1
|
Asad S, Ahl D, Suárez‐López YDC, Erdélyi M, Phillipson M, Teleki A. Click Chemistry-Based Bioconjugation of Iron Oxide Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407883. [PMID: 39924809 PMCID: PMC11922026 DOI: 10.1002/smll.202407883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/19/2025] [Indexed: 02/11/2025]
Abstract
Superparamagnetic iron oxide nanoparticles (SPIONs) exhibit unique properties for diverse biomedical applications, including drug delivery and diagnostic imaging. Actively targeted SPIONs enhance delivery to diseased sites, reducing side effects and enhancing treatment efficacy. However, the development of reproducible functionalization protocols is challenged by the erratic behavior of nanoparticles in suspensions, such as agglomeration and sedimentation. In this study, a functionalization method is developed and systematically optimized to attach the Fc-region of antibodies onto silica-coated SPIONs via click chemistry, ensuring controlled ligand orientation on the particle surface. The synthesis and successive modifications of silica-coated SPIONs with organic moieties are presented resulting in the final click conjugation with antibodies targeting intercellular adhesion molecule 1 (ICAM1). This protein is upregulated on epithelial cell surfaces during gastrointestinal inflammation. Thermogravimetric analysis and infrared spectroscopy confirm successful SPION functionalization after each modification step. Cell viability assessment indicates no adverse effects of bioconjugated particles. Quantitative elemental analysis reveals significantly higher iron concentration in inflammation-induced Caco-2 cells exposed to ICAM1-modified particles compared to non-conjugated counterparts. Furthermore, laser scanning confocal microscopy of these cells suggests surface interaction and internalization of bioconjugated SPIONs, underscoring their potential for targeted imaging and therapy in inflammatory diseases.
Collapse
Affiliation(s)
- Shno Asad
- Department of PharmacyScience for Life LaboratoryUppsala UniversityUppsalaSE‐75123Sweden
| | - David Ahl
- Department of Medical Cell BiologyScience for Life LaboratoryUppsala UniversityUppsalaSE‐75123Sweden
| | | | - Máté Erdélyi
- Department of Chemistry – BMCUppsala UniversityUppsalaSE‐75123Sweden
| | - Mia Phillipson
- Department of Medical Cell BiologyScience for Life LaboratoryUppsala UniversityUppsalaSE‐75123Sweden
| | - Alexandra Teleki
- Department of PharmacyScience for Life LaboratoryUppsala UniversityUppsalaSE‐75123Sweden
| |
Collapse
|
2
|
He J, Zhao G, Chen M, Ren X, Zhu P, Liu Z, Zhou J, Chen H, Xiao C, Li XG. Identification and functional analysis of hub genes involved in deoxynivalenol-induced enterotoxicity in porcine (Sus scrofa). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117544. [PMID: 39675078 DOI: 10.1016/j.ecoenv.2024.117544] [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: 08/05/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
Deoxynivalenol (DON) is a type of mycotoxin commonly found in food and animal feed. When consumed, it can have harmful effects on the intestine. The porcine digestive system is physiologically similar to that of humans, making pigs a suitable model for studying DON-induced enterotoxicity. However, the exact ways DON causes intestinal damage in pigs still need to be fully understood. To address this knowledge gap, this study aimed to identify hub genes associated with enterotoxicity caused by DON exposure. Transcriptomic datasets from porcine jejunal explants exposed to DON were extensively analyzed using bioinformatic techniques in this study. A total of 265 differentially expressed genes (DEGs) were identified, with 238 being up-regulated and 27 being down-regulated, indicating that exposure to DON tends to increase gene expression. Further analysis revealed that the up-regulated DEGs were enriched in tumor necrosis factor, nuclear factor kappa-B, mitogen-activated protein kinases, and Janus kinase/signal transducer and activator of transcription-related signaling pathways. In addition, Weighted gene co-expression network analysis was performed to identify highly co-expressed modules. Then, genes in the highest co-expressed module were intersected with the up-regulated DEGs to construct a Protein-Protein Interaction network, resulting in 237 overlapping genes. Subsequently, 6 hub genes (CXCR4, PTGS2, ICAM1, IL-1A, IL-1B, and IL-10) that played a central role in the response to DON were identified using cytohubba in conjunction with the Molecular Complex Detection. In summary, exposure to DON is more likely to result in increased rather than decreased gene expression. Six of the upregulated genes, which are involved in immunoregulation and inflammation, were identified as hub genes related to DON-induced enterotoxicity in pigs. This study provides new insights into the mechanisms underlying DON-induced enterotoxicity and could guide interventions for this condition.
Collapse
Affiliation(s)
- Jinhua He
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, China
| | - Geng Zhao
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, China; Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Mingxia Chen
- School of Animal Science and Technology, Guangdong Polytechnic of Science and Trade, Qingyuan 511500, China
| | - Ximing Ren
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Peizhi Zhu
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhizhong Liu
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, China; Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jiayi Zhou
- College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hanwei Chen
- Central Laboratory, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou 511400, China; Panyu Health Management Center (Panyu Rehabilitation Hospital), Guangzhou 511450, China
| | - Chuqiao Xiao
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Xiang-Guang Li
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
3
|
Yoshida M, Arzili R, Nikolić MZ. Immune-epithelial cell interactions in lung development, homeostasis and disease. Int J Biochem Cell Biol 2025; 178:106703. [PMID: 39592067 DOI: 10.1016/j.biocel.2024.106703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/25/2024] [Accepted: 11/18/2024] [Indexed: 11/28/2024]
Abstract
The importance of the crosstalk between lung epithelial and immune cells, which emerges from early development and lasts throughout life, is corroborated by a growing body of scientific evidence. This communication not only has a role in driving lung morphogenesis during development, but it is also required in adulthood for the maintenance of homeostasis and repair following infection or injury. Disruption of the intricate immune-epithelial crosstalk can lead to diseases such as COPD and IPF. In this review we summarise the current knowledge regarding the communication between various immune and epithelial cells in development, homeostasis, regeneration and disease, while identifying the current gaps in our knowledge required to facilitate the development of more effective therapies.
Collapse
Affiliation(s)
- Masahiro Yoshida
- UCL Respiratory, Division of Medicine, University College London, London, UK; Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | - Romina Arzili
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Marko Z Nikolić
- UCL Respiratory, Division of Medicine, University College London, London, UK; University College London Hospitals NHS Foundation Trust, London, UK.
| |
Collapse
|
4
|
Bezze A, Mattioda C, Ciardelli G, Mattu C. Harnessing cells to improve transport of nanomedicines. Eur J Pharm Biopharm 2024; 203:114446. [PMID: 39122052 DOI: 10.1016/j.ejpb.2024.114446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Efficient tumour treatment is hampered by the poor selectivity of anticancer drugs, resulting in scarce tumour accumulation and undesired off-target effects. Nano-sized drug-delivery systems in the form of nanoparticles (NPs) have been proposed to improve drug distribution to solid tumours, by virtue of their ability of passive and active tumour targeting. Despite these advantages, literature studies indicated that less than 1% of the administered NPs can successfully reach the tumour mass, highlighting the necessity for more efficient drug transporters in cancer treatment. Living cells, such as blood cells, circulating immune cells, platelets, and stem cells, are often found as an infiltrating component in most solid tumours, because of their ability to naturally circumvent immune recognition, bypass biological barriers, and reach inaccessible tissues through innate tropism and active motility. Therefore, the tumour-homing ability of these cells can be harnessed to design living cell carriers able to improve the transport of drugs and NPs to tumours. Albeit promising, this approach is still in its beginnings and suffers from difficult scalability, high cost, and poor reproducibility. In this review, we present an overview of the most common cell transporters of drugs and NPs, and we discuss how different cell types interact with biological barriers to deliver cargoes of various natures to tumours. Finally, we analyse the different techniques used to load drugs or NPs in living cells and discuss their advantages and disadvantages.
Collapse
Affiliation(s)
- Andrea Bezze
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Carlotta Mattioda
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Gianluca Ciardelli
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Clara Mattu
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
5
|
Miller MR, Landis HE, Miller RE, Tizabi Y. Intercellular Adhesion Molecule 1 (ICAM-1): An Inflammatory Regulator with Potential Implications in Ferroptosis and Parkinson's Disease. Cells 2024; 13:1554. [PMID: 39329738 PMCID: PMC11430830 DOI: 10.3390/cells13181554] [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: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1/CD54), a transmembrane glycoprotein, has been considered as one of the most important adhesion molecules during leukocyte recruitment. It is encoded by the ICAM1 gene and plays a central role in inflammation. Its crucial role in many inflammatory diseases such as ulcerative colitis and rheumatoid arthritis are well established. Given that neuroinflammation, underscored by microglial activation, is a key element in neurodegenerative diseases such as Parkinson's disease (PD), we investigated whether ICAM-1 has a role in this progressive neurological condition and, if so, to elucidate the underpinning mechanisms. Specifically, we were interested in the potential interaction between ICAM-1, glial cells, and ferroptosis, an iron-dependent form of cell death that has recently been implicated in PD. We conclude that there exist direct and indirect (via glial cells and T cells) influences of ICAM-1 on ferroptosis and that further elucidation of these interactions can suggest novel intervention for this devastating disease.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
6
|
Cacho-Navas C, López-Pujante C, Reglero-Real N, Colás-Algora N, Cuervo A, Conesa JJ, Barroso S, de Rivas G, Ciordia S, Paradela A, D'Agostino G, Manzo C, Feito J, Andrés G, Molina-Jiménez F, Majano P, Correas I, Carazo JM, Nourshargh S, Huch M, Millán J. ICAM-1 nanoclusters regulate hepatic epithelial cell polarity by leukocyte adhesion-independent control of apical actomyosin. eLife 2024; 12:RP89261. [PMID: 38597186 PMCID: PMC11006420 DOI: 10.7554/elife.89261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2024] Open
Abstract
Epithelial intercellular adhesion molecule (ICAM)-1 is apically polarized, interacts with, and guides leukocytes across epithelial barriers. Polarized hepatic epithelia organize their apical membrane domain into bile canaliculi and ducts, which are not accessible to circulating immune cells but that nevertheless confine most of ICAM-1. Here, by analyzing ICAM-1_KO human hepatic cells, liver organoids from ICAM-1_KO mice and rescue-of-function experiments, we show that ICAM-1 regulates epithelial apicobasal polarity in a leukocyte adhesion-independent manner. ICAM-1 signals to an actomyosin network at the base of canalicular microvilli, thereby controlling the dynamics and size of bile canalicular-like structures. We identified the scaffolding protein EBP50/NHERF1/SLC9A3R1, which connects membrane proteins with the underlying actin cytoskeleton, in the proximity interactome of ICAM-1. EBP50 and ICAM-1 form nano-scale domains that overlap in microvilli, from which ICAM-1 regulates EBP50 nano-organization. Indeed, EBP50 expression is required for ICAM-1-mediated control of BC morphogenesis and actomyosin. Our findings indicate that ICAM-1 regulates the dynamics of epithelial apical membrane domains beyond its role as a heterotypic cell-cell adhesion molecule and reveal potential therapeutic strategies for preserving epithelial architecture during inflammatory stress.
Collapse
Affiliation(s)
| | | | - Natalia Reglero-Real
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | | | - Ana Cuervo
- Centro Nacional de Biotecnologia (CSIC)MadridSpain
| | | | - Susana Barroso
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | - Gema de Rivas
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | | | | | | | - Carlo Manzo
- Facultat de Ciències, Tecnologia i Enginyeries, Universitat de Vic – Universitat Central de Catalunya (UVic-UCC)VicSpain
| | - Jorge Feito
- Servicio de Anatomía Patológica, Hospital Universitario de SalamancaSalamancaSpain
| | - Germán Andrés
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | - Francisca Molina-Jiménez
- Molecular Biology Unit, Hospital Universitario de la PrincesaMadridSpain
- Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa)MadridSpain
| | - Pedro Majano
- Molecular Biology Unit, Hospital Universitario de la PrincesaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)MadridSpain
- Department of Cellular Biology, Universidad Complutense de MadridMadridSpain
| | - Isabel Correas
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| | | | - Sussan Nourshargh
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Jaime Millán
- Centro de Biologia Molecular Severo Ochoa, CSIC-UAMMadridSpain
| |
Collapse
|
7
|
Wu Q, Luo Y, Lu H, Xie T, Hu Z, Chu Z, Luo F. The Potential Role of Vitamin E and the Mechanism in the Prevention and Treatment of Inflammatory Bowel Disease. Foods 2024; 13:898. [PMID: 38540888 PMCID: PMC10970063 DOI: 10.3390/foods13060898] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/01/2024] [Accepted: 03/12/2024] [Indexed: 01/06/2025] Open
Abstract
Inflammatory bowel disease (IBD) includes ulcerative colitis and Crohn's disease, and it is a multifactorial disease of the intestinal mucosa. Oxidative stress damage and inflammation are major risk factors for IBD. Vitamin E has powerful antioxidant and anti-inflammatory effects. Our previous work and other investigations have shown that vitamin E has a positive effect on the prevention and treatment of IBD. In this paper, the source and structure of vitamin E and the potential mechanism of vitamin E's role in IBD were summarized, and we also analyzed the status of vitamin E deficiency in patients with IBD and the effect of vitamin E supplementation on IBD. The potential mechanisms by which vitamin E plays a role in the prevention and treatment of IBD include improvement of oxidative damage, enhancement of immunity, maintenance of intestinal barrier integrity, and suppression of inflammatory cytokines, modulating the gut microbiota and other relevant factors. The review will improve our understanding of the complex mechanism by which vitamin E inhibits IBD, and it also provides references for doctors in clinical practice and researchers in this field.
Collapse
Affiliation(s)
- Qi Wu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China; (Q.W.); (H.L.); (T.X.); (Z.H.); (Z.C.)
| | - Yi Luo
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Han Lu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China; (Q.W.); (H.L.); (T.X.); (Z.H.); (Z.C.)
| | - Tiantian Xie
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China; (Q.W.); (H.L.); (T.X.); (Z.H.); (Z.C.)
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China; (Q.W.); (H.L.); (T.X.); (Z.H.); (Z.C.)
| | - Zhongxing Chu
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China; (Q.W.); (H.L.); (T.X.); (Z.H.); (Z.C.)
| | - Feijun Luo
- Hunan Key Laboratory of Grain-Oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha 410004, China; (Q.W.); (H.L.); (T.X.); (Z.H.); (Z.C.)
| |
Collapse
|
8
|
Wang Z, Guo Y, Zhang Y, Wu L, Wang L, Lin Q, Wan B. An Intriguing Structural Modification in Neutrophil Migration Across Blood Vessels to Inflammatory Sites: Progress in the Core Mechanisms. Cell Biochem Biophys 2024; 82:67-75. [PMID: 37962751 DOI: 10.1007/s12013-023-01198-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
The role and function of neutrophils are well known, but we still have incomplete understanding of the mechanisms by which neutrophils migrate from blood vessels to inflammatory sites. Neutrophil migration is a complex process that involves several distinct steps. To resist the blood flow and maintain their rolling, neutrophils employ tether and sling formation. They also polarize and form pseudopods and uropods, guided by hierarchical chemotactic agents that enable precise directional movement. Meanwhile, chemotactic agents secreted by neutrophils, such as CXCL1, CXCL8, LTB4, and C5a, can recruit more neutrophils and amplify their response. In the context of diapedesis neutrophils traverse the endothelial cells via two pathways: the transmigratory cup and the lateral border recycling department. These structures aid in overcoming the narrow pore size of the endothelial barrier, resulting in more efficient transmembrane migration. Interestingly, neutrophils exhibit a preference for the paracellular pathway over the transcellular pathway, likely due to the former's lower resistance. In this review, we will delve into the intricate process of neutrophil migration by focusing on critical structures that underpins this process.
Collapse
Affiliation(s)
- Zexu Wang
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Yufang Guo
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Yulei Zhang
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Liangquan Wu
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Li Wang
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Qiuqi Lin
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China
| | - Bing Wan
- Department of Respiratory and Critical Care Medicine, the Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 210002, China.
| |
Collapse
|
9
|
Daei S, Ildarabadi A, Goodarzi S, Mohamadi-Sartang M. Effect of Coenzyme Q10 Supplementation on Vascular Endothelial Function: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. High Blood Press Cardiovasc Prev 2024; 31:113-126. [PMID: 38630421 DOI: 10.1007/s40292-024-00630-8] [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: 09/28/2023] [Accepted: 03/03/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Coenzyme Q10 (CoQ10) has gained attention as a potential therapeutic agent for improving endothelial function. Several randomized clinical trials have investigated CoQ10 supplementation's effect on endothelial function. However, these studies have yielded conflicting results, therefore this systematic review and meta-analysis were conducted. AIM This systematic review and meta-analysis were conducted to assess the effects of CoQ10 supplementation on endothelial factors. METHODS A comprehensive search was done in numerous databases until July 19th, 2023. Quantitative data synthesis was performed using a random-effects model, with weight mean difference (WMD) and 95% confidence intervals (CI). Standard methods were used for the assessment of heterogeneity, meta-regression, sensitivity analysis, and publication bias. RESULTS 12 studies comprising 489 subjects were included in the meta-analysis. The results demonstrated significant increases in Flow Mediated Dilation (FMD) after CoQ10 supplementation (WMD: 1.45; 95% CI: 0.55 to 2.36; p < 0.02), but there is no increase in Vascular cell adhesion protein (VCAM), and Intercellular adhesion molecule (ICAM) following Q10 supplementation (VCAM: SMD: - 0.34; 95% CI: - 0.74 to - 0.06; p < 0.10) (ICAM: SMD: - 0.18; 95% CI: - 0.82 to 0.46; p < 0.57). The sensitivity analysis showed that the effect size was robust in FMD and VCAM. In meta-regression, changes in FMD percent were associated with the dose of supplementation (slope: 0.01; 95% CI: 0.004 to 0.03; p = 0.006). CONCLUSIONS CoQ10 supplementation has a positive effect on FMD in a dose-dependent manner. Our findings show that CoQ10 has an effect on FMD after 8 weeks of consumption. Additional research is warranted to establish the relationship between CoQ10 supplementation and endothelial function.
Collapse
Affiliation(s)
- Shahrzad Daei
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Azam Ildarabadi
- Department of Nutrition, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sima Goodarzi
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Mohsen Mohamadi-Sartang
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Jing X, Qin X, Liu H, Liu H, Wang H, Qin J, Zhang Y, Cao S, Fan X. DNA damage response alterations in clear cell renal cell carcinoma: clinical, molecular, and prognostic implications. Eur J Med Res 2024; 29:107. [PMID: 38326910 PMCID: PMC10848511 DOI: 10.1186/s40001-024-01678-x] [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: 08/11/2023] [Accepted: 09/08/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND DNA damage repair (DDR) pathways modulate cancer risk, progression, and therapeutic responses. Nonetheless, the characteristics and significance of DDR alterations in clear cell renal cell carcinoma (ccRCC) remain undefined. This study aimed to explore the predictive role, molecular mechanism, and tumor immune profile of DDR genes in ccRCC. METHODS We prospectively sequenced 757 tumors and matched blood DNA samples from Chinese patients with ccRCC using next-generation sequencing (NGS) and analyzed data from 537 patients from The Cancer Genome Atlas (TCGA). A comprehensive analysis was performed. RESULTS Fifty-two percent of Chinese patients with ccRCC harbored DDR gene mutations and 57% of TCGA patients. The immunotherapy treatment prognosis of patients with DDR gene mutations was superior to that of patients without DDR gene mutations (p = 0.047). DDR gene mutations were associated with more gene mutations and a higher tumor mutation load (TMB, p < 0.001). Moreover, patients with DDR gene mutations have a distinct mutational signature compared with those with wild-type DDR. Furthermore, the DDR-mut group had elevated neoantigen load (including single-nucleotide variants (SNV) and indel neoantigen load, p = 0.037 and p = 0.002, respectively), TCR Shannon (p = 0.025), and neutrophils (p = 0.010). DDR gene mutations exhibited a distinct immune profile with significantly higher expression levels of TNFSF9, CD70, ICAM1, and indoleamine-2,3-dioxygenase (IDO) and lower expression levels of VTCN1 and IL12A. CONCLUSIONS Our data suggest that the detection of somatic mutations in DDR genes can predict the efficacy of immunotherapy in patients with ccRCC. Furthermore, we revealed the unique molecular and immune mechanisms underlying ccRCC with DDR gene mutations.
Collapse
Affiliation(s)
- Xiao Jing
- Department of Urology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangcheng Qin
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, China
| | - Hao Liu
- Department of Urology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huanhuan Liu
- Acornmed Biotechnology Co., Ltd., Beijing, China
| | - Huina Wang
- Acornmed Biotechnology Co., Ltd., Beijing, China
| | - Jiayue Qin
- Acornmed Biotechnology Co., Ltd., Beijing, China
| | - Yanui Zhang
- Acornmed Biotechnology Co., Ltd., Beijing, China
| | - Shanbo Cao
- Acornmed Biotechnology Co., Ltd., Beijing, China
| | - Xiaodong Fan
- Department of Urology, Ningbo Urology and Nephrology Hospital, Ningbo, China.
| |
Collapse
|
11
|
Ma J, Hermans L, Dierick M, Van der Weken H, Cox E, Devriendt B. Enterotoxigenic Escherichia coli heat labile enterotoxin affects neutrophil effector functions via cAMP/PKA/ERK signaling. Gut Microbes 2024; 16:2399215. [PMID: 39284098 PMCID: PMC11407407 DOI: 10.1080/19490976.2024.2399215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are a major cause of diarrheal illness in humans and animals, induced by enterotoxins produced by these pathogens. Despite the crucial role of neutrophils in combatting bacterial infections, our understanding of how enterotoxins impact neutrophil function is limited. To address this knowledge gap, we used heat-labile enterotoxin (LT) and heat-stable enterotoxin a (STa) to investigate their impact on the effector functions of neutrophils. Our study reveals that pSTa does not exert any discernible effect on the function of neutrophils. In contrast, LT altered the migration and phagocytosis of neutrophils and induced the production of inflammatory factors via activation of cAMP/PKA and ERK1/2 signaling. LT also attenuated the release of neutrophil extracellular traps by neutrophils via the PKA signaling pathway. Our findings provide novel insights into the impact of LT on neutrophil function, shedding light on the underlying mechanisms that govern its immunoregulatory effects. This might help ETEC in subverting the immune system and establishing infection.
Collapse
Affiliation(s)
- Jinglin Ma
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Leen Hermans
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Matthias Dierick
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hans Van der Weken
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bert Devriendt
- Laboratory Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
12
|
Robinson E, Herbert JA, Palor M, Ren L, Larken I, Patel A, Moulding D, Cortina-Borja M, Smyth RL, Smith CM. Trans-epithelial migration is essential for neutrophil activation during RSV infection. J Leukoc Biol 2023; 113:354-364. [PMID: 36807711 PMCID: PMC11334017 DOI: 10.1093/jleuko/qiad011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 02/04/2023] Open
Abstract
The recruitment of neutrophils to the infected airway occurs early following respiratory syncytial virus (RSV) infection, and high numbers of activated neutrophils in the airway and blood are associated with the development of severe disease. The aim of this study was to investigate whether trans-epithelial migration is sufficient and necessary for neutrophil activation during RSV infection. Here, we used flow cytometry and novel live-cell fluorescent microscopy to track neutrophil movement during trans-epithelial migration and measure the expression of key activation markers in a human model of RSV infection. We found that when migration occurred, neutrophil expression of CD11b, CD62L, CD64, NE, and MPO increased. However, the same increase did not occur on basolateral neutrophils when neutrophils were prevented from migrating, suggesting that activated neutrophils reverse migrate from the airway to the bloodstream side, as has been suggested by clinical observations. We then combined our findings with the temporal and spatial profiling and suggest 3 initial phases of neutrophil recruitment and behavior in the airways during RSV infection; (1) initial chemotaxis; (2) neutrophil activation and reverse migration; and (3) amplified chemotaxis and clustering, all of which occur within 20 min. This work and the novel outputs could be used to develop therapeutics and provide new insight into how neutrophil activation and a dysregulated neutrophil response to RSV mediates disease severity.
Collapse
Affiliation(s)
- Elisabeth Robinson
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Jenny Amanda Herbert
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
- School of Medical Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Rd, Manchester M13 9PL, United Kingdom
| | - Machaela Palor
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Luo Ren
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
- Department of Respiratory Medicine, Children's Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Isobel Larken
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Alisha Patel
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Dale Moulding
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Mario Cortina-Borja
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Rosalind Louise Smyth
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| | - Claire Mary Smith
- Infection, Immunity and Inflammation Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N1EH, United Kingdom
| |
Collapse
|
13
|
Du Y, Yan B. Ocular immune privilege and retinal pigment epithelial cells. J Leukoc Biol 2023; 113:288-304. [PMID: 36805720 DOI: 10.1093/jleuko/qiac016] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Indexed: 02/04/2023] Open
Abstract
The ocular tissue microenvironment is immune-privileged and uses multiple immunosuppressive mechanisms to prevent the induction of inflammation. The retinal pigment epithelium plays an essential role in ocular immune privilege. In addition to serving as a blood barrier separating the fenestrated choriocapillaris from the retina, the retinal pigment epithelium is a source of immunosuppressive cytokines and membrane-bound negative regulators that modulate the activity of immune cells within the retina. This article reviews the current understanding of how retinal pigment epithelium cells mediate immune regulation, focusing on the changes under pathologic conditions.
Collapse
Affiliation(s)
- Yuxiang Du
- Institute of Precision Medicine, Jining Medical University, No. 133, Hehua Road, Taibaihu New District, Jining, Shandong 272067, People's Republic of China
| | - Bo Yan
- Institute of Precision Medicine, Jining Medical University, No. 133, Hehua Road, Taibaihu New District, Jining, Shandong 272067, People's Republic of China
| |
Collapse
|
14
|
Liang Z, Hu X, Lin R, Tang Z, Ye Z, Mao R, Chen W, Zhou Y. Identification of shared gene signatures and molecular mechanisms between chronic kidney disease and ulcerative colitis. Front Immunol 2023; 14:1078310. [PMID: 36860851 PMCID: PMC9970095 DOI: 10.3389/fimmu.2023.1078310] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Background There is a complex interaction between chronic kidney disease (CKD) and ulcerative colitis (UC), but the pathophysiological mechanisms underlying the coexistence of CKD and UC are unclear. This study aimed to investigate the key molecules and pathways that may mediate the co-occurrence of CKD and UC through quantitative bioinformatics analysis based on a public RNA-sequencing database. Methods The discovery datasets of CKD (GSE66494) and UC (GSE4183), as well as validation datasets of CKD (GSE115857) and UC (GSE10616), were downloaded from the Gene Expression Omnibus (GEO) database. After identifying differentially expressed genes (DEGs) with GEO2R online tool, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses for the DEGs were performed. Next, protein-protein interaction network was constructed with Search Tool for the Retrieval of Interacting Genes (STRING) and visualized by Cytoscape. Gene modules were identified by the plug-in MCODE and hub genes were screened using the plug-in CytoHubba. Then, correlation between immune cell infiltration and hub genes was analyzed, and the receiver operating characteristic curves were used to assess the predictive value of hub genes. Finally, immunostaining of human specimens was used to validate the relevant findings. Results A total of 462 common DEGs were identified and selected for further analyses. GO and KEGG enrichment analyses indicated that these DEGs were primarily enriched in immune- and inflammation-related pathways. Among them, the PI3K-Akt signaling pathway ranked top in both discovery and validation cohorts, and the key signal molecule phosphorylated Akt (p-Akt) was shown to be significantly overexpressed in human CKD kidneys and UC colons, and further elevated in CKD-UC comorbidity specimens. Moreover, nine candidate hub genes, including CXCL8, CCL2, CD44, ICAM1, IL1A, CXCR2, PTPRC, ITGAX, and CSF3, were identified, of which ICAM1 was validated as a common hub gene. Besides, immune infiltration analysis revealed that neutrophils, macrophages, and CD4+ T memory cells significantly accumulated in both diseases, and ICAM1 was remarkably associated with neutrophil infiltration. Furthermore, intercellular adhesion molecule1 (ICAM1)-mediated neutrophil infiltration was validated to be upregulated in kidney and colon biopsies of CKD and UC patients, and further increased in patients diagnosed with both CKD and UC. Finally, ICAM1 had shown critical value as a diagnostic marker for the co-occurrence of CKD and UC. Conclusions Our study elucidated that immune response, PI3K-Akt signaling pathway, and ICAM1-mediated neutrophil infiltration might be the common pathogenesis of CKD and UC, and identified ICAM1 as a key potential biomarker and therapeutic target for the comorbidity of these two diseases.
Collapse
Affiliation(s)
- Zhou Liang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC), Key Laboratory of Clinical Nephrology (SunYat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Xinrong Hu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC), Key Laboratory of Clinical Nephrology (SunYat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ruoni Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC), Key Laboratory of Clinical Nephrology (SunYat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ziwen Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC), Key Laboratory of Clinical Nephrology (SunYat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ziyin Ye
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ren Mao
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC), Key Laboratory of Clinical Nephrology (SunYat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC), Key Laboratory of Clinical Nephrology (SunYat-Sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
15
|
Xu Y, Li Q, Ge P, Mao H, Yang C. Chitosan nanoparticles attenuate intestinal damage and inflammatory responses in LPS-challenged weaned piglets via prevention of IκB degradation. J Anim Physiol Anim Nutr (Berl) 2023; 107:173-181. [PMID: 34820921 DOI: 10.1111/jpn.13664] [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: 02/08/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 01/10/2023]
Abstract
Chitosan nanoparticles (CNP), widely applied as oral drug/gene/vaccine carrier, were found to have anti-inflammatory properties. In this study, the effects of CNP on lipopolysaccharide (LPS)-induced intestinal damage in weaned piglets and the related mechanisms were investigated. Twenty-four weaned piglets (Duroc × Landrace × Yorkshire, 21 ± 2 day of age, initial mass: 8.58 ± 0.59 kg) were randomly assigned into four groups: control, LPS, CNP and CNP + LPS. The control and LPS groups were fed a corn-soybean meal-based control diet, whereas the CNP and CNP + LPS groups were fed a control diet supplemented with 400 mg/kg CNP. After 28 days of feeding, piglets in LPS and CNP + LPS groups were injected with LPS (100 μg/kg); meanwhile, the piglets in control and CNP groups were injected with sterile saline. After 4 h from the LPS challenge, pigs were sacrificed to collect the intestinal samples for analysis. The results showed that CNP could attenuate the intestinal damages and inflammatory response stimulated by LPS treatment. LPS induced dramatically higher levels of CD177+ neutrophils invasion in jejunum mucosa (p < 0.01), which accompanied by increased secretion of marks of inflammation (p < 0.01) compared with the control, whereas CNP administration obviously inhibited LPS-induced CD177+ neutrophils invasion (p < 0.01) and secretion of marks of inflammation, such as interleukin-8 (p < 0.05), intercellular adhesion molecule-1 (p < 0.05) secretion in jejunum mucosa compared with LPS group. Moreover, CNP was shown to inhibit IκB-α degradation in cytoplasm, which resulted in reduced nuclear translocation of NF-κB p65 in LPS-challenged piglets. These findings suggest that CNP attenuates intestinal damage and inflammatory responses in LPS-challenged weaned piglets by impairing the NF-κB signalling pathway.
Collapse
Affiliation(s)
- Yinglei Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology • College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Qing Li
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology • College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Pu Ge
- Pathophysiology Department of ChongQing Medical University, Chongqing, China
| | - Huiling Mao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology • College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| | - Caimei Yang
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, College of Animal Science and Technology • College of Veterinary Medicine, Zhejiang A&F University, Hangzhou, China
| |
Collapse
|
16
|
Azcutia V, Kelm M, Kim S, Luissint AC, Flemming S, Abernathy-Close L, Young VB, Nusrat A, Miller MJ, Parkos CA. Distinct stimulus-dependent neutrophil dynamics revealed by real-time imaging of intestinal mucosa after acute injury. PNAS NEXUS 2022; 1:pgac249. [PMID: 36712325 PMCID: PMC9802210 DOI: 10.1093/pnasnexus/pgac249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Clinical symptoms in many inflammatory diseases of the intestine are directly related to neutrophil (PMN) migration across colonic mucosa and into the intestinal lumen, yet in-vivo studies detailing this process are lacking. Using real-time intravital microscopy and a new distal colon loop model, we report distinct PMN migratory dynamics in response to several models of acute colonic injury. PMNs exhibited rapid swarming responses after mechanically induced intestinal wounds. Similar numbers of PMNs infiltrated colonic mucosa after wounding in germ-free mice, suggesting microbiota-independent mechanisms. By contrast, acute mucosal injury secondary to either a treatment of mice with dextran sodium sulfate or an IL-10 receptor blockade model of colitis resulted in lamina propria infiltration with PMNs that were largely immotile. Biopsy wounding of colonic mucosa in DSS-treated mice did not result in enhanced PMN swarming however, intraluminal application of the neutrophil chemoattractant LTB4 under such conditions resulted in enhanced transepithelial migration of PMNs. Analyses of PMNs that had migrated into the colonic lumen revealed that the majority of PMNs were directly recruited from the circulation and not from the immotile pool in the mucosa. Decreased PMN motility parallels upregulation of the receptor CXCR4 and apoptosis. Similarly, increased expression of CXCR4 on human PMNs was observed in colonic biopsies from people with active ulcerative colitis. This new approach adds an important tool to investigate mechanisms regulating PMN migration across mucosa within the distal intestine and will provide new insights for developing future anti-inflammatory and pro-repair therapies.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, University of Michigan; Ann Arbor, MI 48109, USA
| | - Matthias Kelm
- Department of Pathology, University of Michigan; Ann Arbor, MI 48109, USA
| | - Seonyoung Kim
- Department of Internal Medicine, Washington University School of Medicine; Saint Louis, MO 63110, USA
| | | | - Sven Flemming
- Department of Pathology, University of Michigan; Ann Arbor, MI 48109, USA
| | - Lisa Abernathy-Close
- Department of Internal Medicine/Division of Infectious Diseases, University of Michigan; Ann Arbor, MI 48109, USA
| | - Vincent B Young
- Department of Internal Medicine/Division of Infectious Diseases, University of Michigan; Ann Arbor, MI 48109, USA
- Department of Microbiology and Immunology, University of Michigan; Ann Arbor, MI 48109, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan; Ann Arbor, MI 48109, USA
| | - Mark J Miller
- Department of Internal Medicine, Washington University School of Medicine; Saint Louis, MO 63110, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan; Ann Arbor, MI 48109, USA
| |
Collapse
|
17
|
Evaluation of Blood Intercellular Adhesion Molecule-1 (ICAM-1) Level in Obstructive Sleep Apnea: A Systematic Review and Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101499. [PMID: 36295659 PMCID: PMC9607021 DOI: 10.3390/medicina58101499] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 12/04/2022]
Abstract
Background and objective: Intercellular adhesion molecule-1 (ICAM-1) appears to be an active and important biomarker for decreasing the risk of cardiovascular issues among individuals with obstructive sleep apnea (OSA). Herein, a systematic review and meta-analysis was designed to probe whether plasma/serum ICAM-1levels are different in adults with OSA compared to adults with no OSA, as well as adults with severe OSA compared to adults with mild/moderate OSA. Materials and methods: A thorough and systematic literature search was performed in four databases (PubMed/Medline, Web of Science, Scopus, and Cochrane Library) until 17 July 2022, without any age and sample size restrictions to retrieve the relevant articles. The standardized mean difference (SMD) along with a 95% confidence interval (CI) of plasma/serum of ICAM-1 levels was reported. Analyses, including sensitivity analysis, subgroup analysis, trial sequential analysis, meta-regression, and a funnel plot analysis, were performed in the pooled analysis. Results: A total of 414 records were identified in the databases, and 17 articles including 22 studies were entered into the meta-analysis. The pooled SMD of serum/plasma ICAM-1 levels in adults with OSA compared to controls was 2.00 (95%CI: 1.41, 2.59; p < 0.00001). The pooled SMD of serum/plasma ICAM-1 levels in adults with severe compared to mild/moderate OSA was 3.62 (95%CI: 1.74, 5.51; p = 0.0002). Higher serum/plasma ICAM-1 levels were associated with a higher mean age of controls, higher scores for the apnea-hypopnea index, and with a lower mean age of adults with OSA and with smaller sample sizes. Conclusions: Th results of the present meta-analysis showed that serum/plasma ICAM-1 levels in adults with OSA was higher than serum/plasma ICAM-1 levels in controls. Similarly, serum/plasma ICAM-1 levels in adults with severe OSA were higher compared to serum/plasma ICAM-1 levels of adults with mild or moderate OSA. Therefore, ICAM-1 may be used as an additional diagnostic and therapeutic biomarker in adults with OSA.
Collapse
|
18
|
Manzanares LD, David J, Ren X, Yalom LK, Piccolo EB, Dehghan Y, David AJ, Hanauer SB, Sumagin R. Atovaquone attenuates experimental colitis by reducing neutrophil infiltration of colonic mucosa. Front Pharmacol 2022; 13:1011115. [PMID: 36313299 PMCID: PMC9614091 DOI: 10.3389/fphar.2022.1011115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2022] [Indexed: 10/08/2023] Open
Abstract
Ulcerative colitis (UC) is a chronic relapsing disease featuring aberrant accumulation of neutrophils in colonic mucosa and the luminal space. Although significant advances in UC therapy have been made with the development of novel biologics and small molecules targeting immune responses, success of most current therapies is still limited, with significant safety concerns. Thus, there is a need to develop additional safe and effective therapies for the treatment of UC. Antimalarial drugs have been safely used for many years to resolve tissue inflammation and the associated pathologies. Atovaquone is a recent FDA-approved antimalarial drug that has shown anti-viral and tumor-suppressive properties in vitro however, its role in mucosal inflammation has not been evaluated. Using pre-clinical murine DSS-induced colitis model combined with complementary in vivo peritonitis and ex vivo human neutrophil activation and chemotaxis assays we investigated functional and mechanistic impacts of atovaquone on disease resolution and neutrophil trafficking. We demonstrate that atovaquone promotes resolution of DSS-induced murine colitis by reducing neutrophil accumulation in the inflamed colonic mucosa. Mechanistically, we show that atovaquone suppressed induction of CD11b expression in neutrophils, reducing their polarization and migratory ability. Thus, our findings identify a new role of atovaquone in promoting resolution of mucosal inflammation, supporting the idea of potential repurposing of this FDA-approved drug as UC therapeutic.
Collapse
Affiliation(s)
- Laura D. Manzanares
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph David
- Department of Medicine, Gastroenterology and Hepatology University of Arizona College of Medicine, Phoenix, AZ, United States
| | - Xingsheng Ren
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lenore K. Yalom
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Enzo B. Piccolo
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Yalda Dehghan
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Aidan J. David
- College of Arts and Sciences, Case Western Reserve Unviersity, Cleveland, OH, United States
| | - Stephen B. Hanauer
- Department of Medicine, Gastroenterology and Hepatology Northwestern Memorial Hospital, Chicago, IL, United States
| | - Ronen Sumagin
- Laboratory 7-065 Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
19
|
Sequestration of gut pathobionts in intraluminal casts, a mechanism to avoid dysregulated T cell activation by pathobionts. Proc Natl Acad Sci U S A 2022; 119:e2209624119. [PMID: 36201539 PMCID: PMC9565271 DOI: 10.1073/pnas.2209624119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T cells that express the transcription factor RORγ, regulatory (Treg), or conventional (Th17) are strongly influenced by intestinal symbionts. In a genetic approach to identify mechanisms underlying this influence, we performed a screen for microbial genes implicated, in germfree mice monocolonized with Escherichia coli Nissle. The loss of capsule-synthesis genes impaired clonal expansion and differentiation of intestinal RORγ+ T cells. Mechanistic exploration revealed that the capsule-less mutants remained able to induce species-specific immunoglobulin A (IgA) and were highly IgA-coated. They could still trigger myeloid cells, and more effectively damaged epithelial cells in vitro. Unlike wild-type microbes, capsule-less mutants were mostly engulfed in intraluminal casts, large agglomerates composed of myeloid cells extravasated into the gut lumen. We speculate that sequestration in luminal casts of potentially harmful microbes, favored by IgA binding, reduces the immune system's actual exposure, preserving host-microbe equilibrium. The variable immunostimulation by microbes that has been charted in recent years may not solely be conditioned by triggering molecules or metabolites but also by physical limits to immune system exposure.
Collapse
|
20
|
Yu L, Lin W, Shen C, Meng T, Jin P, Ding X, Eggenhuizen PJ, Ooi JD, Tang R, Nie W, Li X, Xiao X, Zhong Y. Intrarenal Single-Cell Sequencing of Hepatitis B Virus Associated Membranous Nephropathy. Front Med (Lausanne) 2022; 9:869284. [PMID: 35935760 PMCID: PMC9355751 DOI: 10.3389/fmed.2022.869284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Abstract
To date, the pathogenesis of hepatitis B virus (HBV)-associated membranous nephropathy (MN) remains elusive. This study aimed to decipher the etiopathogenesis of HBV-associated MN by performing single-cell RNA sequencing (scRNA-seq) of kidney biopsy specimens from a patient with HBV-associated MN and two healthy individuals. We generated 4,114 intrarenal single-cell transcriptomes from the HBV-associated MN patient by scRNA-seq. Compared to healthy individuals, podocytes in the HBV-associated MN patient showed an increased expression of extracellular matrix formation-related genes, including HSPA5, CTGF, and EDIL3. Kidney endothelial cells (ECs) in the HBV-associated MN were enriched in inflammatory pathways, including NF-kappa B signaling, IL-17 signaling, TNF signaling and NOD-like receptor signaling. Gene ontology (GO) functional enrichment analysis and Gene Set Variation Analysis (GSVA) further revealed that differentially expressed genes (DEGs) of ECs from the HBV-associated MN patients were enriched in apoptotic signaling pathway, response to cytokine and leukocyte cell-cell adhesion. The up-regulated DEGs in glomerular ECs of HBV-associated MN patients were involved in biological processes such as viral gene expression, and protein targeting to endoplasmic reticulum. We further verified that the overexpressed genes in ECs from HBV-associated MN were mainly enriched in regulation of protein targeting to endoplasmic reticulum, exocytosis, viral gene expression, IL-6 and IL-1 secretion when compared with anti-phospholipase A2 receptor (PLA2R)-positive idiopathic membranous nephropathy (IMN). The receptor-ligand crosstalk analysis revealed potential interactions between endothelial cells and other cells in HBV-associated-MN. These results offer new insight into the pathogenesis of HBV-associated MN and may identify new therapeutic targets for HBV-associated MN.
Collapse
Affiliation(s)
- Leilin Yu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- Jiujiang Traditional Medicine Hospital, Jiujiang, China
| | - Wei Lin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, China
| | - Chanjuan Shen
- Department of Hematology, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Changsha, China
| | - Ting Meng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Jin
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, China
| | - Xiang Ding
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, China
| | | | - Joshua D. Ooi
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Rong Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Wannian Nie
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xia Li
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, China
- Xiangcheng Xiao
| | - Yong Zhong
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yong Zhong
| |
Collapse
|
21
|
Epithelial and Neutrophil Interactions and Coordinated Response to Shigella in a Human Intestinal Enteroid-Neutrophil Coculture Model. mBio 2022; 13:e0094422. [PMID: 35652591 PMCID: PMC9239269 DOI: 10.1128/mbio.00944-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Polymorphonuclear neutrophils (PMN) are recruited to the gastrointestinal mucosa in response to inflammation, injury, and infection. Here, we report the development and the characterization of an ex vivo tissue coculture model consisting of human primary intestinal enteroid monolayers and PMN, and a mechanistic interrogation of PMN-epithelial cell interaction and response to Shigella, a primary cause of childhood dysentery. Cellular adaptation and tissue integration, barrier function, PMN phenotypic and functional attributes, and innate immune responses were examined. PMN within the enteroid monolayers acquired a distinct activated/migratory phenotype that was influenced by direct epithelial cell contact as well as by molecular signals. Seeded on the basal side of the intestinal monolayer, PMN were intercalated within the epithelial cells and moved paracellularly toward the apical side. Cocultured PMN also increased basal secretion of interleukin 8 (IL-8). Shigella added to the apical surface of the monolayers evoked additional PMN phenotypic adaptations, including increased expression of cell surface markers associated with chemotaxis and cell degranulation (CD47, CD66b, and CD88). Apical Shigella infection triggered rapid transmigration of PMN to the luminal side, neutrophil extracellular trap (NET) formation, and bacterial phagocytosis and killing. Shigella infection modulated cytokine production in the coculture; apical monocyte chemoattractant protein (MCP-1), tumor necrosis factor alpha (TNF-α), and basolateral IL-8 production were downregulated, while basolateral IL-6 secretion was increased. We demonstrated, for the first time, PMN phenotypic adaptation and mobilization and coordinated epithelial cell-PMN innate response upon Shigella infection in the human intestinal environment. The enteroid monolayer-PMN coculture represents a technical innovation for mechanistic interrogation of gastrointestinal physiology, host-microbe interaction, innate immunity, and evaluation of preventive/therapeutic tools.
Collapse
|
22
|
Pandurangan AK, Mohebali N, Hasanpourghadi M, Esa NM. Caffeic Acid Phenethyl Ester Attenuates Dextran Sulfate Sodium-Induced Ulcerative Colitis Through Modulation of NF-κB and Cell Adhesion Molecules. Appl Biochem Biotechnol 2022; 194:1091-1104. [PMID: 35040047 DOI: 10.1007/s12010-021-03788-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 12/28/2022]
Abstract
Ulcerative colitis (UC) is a serious health condition and defined as inflammation in the colon. Untreated, UC can develop into colitis-associated cancer (CAC), for which effective medicines are not available. Natural products are a better choice to treat UC by alleviating the inflammation. Caffeic acid phenethyl ester (CAPE) is a phenolic compound and known for its beneficial effects, including antibacterial, anti-inflammatory, anti-diabetic, and anticancer. We aimed to study the effect of CAPE on dextran sulfate sodium (DSS)-induced UC in mouse model. Administration of CAPE to DSS-induced mice protected against colon damage by improving body weight of mice, reducing the weight of spleen, and increased colon length. In addition, administration of CAPE resulted reduced the activity of myeloperoxidase (MPO) and CD68+ positive cells. Furthermore, a significant decrease in the production of key cytokines and the expression of nuclear factor (p65-NF)-κB. Moreover, p65-NF-κB activation was reduced in lipopolysaccharide (LPS)-treated RAW 264.7 macrophage cells from mouse origin. CAPE treatment leads to the reduced expressions of intercellular adhesion molecules (ICAM)-1 and vascular cell adhesion molecules (VCAM), both are key cell adhesion molecules. The results of this study clearly indicate that CAPE can potentially control inflammation in the colon and can be used as a therapy for UC.
Collapse
Affiliation(s)
- Ashok Kumar Pandurangan
- School of Life Sciences, B.S. Abdur Rahman Crescent Institute of Science and Technology, GST road, Vandalur, Chennai, Tamil Nadu, 600048, India.
| | - Nooshin Mohebali
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Mohadeseh Hasanpourghadi
- Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, 43400, Malaysia
| | - Norhaizan Mohd Esa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| |
Collapse
|
23
|
Xian Y, Da P, Chao Y, Hui X, Ligang Y, Shaokang W, Guiju S. Wheat oligopeptides enhance the intestinal mucosal barrier and alleviate inflammation via the TLR4/Myd88/MAPK signaling pathway in aged mice. Food Nutr Res 2022; 66:5690. [PMID: 35261579 PMCID: PMC8861859 DOI: 10.29219/fnr.v66.5690] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/04/2021] [Accepted: 11/30/2021] [Indexed: 11/24/2022] Open
Abstract
Background Aging can induce oxidative stress, inflammation and mucosal impairment, and few works have been conducted to investigate the protective effects of WP on the natural intestinal aging process. Objective The present work aimed to examine the protective effect of wheat oligopeptides (WP) on intestine mucosal impairment in aged mice, and investigate the potential antioxidation, anti-inflammatory effects of WP. Design Seventy-two aged mice (24 months old) were randomly divided into six groups, 12 for each group. Twelve young mice (6 months old) were regarded as the young control group. WP (25, 50, 100, 200, or 400 mg/kg) or distilled water were administered daily by gavage for 30 days. Results Histological observations showed that intestinal mucosal degeneration was attenuated by WP pretreatment. WP exhibited remarkable antioxidant activity via increasing superoxide dismutase, glutathione peroxidase, total antioxidant capacity and catalase activities, and decreasing the malondialdehyde levels in small intestine mucosa. WP pretreatment significantly suppressed intestinal mucosa inflammation through the reduction of TNF-α, TGF-β, IFN-γ IL-1β and IL-6. WP markedly protect the intestinal mucosal barrier by decreasing the ICAM-1 level, and increasing ZO-1 and JAMA-A levels. WP significantly down-regulated protein expression levels of TLR4, Myd88, and MAPK, suggesting that WP have a potential effect on inhibiting aging-induced inflammatory responses by blocking TLR4/Myd88/MAPK signal transduction. Conclusion WP administration effectively alleviated intestinal mucosal impairment in aged mice. The potential mechanism was associated with enhancement of antioxidation and anti-inflammatory action and protection of the intestinal mucosal barrier.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Sun Guiju
- Sun Guiju, Key Laboratory of Environmental Medicine and Engineering, Ministry of Education/Department of Nutrition and Food, Hygiene, School of Public Health, Southeast University, Nanjing, P.R. China.
| |
Collapse
|
24
|
Plasmolipin regulates basolateral-to-apical transcytosis of ICAM-1 and leukocyte adhesion in polarized hepatic epithelial cells. Cell Mol Life Sci 2022; 79:61. [PMID: 34999972 PMCID: PMC8743267 DOI: 10.1007/s00018-021-04095-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/30/2022]
Abstract
Apical localization of Intercellular Adhesion Receptor (ICAM)-1 regulates the adhesion and guidance of leukocytes across polarized epithelial barriers. Here, we investigate the molecular mechanisms that determine ICAM-1 localization into apical membrane domains of polarized hepatic epithelial cells, and their effect on lymphocyte-hepatic epithelial cell interaction. We had previously shown that segregation of ICAM-1 into apical membrane domains, which form bile canaliculi and bile ducts in hepatic epithelial cells, requires basolateral-to-apical transcytosis. Searching for protein machinery potentially involved in ICAM-1 polarization we found that the SNARE-associated protein plasmolipin (PLLP) is expressed in the subapical compartment of hepatic epithelial cells in vitro and in vivo. BioID analysis of ICAM-1 revealed proximal interaction between this adhesion receptor and PLLP. ICAM-1 colocalized and interacted with PLLP during the transcytosis of the receptor. PLLP gene editing and silencing increased the basolateral localization and reduced the apical confinement of ICAM-1 without affecting apicobasal polarity of hepatic epithelial cells, indicating that ICAM-1 transcytosis is specifically impaired in the absence of PLLP. Importantly, PLLP depletion was sufficient to increase T-cell adhesion to hepatic epithelial cells. Such an increase depended on the epithelial cell polarity and ICAM-1 expression, showing that the epithelial transcytotic machinery regulates the adhesion of lymphocytes to polarized epithelial cells. Our findings strongly suggest that the polarized intracellular transport of adhesion receptors constitutes a new regulatory layer of the epithelial inflammatory response.
Collapse
|
25
|
Crifo B, MacNaughton WK. Cells and mediators of inflammation as effectors of epithelial repair in the inflamed intestine. Am J Physiol Gastrointest Liver Physiol 2022; 322:G169-G182. [PMID: 34878937 DOI: 10.1152/ajpgi.00194.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mucosal and histological healing have become the gold standards for assessing the efficacy of therapy in patients living with inflammatory bowel diseases (IBD). Despite these being the accepted goals in therapy, the mechanisms that underlie the healing of the mucosa after an inflammatory insult are not well understood, and many patients fail to meet this therapeutic endpoint. Here we review the emerging evidence that mediators (e.g., prostaglandins, cytokines, proteases, reactive oxygen, and nitrogen species) and innate immune cells (e.g., neutrophils and monocytes/macrophages), that are involved in the initiation of the inflammatory response, are also key players in the mechanisms underlying mucosal healing to resolve chronic inflammation in the colon. The dual function mediators comprise an inflammation/repair program that returns damaged tissue to homeostasis. Understanding details of the dual mechanisms of these mediators and cells may provide the basis for the development of drugs that can help to stimulate epithelial repair in patients affected by IBD.
Collapse
Affiliation(s)
- Bianca Crifo
- Department of Physiology and Pharmacology, Inflammation Research Network and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Department of Physiology and Pharmacology, Inflammation Research Network and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
26
|
Parkos CA. LEUKOCYTE-EPITHELIAL INTERACTIONS: A DOUBLE-EDGED SWORD THAT PROTECTS AND INJURES DURING HEALTH AND DISEASE. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2022; 132:22-33. [PMID: 36196189 PMCID: PMC9480551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Neutrophils (PMNs) play a critical role in innate immunity, yet many pathologic conditions are associated with dysregulated infiltration of PMNs into tissues. In the gut, robust PMN accumulation and migration across the intestinal epithelium closely correlates with clinical symptoms in conditions such as ulcerative colitis. While much is known about how PMNs migrate out of blood vessels, far less is understood about how PMNs traverse epithelial barriers. Until fairly recently, in vitro models of PMN transepithelial migration (TEpM) across cultured intestinal epithelial cell lines provided many of the insights into the molecular basis of TEpM. However, innovative animal models have provided new avenues for investigating in vivo mechanisms regulating PMN TEpM. This report will highlight molecular insights gained from studies on PMN TEpM and provide a rationale for developing tissue targeted strategies directed at reducing pathologic consequences of dysregulated PMN trafficking in the gut.
Collapse
|
27
|
Li T, Liu H, Jiang N, Wang Y, Wang Y, Zhang J, Shen Y, Cao J. Comparative proteomics reveals Cryptosporidium parvum manipulation of the host cell molecular expression and immune response. PLoS Negl Trop Dis 2021; 15:e0009949. [PMID: 34818332 PMCID: PMC8612570 DOI: 10.1371/journal.pntd.0009949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/25/2021] [Indexed: 02/01/2023] Open
Abstract
Cryptosporidium is a life-threating protozoan parasite belonging to the phylum Apicomplexa, which mainly causes gastroenteritis in a variety of vertebrate hosts. Currently, there is a re-emergence of Cryptosporidium infection; however, no fully effective drug or vaccine is available to treat Cryptosporidiosis. In the present study, to better understand the detailed interaction between the host and Cryptosporidium parvum, a large-scale label-free proteomics study was conducted to characterize the changes to the proteome induced by C. parvum infection. Among 4406 proteins identified, 121 proteins were identified as differentially abundant (> 1.5-fold cutoff, P < 0.05) in C. parvum infected HCT-8 cells compared with uninfected cells. Among them, 67 proteins were upregulated, and 54 proteins were downregulated at 36 h post infection. Analysis of the differentially abundant proteins revealed an interferon-centered immune response of the host cells against C. parvum infection and extensive inhibition of metabolism-related enzymes in the host cells caused by infection. Several proteins were further verified using quantitative real-time reverse transcription polymerase chain reaction and western blotting. This systematic analysis of the proteomics of C. parvum-infected HCT-8 cells identified a wide range of functional proteins that participate in host anti-parasite immunity or act as potential targets during infection, providing new insights into the molecular mechanism of C. parvum infection. Cryptosporidium parvum is an emerging zoonotic pathogen transmitted via the fecal–oral route, and is considered a leading cause of moderate-to-severe diarrheal disease in young children in resource limited areas. After infection, C. parvum parasitizes intestinal epithelial cells and evokes an inflammatory immune response, leading to severe damage of the intestinal mucosa. The infection can be lethal to immunosuppressed individuals. However, no fully effective drug or vaccine is available for cryptosporidiosis, and the pathogenesis and immune mechanisms during C. parvum infection are obscure. Thus, an in-depth understanding of host-parasite interaction is needed. Hence, we established a C. parvum-infected HCT-8 cell model and performed comparative quantitative proteomic analyses to profile global host-parasite interactions and determine the molecular mechanisms that are activated during infection, aiming to offer new insights into the treatment of Cryptosporidium.
Collapse
Affiliation(s)
- Teng Li
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
| | - Nan Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
| | - Yiluo Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
| | - Ying Wang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
| | - Jing Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
| | - Yujuan Shen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (YS); (JC)
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; WHO Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (YS); (JC)
| |
Collapse
|
28
|
Heimroth RD, Casadei E, Benedicenti O, Amemiya CT, Muñoz P, Salinas I. The lungfish cocoon is a living tissue with antimicrobial functions. SCIENCE ADVANCES 2021; 7:eabj0829. [PMID: 34788085 PMCID: PMC8597997 DOI: 10.1126/sciadv.abj0829] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
Terrestrialization is an extreme physiological adaptation by which African lungfish survive dry seasons. For months and up to several years, lungfish live inside a dry mucus cocoon that protects them from desiccation. Light and electron microscopy reveal that the lungfish cocoon is a living tissue that traps bacteria. Transcriptomic analyses identify a global state of inflammation in the terrestrialized lungfish skin characterized by granulocyte recruitment. Recruited granulocytes transmigrate into the cocoon where they release extracellular traps. In vivo DNase I surface spraying during terrestrialization results in dysbiosis, septicemia, skin wounds, and hemorrhages. Thus, lungfish have evolved unique immunological adaptations to protect their bodies from infection for extended periods of time while living on land. Trapping bacteria outside their bodies may benefit estivating vertebrates that undergo metabolic torpor.
Collapse
Affiliation(s)
- Ryan Darby Heimroth
- Center for Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM, USA
| | - Elisa Casadei
- Center for Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM, USA
| | - Ottavia Benedicenti
- Center for Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM, USA
| | - Chris Tsuyoshi Amemiya
- Department of Molecular and Cell Biology, University of California, Merced, Merced, CA, USA
| | - Pilar Muñoz
- Department of Animal Health, Campus de Espinardo, University of Murcia, 30100 Murcia, Spain
| | - Irene Salinas
- Center for Evolutionary and Theoretical Immunology, Biology Department, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
29
|
Sun JN, Yu XY, Hou B, Ai M, Qi MT, Ma XY, Cai MJ, Gao M, Cai WW, Ni LL, Xu F, Zhou YT, Qiu LY. Vaccarin enhances intestinal barrier function in type 2 diabetic mice. Eur J Pharmacol 2021; 908:174375. [PMID: 34303666 DOI: 10.1016/j.ejphar.2021.174375] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/16/2021] [Accepted: 07/21/2021] [Indexed: 01/12/2023]
Abstract
AIMS Hyperglycemia and insulin resistance drive intestinal barrier dysfunction in type 2 diabetes (T2DM). Vaccarin, the main active component in the semen of traditional Chinese medicine Vaccaria has a definite effect on T2DM mice. The purpose of this study was to investigate whether vaccarin can enhance the intestinal barrier function in T2DM. MAIN METHODS The T2DM mice model was established by streptozocin and high-fat diet. Vaccarin at a dose of 1 mg/kg/day was administered. We evaluated the effects of vaccarin on gut microbiota and intestinal barrier function by 16S rRNA sequencing, Western blot, quantitative fluorescent PCR (qPCR), and morphological observation. Moreover, we constructed a single layer of the human intestinal epithelium model to determine the effect of vaccarin in vitro. RESULTS The experimental results showed that vaccarin alleviated inflammatory mediators in serum and intestinal tissue of mice (P < 0.05), which may depend on the improvement of tight junctions and gut microbiota (P < 0.05). Activation of extracellular regulated protein kinases (Erk1/2) stimulated myosin light chain kinase (MLCK). By inhibiting ERK expression (P < 0.05), vaccarin had similar effects to ERK inhibitors. In addition, the regulation of tight junction barriers also involved the abovementioned pathways in vivo. CONCLUSION Vaccarin could protect the intestinal barrier by inhibiting the ERK/MLCK signaling pathway and modulate the composition of the microbiota. These results suggested that vaccarin may be an effective candidate for improving intestinal barrier changes in T2DM.
Collapse
Affiliation(s)
- Jiang-Nan Sun
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Xiao-Yi Yu
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Bao Hou
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Min Ai
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Meng-Ting Qi
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Xin-Yu Ma
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Ming-Jie Cai
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Min Gao
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Wei-Wei Cai
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Lu-Lu Ni
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Fei Xu
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Yue-Tao Zhou
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China
| | - Li-Ying Qiu
- Wuxi Medical School, Jiangnan University, Wuxi, 214122, Jiangsu Province, PR China.
| |
Collapse
|
30
|
Cazzato G, Colagrande A, Andriola V, Lettini T, Cicco S, Candance PMV, Resta L, Vincenti L, Ingravallo G. Histological Hallmarks of Mucosal Healing in Inflammatory Bowel Diseases in the Era of Monoclonal Antibodies Therapy: New Insights and Perspectives. Diagnostics (Basel) 2021; 11:1570. [PMID: 34573912 PMCID: PMC8470623 DOI: 10.3390/diagnostics11091570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/26/2021] [Accepted: 08/27/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Chronic inflammatory bowel diseases (IBDs) are gaining increasing attention, both because they can severely reduce the quantity and quality of life, and because the advent of monoclonal antibodies has profoundly changed the natural history of these diseases. In recent years, the concept of mucosal healing has assumed a certain importance, and there are more and more clinical and pharmacological trials that consider this parameter among their endpoints, so much so that it may soon be included among the desirable clinical outcomes of patients with IBD. METHODS We performed a literature review of the Pubmed, Medline, and Web of Science (WoS) databases. RESULTS We selected 88 articles and then removed 6 duplicates; the final sample after accurate application of the inclusion criteria numbered 73 articles, with a level of evidence rating of three or four, according to Oxfords Evidence-based medicine. Our aim was to study the histological impact of monoclonal antibody therapies on mucosal healing, taking into consideration the few studies present in the literature. To perform this review, we compared studies that examined patients with Crohn's disease (CD) and/or ulcerative colitis (UC) undergoing monoclonal therapy versus patients undergoing other non-biological therapies (PICO statements). CONCLUSIONS We try to delineate how monoclonal antibodies have changed the natural history of IBD, acting at the microscopic level, and we believe that a careful analysis of the histopathology and the definition of the objective criteria for "Mucosa Healing" should enable this concept to be included among the clinical endpoints of patients affected by IBD, thus contributing to a better therapeutic management of these patients.
Collapse
Affiliation(s)
- Gerardo Cazzato
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.C.); (T.L.); (P.M.V.C.); (L.R.)
| | - Anna Colagrande
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.C.); (T.L.); (P.M.V.C.); (L.R.)
| | - Valeria Andriola
- Section of General Surgery, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.A.); (L.V.)
| | - Teresa Lettini
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.C.); (T.L.); (P.M.V.C.); (L.R.)
| | - Sebastiano Cicco
- Section of Internal Medicine, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Pragnell Mary Victoria Candance
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.C.); (T.L.); (P.M.V.C.); (L.R.)
| | - Leonardo Resta
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.C.); (T.L.); (P.M.V.C.); (L.R.)
| | - Leonardo Vincenti
- Section of General Surgery, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (V.A.); (L.V.)
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.C.); (T.L.); (P.M.V.C.); (L.R.)
| |
Collapse
|
31
|
Chong DLW, Rebeyrol C, José RJ, Williams AE, Brown JS, Scotton CJ, Porter JC. ICAM-1 and ICAM-2 Are Differentially Expressed and Up-Regulated on Inflamed Pulmonary Epithelium, but Neither ICAM-2 nor LFA-1: ICAM-1 Are Required for Neutrophil Migration Into the Airways In Vivo. Front Immunol 2021; 12:691957. [PMID: 34484188 PMCID: PMC8415445 DOI: 10.3389/fimmu.2021.691957] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/29/2021] [Indexed: 01/21/2023] Open
Abstract
Neutrophil migration into the airways is an important process to fight infection and is mediated by cell adhesion molecules. The intercellular adhesion molecules, ICAM-1 (CD54) and ICAM-2 (CD102) are known ligands for the neutrophil integrins, lymphocyte function associated antigen (LFA)-1 (αLβ2; CD11a/CD18), and macrophage-1 antigen (Mac-1;αMβ2;CD11b/CD18) and are implicated in leukocyte migration into the lung. However, it is ill-defined how neutrophils exit the lung and the role for ICAMs in trans-epithelial migration (TEpM) across the bronchial or alveolar epithelium. We found that human and murine alveolar epithelium expressed ICAM-1, whilst the bronchial epithelium expressed ICAM-2, and both were up-regulated during inflammatory stimulation in vitro and in inflammatory lung diseases such as cystic fibrosis. Although β2 integrins interacting with ICAM-1 and -2 mediated neutrophil migration across human bronchial epithelium in vitro, neither ICAM-2 nor LFA-1 binding of ICAM-1 mediated murine neutrophil migration into the lung or broncho-alveolar space during LPS-induced inflammation in vivo. Furthermore, TEpM of neutrophils themselves resulted in increased epithelial junctional permeability and reduced barrier function in vitro. This suggests that although β2 integrins interacting with ICAMs may regulate low levels of neutrophil traffic in healthy lung or early in inflammation when the epithelial barrier is intact; these interactions may be redundant later in inflammation when epithelial junctions are disrupted and no longer limit TEpM.
Collapse
Affiliation(s)
- Deborah L. W. Chong
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Carine Rebeyrol
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Ricardo J. José
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Andrew E. Williams
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Jeremy S. Brown
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| | - Chris J. Scotton
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
- Institute of Biomedical and Clinical Sciences, College of Medicine & Health, Exeter, United Kingdom
| | - Joanna C. Porter
- Centre for Inflammation and Tissue Repair, Division of Medicine, University College London, London, United Kingdom
| |
Collapse
|
32
|
Sfera A, Osorio C, Zapata Martín del Campo CM, Pereida S, Maurer S, Maldonado JC, Kozlakidis Z. Endothelial Senescence and Chronic Fatigue Syndrome, a COVID-19 Based Hypothesis. Front Cell Neurosci 2021; 15:673217. [PMID: 34248502 PMCID: PMC8267916 DOI: 10.3389/fncel.2021.673217] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome is a serious illness of unknown etiology, characterized by debilitating exhaustion, memory impairment, pain and sleep abnormalities. Viral infections are believed to initiate the pathogenesis of this syndrome although the definite proof remains elusive. With the unfolding of COVID-19 pandemic, the interest in this condition has resurfaced as excessive tiredness, a major complaint of patients infected with the SARS-CoV-2 virus, often lingers for a long time, resulting in disability, and poor life quality. In a previous article, we hypothesized that COVID-19-upregulated angiotensin II triggered premature endothelial cell senescence, disrupting the intestinal and blood brain barriers. Here, we hypothesize further that post-viral sequelae, including myalgic encephalomyelitis/chronic fatigue syndrome, are promoted by the gut microbes or toxin translocation from the gastrointestinal tract into other tissues, including the brain. This model is supported by the SARS-CoV-2 interaction with host proteins and bacterial lipopolysaccharide. Conversely, targeting microbial translocation and cellular senescence may ameliorate the symptoms of this disabling illness.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | | | | | | | - Steve Maurer
- Patton State Hospital, San Bernardino, CA, United States
| | - Jose Campo Maldonado
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
33
|
Superoxide Dismutase 3-Transduced Mesenchymal Stem Cells Preserve Epithelial Tight Junction Barrier in Murine Colitis and Attenuate Inflammatory Damage in Epithelial Organoids. Int J Mol Sci 2021; 22:ijms22126431. [PMID: 34208517 PMCID: PMC8233984 DOI: 10.3390/ijms22126431] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/04/2021] [Accepted: 06/11/2021] [Indexed: 01/08/2023] Open
Abstract
Superoxide dismutase 3 (SOD3), also known as extracellular superoxide dismutase, is an enzyme that scavenges reactive oxygen species (ROS). It has been reported that SOD3 exerts anti-inflammatory abilities in several immune disorders. However, the effect of SOD3 and the underlying mechanism in inflammatory bowel disease (IBD) have not been uncovered. Therefore, in the present study, we investigated whether SOD3 can protect intestinal cells or organoids from inflammation-mediated epithelial damage. Cells or mice were treated with SOD3 protein or SOD3-transduced mesenchymal stem cells (MSCs). Caco-2 cells or intestinal organoids stimulated with pro-inflammatory cytokines were used to evaluate the protective effect of SOD3 on epithelial junctional integrity. Dextran sulfate sodium (DSS)-induced colitis mice received SOD3 or SOD3-transduced MSCs (SOD3-MSCs), and were assessed for severity of disease and junctional protein expression. The activation of the mitogen-activated protein kinase (MAPK) pathway and elevated expression of cytokine-encoding genes decreased in TNF-α-treated Caco-2 cells or DSS-induced colitis mice when treated with SOD3 or SOD3-MSCs. Moreover, the SOD3 supply preserved the expression of tight junction (ZO-1, occludin) or adherence junction (E-cadherin) proteins when inflammation was induced. SOD3 also exerted a protective effect against cytokine- or ROS-mediated damage to intestinal organoids. These results indicate that SOD3 can effectively alleviate enteritis symptoms by maintaining the integrity of epithelial junctions and regulating inflammatory- and oxidative stress.
Collapse
|
34
|
Tong Xie Yao Fang: A Classic Chinese Medicine Prescription with Potential for the Treatment of Ulcerative Colitis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5548764. [PMID: 34211567 PMCID: PMC8208878 DOI: 10.1155/2021/5548764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/05/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023]
Abstract
The prescription of Tong Xie Yao Fang (TXYF) was derived from the Yuan dynasty “Dan Brook Heart Law,” which was a representative formula for treating liver-spleen disharmony, diarrhea, and abdominal pain. The prescription is composed of four herbs for soothing the liver and strengthening the spleen. TXYF is reportedly capable of eliminating discomfort in ulcerative colitis (UC). This classic formula has been widely used for regulating gastrointestinal motor dysfunction and repairing colon mucosa. This review aims to provide current information on the pharmacology and clinical research of TXYF in the treatment of UC, and to critically appraise that information, in order to guide the future clinical use and experimental study of TXYF in the treatment of UC. We searched online databases including PubMed, CNKI, and Google Scholar for research published between 2010 and 2020 on TXYF and its efficacy in the treatment of UC. The findings indicated that TXYF has anti-inflammatory and immunomodulatory effects, regulates cell signal transduction, brain-gut axis, and intestinal flora in UC, and may promote targeting of bone mesenchymal stem cells (BMSCs) to the colonic mucosa and accelerate healing of the colonic mucosal barrier. In addition, the results of clinical studies showed that TXYF has good efficacy and few adverse reactions in the treatment of UC. Although it has achieved some success, the research is limited by deficiencies; there is a lack of unified standards for the construction of UC animal models and for administration regimen. In addition, the dosage of TXYF is not consistent and lacks pharmacological verification, and clinical trial data are not detailed or sufficiently rigorous. Therefore, a more rigorous, comprehensive, and in-depth study of TXYF in the treatment of UC is needed.
Collapse
|
35
|
Labarta-Bajo L, Nilsen SP, Humphrey G, Schwartz T, Sanders K, Swafford A, Knight R, Turner JR, Zúñiga EI. Type I IFNs and CD8 T cells increase intestinal barrier permeability after chronic viral infection. J Exp Med 2021; 217:152069. [PMID: 32880630 PMCID: PMC7953738 DOI: 10.1084/jem.20192276] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/29/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Intestinal barrier leakage constitutes a potential therapeutic target for many inflammatory diseases and represents a disease progression marker during chronic viral infections. However, the causes of altered gut barrier remain mostly unknown. Using murine infection with lymphocytic choriomeningitis virus, we demonstrate that, in contrast to an acute viral strain, a persistent viral isolate leads to long-term viral replication in hematopoietic and mesenchymal cells, but not epithelial cells (IECs), in the intestine. Viral persistence drove sustained intestinal epithelial barrier leakage, which was characterized by increased paracellular flux of small molecules and was associated with enhanced colitis susceptibility. Type I IFN signaling caused tight junction dysregulation in IECs, promoted gut microbiome shifts and enhanced intestinal CD8 T cell responses. Notably, both type I IFN receptor blockade and CD8 T cell depletion prevented infection-induced barrier leakage. Our study demonstrates that infection with a virus that persistently replicates in the intestinal mucosa increases epithelial barrier permeability and reveals type I IFNs and CD8 T cells as causative factors of intestinal leakage during chronic infections.
Collapse
Affiliation(s)
- Lara Labarta-Bajo
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
| | - Steven P Nilsen
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Gregory Humphrey
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Tara Schwartz
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Karenina Sanders
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| | - Austin Swafford
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, La Jolla, CA.,Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA.,Department of Bioengineering, University of California, San Diego, La Jolla, CA.,Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Elina I Zúñiga
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA
| |
Collapse
|
36
|
LasR-deficient Pseudomonas aeruginosa variants increase airway epithelial mICAM-1 expression and enhance neutrophilic lung inflammation. PLoS Pathog 2021; 17:e1009375. [PMID: 33690714 PMCID: PMC7984618 DOI: 10.1371/journal.ppat.1009375] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 03/22/2021] [Accepted: 02/13/2021] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa causes chronic airway infections, a major determinant of lung inflammation and damage in cystic fibrosis (CF). Loss-of-function lasR mutants commonly arise during chronic CF infections, are associated with accelerated lung function decline in CF patients and induce exaggerated neutrophilic inflammation in model systems. In this study, we investigated how lasR mutants modulate airway epithelial membrane bound ICAM-1 (mICAM-1), a surface adhesion molecule, and determined its impact on neutrophilic inflammation in vitro and in vivo. We demonstrated that LasR-deficient strains induce increased mICAM-1 levels in airway epithelial cells compared to wild-type strains, an effect attributable to the loss of mICAM-1 degradation by LasR-regulated proteases and associated with enhanced neutrophil adhesion. In a subacute airway infection model, we also observed that lasR mutant-infected mice displayed greater airway epithelial ICAM-1 expression and increased neutrophilic pulmonary inflammation. Our findings provide new insights into the intricate interplay between lasR mutants, LasR-regulated proteases and airway epithelial ICAM-1 expression, and reveal a new mechanism involved in the exaggerated inflammatory response induced by lasR mutants.
Collapse
|
37
|
Azcutia V, Kelm M, Luissint AC, Boerner K, Flemming S, Quiros M, Newton G, Nusrat A, Luscinskas FW, Parkos CA. Neutrophil expressed CD47 regulates CD11b/CD18-dependent neutrophil transepithelial migration in the intestine in vivo. Mucosal Immunol 2021; 14:331-341. [PMID: 32561828 PMCID: PMC7749029 DOI: 10.1038/s41385-020-0316-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/24/2020] [Accepted: 06/04/2020] [Indexed: 02/04/2023]
Abstract
Dysregulated neutrophil (PMN) transmigration across epithelial surfaces (TEpM) significantly contributes to chronic inflammatory diseases, yet mechanisms defining this process remain poorly understood. In the intestine, uncontrolled PMN TEpM is a hallmark of disease flares in ulcerative colitis. Previous in vitro studies directed at identifying molecular determinants that mediate TEpM have shown that plasma membrane proteins including CD47 and CD11b/CD18 play key roles in regulating PMN TEpM across monolayers of intestinal epithelial cells. Here, we show that CD47 modulates PMN TEpM in vivo using an ileal loop assay. Importantly, using novel tissue-specific CD47 knockout mice and in vitro approaches, we report that PMN-expressed, but not epithelial-expressed CD47 plays a major role in regulating PMN TEpM. We show that CD47 associates with CD11b/CD18 in the plasma membrane of PMN, and that loss of CD47 results in impaired CD11b/CD18 activation. In addition, in vitro and in vivo studies using function blocking antibodies support a role of CD47 in regulating CD11b-dependent PMN TEpM and chemotaxis. Taken together, these findings provide new insights for developing approaches to target dysregulated PMN infiltration in the intestine. Moreover, tissue-specific CD47 knockout mice constitute an important new tool to study contributions of cells expressing CD47 to inflammation in vivo.
Collapse
Affiliation(s)
- Veronica Azcutia
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.,Correspondence to:Veronica Azcutia, PhD. Department of Pathology, University of Michigan School of Medicine, 109 Zina Pitcher, BSRB Rm-4620. Ann Arbor, Michigan 48109, USA. Tel: (734) 936-1856 ; Charles A. Parkos, MD, PhD. Department of Pathology, University of Michigan School of Medicine, 2800 Plymouth Road, NCRC 30-1537. Ann Arbor, Michigan 48109, USA. Tel: (734) 763-6384 Fax: (734) 763-4782
| | - Matthias Kelm
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Anny-Claude Luissint
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Kevin Boerner
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Sven Flemming
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Miguel Quiros
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Gail Newton
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Francis W. Luscinskas
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Charles A. Parkos
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.,Correspondence to:Veronica Azcutia, PhD. Department of Pathology, University of Michigan School of Medicine, 109 Zina Pitcher, BSRB Rm-4620. Ann Arbor, Michigan 48109, USA. Tel: (734) 936-1856 ; Charles A. Parkos, MD, PhD. Department of Pathology, University of Michigan School of Medicine, 2800 Plymouth Road, NCRC 30-1537. Ann Arbor, Michigan 48109, USA. Tel: (734) 763-6384 Fax: (734) 763-4782
| |
Collapse
|
38
|
Croasdell Lucchini A, Gachanja NN, Rossi AG, Dorward DA, Lucas CD. Epithelial Cells and Inflammation in Pulmonary Wound Repair. Cells 2021; 10:339. [PMID: 33562816 PMCID: PMC7914803 DOI: 10.3390/cells10020339] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/15/2021] [Accepted: 01/30/2021] [Indexed: 12/15/2022] Open
Abstract
Respiratory diseases are frequently characterised by epithelial injury, airway inflammation, defective tissue repair, and airway remodelling. This may occur in a subacute or chronic context, such as asthma and chronic obstructive pulmonary disease, or occur acutely as in pathogen challenge and acute respiratory distress syndrome (ARDS). Despite the frequent challenge of lung homeostasis, not all pulmonary insults lead to disease. Traditionally thought of as a quiescent organ, emerging evidence highlights that the lung has significant capacity to respond to injury by repairing and replacing damaged cells. This occurs with the appropriate and timely resolution of inflammation and concurrent initiation of tissue repair programmes. Airway epithelial cells are key effectors in lung homeostasis and host defence; continual exposure to pathogens, toxins, and particulate matter challenge homeostasis, requiring robust defence and repair mechanisms. As such, the epithelium is critically involved in the return to homeostasis, orchestrating the resolution of inflammation and initiating tissue repair. This review examines the pivotal role of pulmonary airway epithelial cells in initiating and moderating tissue repair and restitution. We discuss emerging evidence of the interactions between airway epithelial cells and candidate stem or progenitor cells to initiate tissue repair as well as with cells of the innate and adaptive immune systems in driving successful tissue regeneration. Understanding the mechanisms of intercellular communication is rapidly increasing, and a major focus of this review includes the various mediators involved, including growth factors, extracellular vesicles, soluble lipid mediators, cytokines, and chemokines. Understanding these areas will ultimately identify potential cells, mediators, and interactions for therapeutic targeting.
Collapse
Affiliation(s)
| | | | | | | | - Christopher D. Lucas
- University of Edinburgh Centre for Inflammation Research, Queen’s Medical Research Institute, Edinburgh Bioquarter, Edinburgh EH16 4TJ, UK; (A.C.L.); (N.N.G.); (A.G.R.); (D.A.D.)
| |
Collapse
|
39
|
Lin WC, Fessler MB. Regulatory mechanisms of neutrophil migration from the circulation to the airspace. Cell Mol Life Sci 2021; 78:4095-4124. [PMID: 33544156 PMCID: PMC7863617 DOI: 10.1007/s00018-021-03768-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/22/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
The neutrophil, a short-lived effector leukocyte of the innate immune system best known for its proteases and other degradative cargo, has unique, reciprocal physiological interactions with the lung. During health, large numbers of ‘marginated’ neutrophils reside within the pulmonary vasculature, where they patrol the endothelial surface for pathogens and complete their life cycle. Upon respiratory infection, rapid and sustained recruitment of neutrophils through the endothelial barrier, across the extravascular pulmonary interstitium, and again through the respiratory epithelium into the airspace lumen, is required for pathogen killing. Overexuberant neutrophil trafficking to the lung, however, causes bystander tissue injury and underlies several acute and chronic lung diseases. Due in part to the unique architecture of the lung’s capillary network, the neutrophil follows a microanatomic passage into the distal airspace unlike that observed in other end-organs that it infiltrates. Several of the regulatory mechanisms underlying the stepwise recruitment of circulating neutrophils to the infected lung have been defined over the past few decades; however, fundamental questions remain. In this article, we provide an updated review and perspective on emerging roles for the neutrophil in lung biology, on the molecular mechanisms that control the trafficking of neutrophils to the lung, and on past and ongoing efforts to design therapeutics to intervene upon pulmonary neutrophilia in lung disease.
Collapse
Affiliation(s)
- Wan-Chi Lin
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, P.O. Box 12233, MD D2-01, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
40
|
Peixoto R, Silva LMR, López-Osório S, Zhou E, Gärtner U, Conejeros I, Taubert A, Hermosilla C. Fasciola hepatica induces weak NETosis and low production of intra- and extracellular ROS in exposed bovine polymorphonuclear neutrophils. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103787. [PMID: 32791176 DOI: 10.1016/j.dci.2020.103787] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/26/2020] [Accepted: 06/28/2020] [Indexed: 06/11/2023]
Abstract
Fasciola hepatica is the causative agent of fasciolosis, a worldwide distributed zoonotic disease, leading to hepatitis in humans and livestock. Newly excysted juveniles (NEJ) of F. hepatica are the first invasive stages to encounter leukocytes of host innate immune system in vivo. Among leukocytes, polymorphonuclear neutrophils (PMN) are the most abundant granulocytes of blood system and first ones to migrate into infection sites. PMN are able to cast neutrophil extracellular traps (NETs), also known as NETosis, consisting of nuclear DNA, decorated with histones, enzymes and antimicrobial peptides, which can entrap and eventually kill invasive parasites. Given that only few large parasitic helminths have been identified as potent NETosis inducers, here we studied for first time whether different F. hepatica stages can also trigger NETosis. Therefore, isolated bovine PMN were co-cultured with viable F. hepatica-NEJ, -metacercariae, -eggs and soluble antigen (FhAg). Interestingly, all stages failed to induce considerable levels of NETosis as detected by immunofluorescence- and scanning electron microscopy (SEM) analyses. NEJ remained motile until the end of incubation period. In line, NETosis quantification via nuclear area expansion (NAE) analysis revealed NEJ as weak NETosis inducers. However, bovine PMN frequently displaced towards motile NEJ and were found attached to NEJ surfaces. Functional PMN chemotaxis assays using vital F. hepatica-NEJ revealed a slight increase of PMN migration when compared to non-exposed controls. Additional experiments on intra- and extracellular reactive oxygen species (ROS) production revealed that soluble FhAg failed to induce ROS production of exposed PMN. Finally, mitochondrial oxygen consumption rates (OCR), extracellular acidification rates (ERAC) and proton production rates (PPR) were not significantly increased in FhAg-stimulated PMN. In summary, data suggest that F. hepatica might effectively evade PMN activation and NETosis by secreting parasite-specific molecules to either resolve NETs or to impair NETosis signaling pathways. We call for future molecular analysis not only on F. hepatica-derived NETosis modulation but also on its possible role in fasciolosis-associated pathology in vivo.
Collapse
Affiliation(s)
- Raquel Peixoto
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany.
| | - Liliana M R Silva
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Sara López-Osório
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany; CIBAV Research Group, Faculty of Agrarian Sciences, University of Antioquia, Medellin, Colombia
| | - Ershun Zhou
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Ulrich Gärtner
- Institute of Anatomy and Cell Biology, Faculty of Human Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Ivan Conejeros
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Anja Taubert
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
41
|
Semin I, Ninnemann J, Bondareva M, Gimaev I, Kruglov AA. Interplay Between Microbiota, Toll-Like Receptors and Cytokines for the Maintenance of Epithelial Barrier Integrity. Front Med (Lausanne) 2021; 8:644333. [PMID: 34124086 PMCID: PMC8194074 DOI: 10.3389/fmed.2021.644333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 05/06/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal tract is densely populated by microbiota consisting of various commensal microorganisms that are instrumental for the healthy state of the living organism. Such commensals generate various molecules that can be recognized by the Toll-like receptors of the immune system leading to the inflammation marked by strong upregulation of various proinflammatory cytokines, such as TNF, IL-6, and IL-1β. To prevent excessive inflammation, a single layer of constantly renewing, highly proliferating epithelial cells (IEC) provides proper segregation of such microorganisms from the body cavities. There are various triggers which facilitate the disturbance of the epithelial barrier which often leads to inflammation. However, the nature and duration of the stress may determine the state of the epithelial cells and their responses to cytokines. Here we discuss the role of the microbiota-TLR-cytokine axis in the maintenance of the epithelial tissue integrity. In particular, we highlight discrepancies in the function of TLR and cytokines in IEC barrier during acute or chronic inflammation and we suggest that intervention strategies should be applied based on the type of inflammation.
Collapse
Affiliation(s)
- Iaroslav Semin
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Justus Ninnemann
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Marina Bondareva
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ilia Gimaev
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey A. Kruglov
- German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- *Correspondence: Andrey A. Kruglov
| |
Collapse
|
42
|
Barros-Becker F, Squirrell JM, Burke R, Chini J, Rindy J, Karim A, Eliceiri KW, Gibson A, Huttenlocher A. Distinct Tissue Damage and Microbial Cues Drive Neutrophil and Macrophage Recruitment to Thermal Injury. iScience 2020; 23:101699. [PMID: 33196024 PMCID: PMC7644964 DOI: 10.1016/j.isci.2020.101699] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/17/2020] [Accepted: 10/14/2020] [Indexed: 12/21/2022] Open
Abstract
Tissue damage triggers a rapid innate immune response that mediates host defense. Previously we reported that thermal damage of the larval zebrafish fin disrupts collagen organization and induces a robust and potentially damaging innate immune response. The mechanisms that drive damaging versus protective neutrophil inflammation in interstitial tissues remain unclear. Here we identify distinct cues in the tissue microenvironment that differentially drive neutrophil and macrophage responses to sterile injury. Using live imaging, we found a motile zone for neutrophils, but not macrophages, in collagen-free regions and identified a specific role for interleukin-6 (IL-6) receptor signaling in neutrophil responses to thermal damage. IL-6 receptor mutants show impaired neutrophil recruitment to sterile thermal injury that was not present in tissues infected with Pseudomonas aeruginosa. These findings identify distinct signaling networks during neutrophil recruitment to sterile and microbial damage cues and provide a framework to limit potentially damaging neutrophil inflammation.
Collapse
Affiliation(s)
- Francisco Barros-Becker
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.,Cellular and Molecular Biology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Jayne M Squirrell
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA
| | - Russell Burke
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia Chini
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.,School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Julie Rindy
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| | - Aos Karim
- Department of Surgery, University of Wisconsin-Madison, Madison WI, USA
| | - Kevin W Eliceiri
- Laboratory for Optical and Computational Instrumentation, Center for Quantitative Cell Imaging, University of Wisconsin-Madison, Madison, WI, USA.,Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, USA
| | - Angela Gibson
- Department of Surgery, University of Wisconsin-Madison, Madison WI, USA
| | - Anna Huttenlocher
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.,Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
43
|
Dalal PJ, Sumagin R. Emerging Functions of ICAM-1 in Macrophage Efferocytosis and Wound Healing. JOURNAL OF CELLULAR IMMUNOLOGY 2020; 2:250-253. [PMID: 33426539 PMCID: PMC7793567 DOI: 10.33696/immunology.2.051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Prarthana J Dalal
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
44
|
Ji E, Wang T, Xu J, Fan J, Zhang Y, Guan Y, Yang H, Wei J, Zhang G, Huang L. Systematic Investigation of the Efficacy of Sinitang Decoction Against Ulcerative Colitis. Front Pharmacol 2020; 11:1337. [PMID: 32982747 PMCID: PMC7490561 DOI: 10.3389/fphar.2020.01337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to investigate the precise clinical use of Sinitang decoction (SNT) in ulcerative colitis (UC). Network pharmacology-based analysis of the drug components-targets-diseases-pathways was used to predict the possible clinical applications of SNT. Next, 2,4,6-trinitrobenzenesulfonic acid (TNBS) was used to establish a rat model of UC, and the efficacy of SNT against UC was tested, followed by a proteomic analysis of the specific signatures regulated by SNT against UC. SNT was predicted to be effective in inflammatory bowel disease, UC, and several other diseases. In the rats with UC, SNT decreased the disease activity index and colon mucosal damage index compared to the untreated UC model rats. Additionally, SNT reversed the upregulated levels of serum tumor necrosis factor (TNF)-α, prostaglandin E2 (PGE2), interleukin (IL)-6, and nitric oxide (NO) in UC model rats. The proteomic analysis identified 78 proteins that were differentially regulated by SNT in the rats with UC, which were associated with the Gene Ontology terms sulfur compound binding, calcium ion binding, and Toll-like receptor (TLR)-4 binding. Among these differentially regulated proteins, C-reactive protein (CRP) and collagen alpha-1(XII) chain (COL12A1) were found to be signature proteins associated with the efficacy of SNT against UC. This study represents the first precise investigation of the efficacy and mechanisms of SNT against UC, and shows that SNT is a promising candidate for personalized management of UC.
Collapse
Affiliation(s)
- Enhui Ji
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tingting Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianwei Fan
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Yi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongxia Guan
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Hongjun Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junying Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guimin Zhang
- State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Luqi Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
45
|
Lin WC, Gowdy KM, Madenspacher JH, Zemans RL, Yamamoto K, Lyons-Cohen M, Nakano H, Janardhan K, Williams CJ, Cook DN, Mizgerd JP, Fessler MB. Epithelial membrane protein 2 governs transepithelial migration of neutrophils into the airspace. J Clin Invest 2020; 130:157-170. [PMID: 31550239 DOI: 10.1172/jci127144] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 09/18/2019] [Indexed: 02/06/2023] Open
Abstract
Whether respiratory epithelial cells regulate the final transit of extravasated neutrophils into the inflamed airspace or are a passive barrier is poorly understood. Alveolar epithelial type 1 (AT1) cells, best known for solute transport and gas exchange, have few established immune roles. Epithelial membrane protein 2 (EMP2), a tetraspan protein that promotes recruitment of integrins to lipid rafts, is highly expressed in AT1 cells but has no known function in lung biology. Here, we show that Emp2-/- mice exhibit reduced neutrophil influx into the airspace after a wide range of inhaled exposures. During bacterial pneumonia, Emp2-/- mice had attenuated neutrophilic lung injury and improved survival. Bone marrow chimeras, intravital neutrophil labeling, and in vitro assays suggested that defective transepithelial migration of neutrophils into the alveolar lumen occurs in Emp2-/- lungs. Emp2-/- AT1 cells had dysregulated surface display of multiple adhesion molecules, associated with reduced raft abundance. Epithelial raft abundance was dependent upon putative cholesterol-binding motifs in EMP2, whereas EMP2 supported adhesion molecule display and neutrophil transmigration through suppression of caveolins. Taken together, we propose that EMP2-dependent membrane organization ensures proper display on AT1 cells of a suite of proteins required to instruct paracellular neutrophil traffic into the alveolus.
Collapse
Affiliation(s)
- Wan-Chi Lin
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kymberly M Gowdy
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Jennifer H Madenspacher
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Rachel L Zemans
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kazuko Yamamoto
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA.,Second Department of Internal Medicine, Nagasaki University Hospital, Nagasaki, Japan.,Department of Clinical Research Center, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Miranda Lyons-Cohen
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Hideki Nakano
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Kyathanahalli Janardhan
- Cellular & Molecular Pathology Branch, National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA.,Integrated Laboratory Systems Inc., Research Triangle Park, North Carolina, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
46
|
Wiesolek HL, Bui TM, Lee JJ, Dalal P, Finkielsztein A, Batra A, Thorp EB, Sumagin R. Intercellular Adhesion Molecule 1 Functions as an Efferocytosis Receptor in Inflammatory Macrophages. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:874-885. [PMID: 32035057 PMCID: PMC7180595 DOI: 10.1016/j.ajpath.2019.12.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/26/2022]
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is up-regulated during inflammation by several cell types. ICAM-1 is best known for its role in mediating leukocyte adhesion to endothelial cells and guiding leukocytes across the vascular wall. Recently, macrophages have been shown to express ICAM-1, however, their role in macrophage function is unclear. We found that ICAM-1 expression was induced during inflammatory macrophage polarization and high numbers of ICAM-1-expressing macrophages were noted in inflamed colon tissue in a murine colitis model and in human inflammatory bowel disease. Because tissue macrophages play a critical role in removing apoptotic/necrotic cells in inflammation and injury, a process termed efferocytosis, it was examined whether ICAM-1 contributes to this process. Genetic deletion (ICAM-1 knockout mice) or siRNA-mediated knockdown of ICAM-1 in isolated murine and human macrophages significantly impaired apoptotic cell (AC) engulfment. Impairment in the engulfment of Jurkat T cells, neutrophils, and epithelial cells was confirmed ex vivo by inflammatory macrophages and in vivo by thioglycolate-recruited peritoneal macrophages. Decreased efferocytosis was also seen in vitro and in vivo with inhibition of ICAM-1 adhesive interactions, using a function blocking anti-ICAM-1 antibody. Mechanistically, it was found that ICAM-1 actively redistributes to cluster around engulfed ACs to facilitate macrophage-AC binding. Our findings define a new role for ICAM-1 in promoting macrophage efferocytosis, a critical process in the resolution of inflammation and restoration of tissue homeostasis.
Collapse
Affiliation(s)
- Hannah L Wiesolek
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Triet M Bui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joseph J Lee
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Prarthana Dalal
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ariel Finkielsztein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ayush Batra
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Edward B Thorp
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
47
|
Bui TM, Wiesolek HL, Sumagin R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J Leukoc Biol 2020; 108:787-799. [PMID: 32182390 DOI: 10.1002/jlb.2mr0220-549r] [Citation(s) in RCA: 549] [Impact Index Per Article: 109.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/17/2020] [Accepted: 02/21/2020] [Indexed: 02/06/2023] Open
Abstract
ICAM-1 is a cell surface glycoprotein and an adhesion receptor that is best known for regulating leukocyte recruitment from circulation to sites of inflammation. However, in addition to vascular endothelial cells, ICAM-1 expression is also robustly induced on epithelial and immune cells in response to inflammatory stimulation. Importantly, ICAM-1 serves as a biosensor to transduce outside-in-signaling via association of its cytoplasmic domain with the actin cytoskeleton following ligand engagement of the extracellular domain. Thus, ICAM-1 has emerged as a master regulator of many essential cellular functions both at the onset and at the resolution of pathologic conditions. Because the role of ICAM-1 in driving inflammatory responses is well recognized, this review will mainly focus on newly emerging roles of ICAM-1 in epithelial injury-resolution responses, as well as immune cell effector function in inflammation and tumorigenesis. ICAM-1 has been of clinical and therapeutic interest for some time now; however, several attempts at inhibiting its function to improve injury resolution have failed. Perhaps, better understanding of its beneficial roles in resolution of inflammation or its emerging function in tumorigenesis will spark new interest in revisiting the clinical value of ICAM-1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Triet M Bui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hannah L Wiesolek
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
48
|
Kelm M, Lehoux S, Azcutia V, Cummings RD, Nusrat A, Parkos CA, Brazil JC. Regulation of neutrophil function by selective targeting of glycan epitopes expressed on the integrin CD11b/CD18. FASEB J 2019; 34:2326-2343. [PMID: 31907993 DOI: 10.1096/fj.201902542r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/30/2022]
Abstract
Polymorphonuclear neutrophils (PMNs) play a critical role in the innate immune response to invading pathogens. However, dysregulated mucosal trafficking of PMNs and associated epithelial tissue damage is a pathological hallmark of numerous inflammatory conditions including inflammatory bowel disease. The glycoprotein CD11b/CD18 plays a well-described role in regulating PMN transepithelial migration and PMN inflammatory functions. Previous studies have demonstrated that targeting of the N-linked glycan Lewis X on CD11b blocks PMN transepithelial migration (TEpM). Given evidence of glycosylation-dependent regulation of CD11b/CD18 function, we performed MALDI TOF Mass Spectrometry (MS) analyses on CD11b/CD18 purified from human PMNs. Unusual glycan epitopes identified on CD11b/CD18 included high Mannose oligosaccharides recognized by the Galanthus Nivalis lectin and biantennary galactosylated N-glycans recognized by the Phaseolus Vulgaris erythroagglutinin lectin. Importantly, we show that selective targeting of glycans on CD11b with such lectins results in altered intracellular signaling events that inhibit TEpM and differentially affect key PMN inflammatory functions including phagocytosis, superoxide release and apoptosis. Taken together, these data demonstrate that discrete glycan motifs expressed on CD11b/CD18 such as biantennary galactose could represent novel targets for selective manipulation of CD11b function and reduction of PMN-associated tissue damage in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Matthias Kelm
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Sylvain Lehoux
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Veronica Azcutia
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Asma Nusrat
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Charles A Parkos
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
49
|
The Role of Immune Cells and Cytokines in Intestinal Wound Healing. Int J Mol Sci 2019; 20:ijms20236097. [PMID: 31816903 PMCID: PMC6929186 DOI: 10.3390/ijms20236097] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/08/2019] [Accepted: 11/29/2019] [Indexed: 12/12/2022] Open
Abstract
Intestinal wound healing is a complicated process that not only involves epithelial cells but also immune cells. In this brief review, we will focus on discussing the contribution and regulation of four major immune cell types (neutrophils, macrophages, regulatory T cells, and innate lymphoid cells) and four cytokines (interleukin-10, tumor necrosis factor alpha, interleukin-6, and interleukin-22) to the wound repair process in the gut. Better understanding of these immune factors will be important for developing novel targeted therapy.
Collapse
|
50
|
Arebro J, Drakskog C, Winqvist O, Bachert C, Kumlien Georén S, Cardell L. Subsetting reveals CD16 high CD62L dim neutrophils in chronic rhinosinusitis with nasal polyps. Allergy 2019; 74:2499-2501. [PMID: 31116882 DOI: 10.1111/all.13919] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Julia Arebro
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology Karolinska Institutet Stockholm Sweden
- Department of ENT Diseases Karolinska University Hospital Stockholm Sweden
| | - Cecilia Drakskog
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology Karolinska Institutet Stockholm Sweden
| | - Ola Winqvist
- Department of Medicine, Immunology and Allergy Unit Karolinska University Hospital Stockholm Sweden
| | - Claus Bachert
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology Karolinska Institutet Stockholm Sweden
- Department of ENT Diseases Karolinska University Hospital Stockholm Sweden
- Upper Airways Research Laboratory Ghent University Ghent Belgium
| | - Susanna Kumlien Georén
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology Karolinska Institutet Stockholm Sweden
| | - Lars‐Olaf Cardell
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology Karolinska Institutet Stockholm Sweden
- Department of ENT Diseases Karolinska University Hospital Stockholm Sweden
| |
Collapse
|