents and agents that downregulate MPO activation, and suggest CAMPs as prototypes for the development of antioxidant compounds to prevent MPO-mediated ROS/RHS overproduction.
Collapse
Affiliation(s)
- Tatyana V. Vakhrusheva
- Department of Biophysics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Correspondence:
| | - Alexey V. Sokolov
- Department of Biophysics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Department of Molecular Genetics, Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Grigoriy D. Moroz
- Department of Biophysics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology (State University), Moscow Region, 141701 Dolgoprudny, Russia
| | - Valeria A. Kostevich
- Department of Biophysics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Department of Molecular Genetics, Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Nikolay P. Gorbunov
- Department of Biophysics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Department of Molecular Genetics, Institute of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Igor P. Smirnov
- Department of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Ekaterina N. Grafskaia
- Department of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Ivan A. Latsis
- Department of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Oleg M. Panasenko
- Department of Biophysics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Vassili N. Lazarev
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology (State University), Moscow Region, 141701 Dolgoprudny, Russia
- Department of Cell Biology, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| |
Collapse
|
41
|
Albaladejo-Riad N, Cuesta A, Esteban MÁ. Induction and characterization of extracellular traps by gilthead seabream (Sparus aurata L.) head-kidney leucocytes. FISH & SHELLFISH IMMUNOLOGY 2022; 130:582-590. [PMID: 36152801 DOI: 10.1016/j.fsi.2022.09.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 06/16/2023]
Abstract
The aim of this study was the induction and characterization of extracellular traps (ETs) produced by gilthead seabream (Sparus aurata L.) head-kidney leucocytes. The cells were incubated several times (10, 30, 60, 120, and 180 min) with different concentrations of the stimulants diluted in RPMI-1640 culture medium: RPMI-1640 (control), β-glucan from Saccharomyces cerevisiae (BG, 0-400 μg mL-1), lipopolysaccharide from Escherichia coli (LPS, 0-10 μg mL-1), calcium ionophore A23187 (CaI, 0-5 μg mL-1), Phorbol 12-myristate 13-acetate (PMA, 0-1000 ng mL-1) and polyinosinic-polycytidylic acid sodium salt (Poly I:C, 0-200 μg mL-1). BG, LPS and CaI exerted only weak stimulatory activity, while PMA and poly I:C exerted a potent one. After stimulation of the leucocytes, ETs structures were quantified and visualised through staining of the chromatin with nucleic acid-specific dyes and immunocytochemical probing of characteristic proteins expected to decorate the structure. ETs structures had DNA and myeloperoxidase. The ETs morphology was studied by light and scanning electron microscopy. These data confirm that seabream leucocytes form ETs with different morphological properties, depending on the used stimulant. These results will be the basis for new studies to analyse the implication of this mechanism in fish immunity. All this new knowledge will have its application in fish farms when we learn to manipulate the innate immune response in order to mitigate microbial infections.
Collapse
Affiliation(s)
- Nora Albaladejo-Riad
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain
| | - Alberto Cuesta
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain
| | - M Ángeles Esteban
- Immunobiology for Aquaculture Group, Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, 30100, Murcia, Spain.
| |
Collapse
|
42
|
Gimeno-Molina B, Muller I, Kropf P, Sykes L. The Role of Neutrophils in Pregnancy, Term and Preterm Labour. Life (Basel) 2022; 12:life12101512. [PMID: 36294949 PMCID: PMC9605051 DOI: 10.3390/life12101512] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/20/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Neutrophils are surveillance cells, and the first to react and migrate to sites of inflammation and infection following a chemotactic gradient. Neutrophils play a key role in both sterile inflammation and infection, performing a wide variety of effector functions such as degranulation, phagocytosis, ROS production and release of neutrophil extracellular traps (NETs). Healthy term labour requires a sterile pro-inflammatory process, whereas one of the most common causes of spontaneous preterm birth is microbial driven. Peripheral neutrophilia has long been described during pregnancy, and evidence exists demonstrating neutrophils infiltrating the cervix, uterus and foetal membranes during both term and preterm deliveries. Their presence supports a role in tissue remodelling via their effector functions. In this review, we describe the effector functions of neutrophils. We summarise the evidence to support their role in healthy pregnancy and labour and describe their potential contribution to microbial driven preterm birth.
Collapse
Affiliation(s)
- Belen Gimeno-Molina
- Imperial College Parturition Research Group, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0HS, UK
- March of Dimes European Prematurity Research Centre, Imperial College London, London W12 0HS, UK
| | - Ingrid Muller
- Department of Infectious Diseases, Imperial College London, London W2 1NY, UK
| | - Pascale Kropf
- March of Dimes European Prematurity Research Centre, Imperial College London, London W12 0HS, UK
- Department of Infectious Diseases, Imperial College London, London W2 1NY, UK
| | - Lynne Sykes
- Imperial College Parturition Research Group, Department of Metabolism, Digestion and Reproduction, Imperial College London, London W12 0HS, UK
- March of Dimes European Prematurity Research Centre, Imperial College London, London W12 0HS, UK
- The Parasol Foundation Centre for Women’s Health and Cancer Research, St. Mary’s Hospital, Imperial College Healthcare NHS Trust, London W2 1NY, UK
- Correspondence:
| |
Collapse
|
43
|
Safrastyan A, Wollny D. Network analysis of hepatocellular carcinoma liquid biopsies augmented by single-cell sequencing data. Front Genet 2022; 13:921195. [PMID: 36092896 PMCID: PMC9452847 DOI: 10.3389/fgene.2022.921195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
Liquid biopsy, the analysis of body fluids, represents a promising approach for disease diagnosis and prognosis with minimal intervention. Sequencing cell-free RNA derived from liquid biopsies has been very promising for the diagnosis of several diseases. Cancer research, in particular, has emerged as a prominent candidate since early diagnosis has been shown to be a critical determinant of disease prognosis. Although high-throughput analysis of liquid biopsies has uncovered many differentially expressed genes in the context of cancer, the functional connection between these genes is not investigated in depth. An important approach to remedy this issue is the construction of gene networks which describes the correlation patterns between different genes, thereby allowing to infer their functional organization. In this study, we aimed at characterizing extracellular transcriptome gene networks of hepatocellular carcinoma patients compared to healthy controls. Our analysis revealed a number of genes previously associated with hepatocellular carcinoma and uncovered their association network in the blood. Our study thus demonstrates the feasibility of performing gene co-expression network analysis from cell-free RNA data and its utility in studying hepatocellular carcinoma. Furthermore, we augmented cell-free RNA network analysis with single-cell RNA sequencing data which enables the contextualization of the identified network modules with cell-type specific transcriptomes from the liver.
Collapse
Affiliation(s)
- Aram Safrastyan
- RNA Bioinformatics and High Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
| | - Damian Wollny
- RNA Bioinformatics and High Throughput Analysis, Friedrich Schiller University Jena, Jena, Germany
- Leibniz Institute on Aging-Fritz Lipmann Institute (FLI), Jena, Germany
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- *Correspondence: Damian Wollny,
| |
Collapse
|
44
|
Myeloperoxidase as a Marker to Differentiate Mouse Monocyte/Macrophage Subsets. Int J Mol Sci 2022; 23:ijms23158246. [PMID: 35897821 PMCID: PMC9330004 DOI: 10.3390/ijms23158246] [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: 06/30/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 02/01/2023] Open
Abstract
Macrophages are present in every tissue in the body and play essential roles in homeostasis and host defense against microorganisms. Some tissue macrophages derive from the yolk sac/fetal liver that populate tissues for life. Other tissue macrophages derive from monocytes that differentiate in the bone marrow and circulate through tissues via the blood and lymphatics. Circulating monocytes are very plastic and differentiate into macrophages with specialized functions upon entering tissues. Specialized monocyte/macrophage subsets have been difficult to differentiate based on cell surface markers. Here, using a combination of "pan" monocyte/macrophage markers and flow cytometry, we asked whether myeloperoxidase (MPO) could be used as a marker of pro-inflammatory monocyte/macrophage subsets. MPO is of interest because of its potent microbicidal activity. In wild-type SPF housed mice, we found that MPO+ monocytes/macrophages were present in peripheral blood, spleen, small and large intestines, and mesenteric lymph nodes, but not the central nervous system. Only monocytes/macrophages that expressed cell surface F4/80 and/or Ly6C co-expressed MPO with the highest expression in F4/80HiLy6CHi subsets regardless of tissue. These cumulative data indicate that MPO expression can be used as an additional marker to differentiate between monocyte/macrophage subsets with pro-inflammatory and microbicidal activity in a variety of tissues.
Collapse
|
45
|
Afonina IS, Hoffmann E. Infection and immunity: ‘There Are Things Out There You (Don't) Need To Know About’. FEBS J 2022; 289:3920-3925. [DOI: 10.1111/febs.16569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Inna S. Afonina
- Unit of Molecular Signal Transduction in Inflammation VIB‐UGent Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Belgium
| | - Eik Hoffmann
- Institute Pasteur Lille Center for Infection and Immunity of Lille (CIIL) France
- European Regimen Accelerator for Tuberculosis (ERA4TB), Innovative Medicines Initiative 2 Joint Undertaking (JU no. 853989)
| |
Collapse
|
46
|
Liu ML, Lyu X, Werth VP. Recent progress in the mechanistic understanding of NET formation in neutrophils. FEBS J 2022; 289:3954-3966. [PMID: 34042290 PMCID: PMC9107956 DOI: 10.1111/febs.16036] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 01/03/2023]
Abstract
Neutrophils are the most abundant circulating white blood cells and one of the major cell types of the innate immune system. Neutrophil extracellular traps (NETs) are a result of the extracellular release of nuclear chromatin from the ruptured nuclear envelope and plasma membrane. The externalized chromatin is an ancient defense weapon for animals to entrap and kill microorganisms in the extracellular milieu, thus protecting animals ranging from lower invertebrates to higher vertebrates. Although the externalized chromatin has the advantage of acting as anti-infective to protect against infections, extracellular chromatin might be problematic in higher vertebrate animals as they have an adaptive immune system that can trigger further immune or autoimmune responses. NETs and their associated nuclear and/or cytoplasmic components may induce sterile inflammation, immune, and autoimmune responses, leading to various human diseases. Though important in human pathophysiology, the cellular and molecular mechanisms of NET formation (also called NETosis) are not well understood. Given that nuclear chromatin forms the backbone of NETs, the nucleus is the root of the nuclear DNA extracellular traps. Thus, nuclear chromatin decondensation, along with the rupture of nuclear envelope and plasma membrane, is required for nuclear chromatin extracellular release and NET formation. So far, most of the literature focuses on certain signaling pathways, which are involved in NET formation but without explanation of cellular events and morphological changes described above. Here, we have summarized emerging evidence and discuss new mechanistic understanding, with our perspectives, in NET formation in neutrophils.
Collapse
Affiliation(s)
- Ming-Lin Liu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xing Lyu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA,Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Victoria P. Werth
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
47
|
Hu B, Li D, Zeng Z, Zhang Z, Cao R, Dong X, Yun C, Li L, Krämer B, Morgera S, Hocher B, Tang D, Yin L, Dai Y. Integrated proteome and malonylome analyses reveal the neutrophil extracellular trap formation pathway in rheumatoid arthritis. J Proteomics 2022; 262:104597. [PMID: 35489682 DOI: 10.1016/j.jprot.2022.104597] [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: 10/21/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 12/09/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease of unknown etiology in which the posttranslational modifications (PTMs) of proteins play an important role. PTMs, such as those involved in the formation of neutrophil extracellular traps (NETs), have been well studied. The excessive formation and release of NETs can mediate inflammation and joint destruction in RA. It has been gradually recognized that lysine malonylation (Kmal) can regulate some biological processes in some prokaryotes and eukaryotes. However, less is known about the role of Kmal in RA. We therefore performed proteome and malonylome analyses to explore the proteomic characteristics of the peripheral blood mononuclear cells from 36 RA patients and 82 healthy subjects. In total, 938 differentially expressed proteins (DEPs) and 42 differentially malonylated proteins (DMPs) with 55 Kmal sites were detected through a liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based analysis. Functional analysis showed that two DEPs with four malonylated sites and one DMP with a malonylated site were identified in the neutrophil extracellular trap formation (NETosis) pathway. Altogether, this study not only describes the characteristics of the malonylome in RA for the first time, but it also reveals that malonylation may be involved in the NETosis pathway. SIGNIFICANCE: This is the first report that reveals the proteomic features of Kmal in RA through a LC-MS/MS-based method. In this study, we found that several key DMPs were associated with the NETosis pathway, which contributes to the development of RA. The present results provide an informative dataset for the future exploration of Kmal in RA.
Collapse
Affiliation(s)
- Biying Hu
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Dandan Li
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China; Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China
| | - Zhipeng Zeng
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China
| | - Zeyu Zhang
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Rui Cao
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - XiangNan Dong
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China
| | - Chen Yun
- Guangzhou Enttxs Medical Products Co., Ltd. P.R. Guangzhou, Guangdong, 510663, China; Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Ling Li
- Hospital of South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Bernhard Krämer
- Department of Medicine Nephrologh, Medical Faculty Mannheim Heideiberg University, 68167 Mannheim, Germany
| | | | - Berthold Hocher
- Department of Medicine Nephrologh, Medical Faculty Mannheim Heideiberg University, 68167 Mannheim, Germany; Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Donge Tang
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China.
| | - Lianghong Yin
- The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong 510632, China; Huangpu Institute of Materials, Guangzhou, Guangdong, 510663, China.
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong 518020, China.
| |
Collapse
|
48
|
Filep JG. Targeting Neutrophils for Promoting the Resolution of Inflammation. Front Immunol 2022; 13:866747. [PMID: 35371088 PMCID: PMC8966391 DOI: 10.3389/fimmu.2022.866747] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a localized and self-limited innate host-defense mechanism against invading pathogens and tissue injury. Neutrophils, the most abundant immune cells in humans, play pivotal roles in host defense by eradicating invading pathogens and debris. Ideally, elimination of the offending insult prompts repair and return to homeostasis. However, the neutrophils` powerful weaponry to combat microbes can also cause tissue damage and neutrophil-driven inflammation is a unifying mechanism for many diseases. For timely resolution of inflammation, in addition to stopping neutrophil recruitment, emigrated neutrophils need to be disarmed and removed from the affected site. Accumulating evidence documents the phenotypic and functional versatility of neutrophils far beyond their antimicrobial functions. Hence, understanding the receptors that integrate opposing cues and checkpoints that determine the fate of neutrophils in inflamed tissues provides insight into the mechanisms that distinguish protective and dysregulated, excessive inflammation and govern resolution. This review aims to provide a brief overview and update with key points from recent advances on neutrophil heterogeneity, functional versatility and signaling, and discusses challenges and emerging therapeutic approaches that target neutrophils to enhance the resolution of inflammation.
Collapse
Affiliation(s)
- János G Filep
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| |
Collapse
|
49
|
The role of neutrophils in rheumatic disease-associated vascular inflammation. Nat Rev Rheumatol 2022; 18:158-170. [PMID: 35039664 DOI: 10.1038/s41584-021-00738-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2021] [Indexed: 12/13/2022]
Abstract
Vascular pathologies underpin and intertwine autoimmune rheumatic diseases and cardiovascular conditions, and atherosclerosis is increasingly recognized as the leading cause of morbidity in conditions such as systemic lupus erythematosus (SLE), rheumatoid arthritis and antineutrophil cytoplasmic antibody-associated vasculitis. Neutrophils, important cells in the innate immune system, exert their functional effects in tissues via a variety of mechanisms, including the generation of neutrophil extracellular traps and the production of reactive oxygen species. Neutrophils have been implicated in the pathogenesis of several rheumatic diseases, and can also intimately interact with the vascular system, either through modulating endothelial barriers at the blood-vessel interface, or through associations with platelets. Emerging data suggest that neutrophils also have an important role maintaining homeostasis in individual organs and can protect the vascular system. Furthermore, studies using high-dimensional omics technologies have advanced our understanding of neutrophil diversity, and immature neutrophils are receiving new attention in rheumatic diseases including SLE and systemic vasculitis. Developments in genomic, imaging and organoid technologies are beginning to enable more in-depth investigations into the pathophysiology of vascular inflammation in rheumatic diseases, making now a good time to re-examine the full scope of roles of neutrophils in these processes.
Collapse
|
50
|
Oarada M, Okumura Y, Hirasaka K, Sugiura K, Tachibana N, Tsurusaki Y, Nikawa T. Dietary Sodium Nitrite Causes Similar Modifications to Splenic Inflammatory Gene Expression as a High-Fat Diet. J Nutr Sci Vitaminol (Tokyo) 2022; 67:404-416. [PMID: 34980719 DOI: 10.3177/jnsv.67.404] [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: 11/27/2022]
Abstract
Sodium nitrite (NaNO2) is a widely used food additive. The present study compared the outcomes from intakes of dietary NaNO2 and a high-fat diet (HFD), and assessed their combined effects on inflammatory gene expression in the immune tissues of the mouse. In experiment I, mice were fed a standard low-fat diet (LFD) without or with NaNO2 (0.02 and 0.08%, w/w) for 11 wk. In experiment II, mice were fed an LFD without or with NaNO2 (0.02%) or HFD without or with NaNO2 (0.02%) for 11 wk. Inflammatory gene expression in the immune tissues was then measured. NaNO2 consumption and HFD feeding each resulted in increased splenic mRNAs for cell markers of neutrophils (Ngp, NE, Ly6g, Mpo) and eosinophils (Epo, Ear6), and an S100 family member (S100A8). In contrast, NaNO2 consumption and HFD feeding each resulted in decreased splenic mRNAs for cell markers of macrophages (Emr1, Itgax, CD68, CD206, Dectin-1, TLRs 4, 6, and 7), T- (CD3, CD4), NK- (CD56) and B-cells (CD20, CD40), pro- and anti-inflammatory cytokines (TNF-α, IL-6, IL-1β, IFN-γ, IL-18, IL-10, TGF-β), interleukin receptor antagonists (IL1ra, IL6ra) and cell adhesion molecules (ICAM-1, VCAM-1). However, dietary NaNO2 combined with HFD feeding caused no further decrease in these transcript levels compared with dietary NaNO2 alone. These NaNO2- or HFD-induced modifications were less profound in the liver and abdominal adipose tissues than in the spleen. These findings indicate that dietary NaNO2 has similar modulatory effects to HFD feeding on splenic inflammatory genes.
Collapse
Affiliation(s)
- Motoko Oarada
- Faculty of Nutritional Science, Sagami Women's University
| | - Yuushi Okumura
- Faculty of Nutritional Science, Sagami Women's University
| | - Katsuya Hirasaka
- Graduate School of Fisheries Science and Environmental Studies, Nagasaki University
| | - Kosuke Sugiura
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School
| | | | | | - Takeshi Nikawa
- Department of Nutritional Physiology, Institute of Medical Nutrition, Tokushima University Graduate School
| |
Collapse
|