1
|
Hopke A, Viens AL, Alexander NJ, Mun SJ, Mansour MK, Irimia D. Spleen tyrosine kinase inhibitors disrupt human neutrophil swarming and antifungal functions. Microbiol Spectr 2025; 13:e0254921. [PMID: 39601545 PMCID: PMC11705959 DOI: 10.1128/spectrum.02549-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 10/26/2024] [Indexed: 11/29/2024] Open
Abstract
Neutrophils communicate with one another and amplify their destructive power through swarming, a collective process that synchronizes the activities of multiple neutrophils against one target. The sequence of activities contributing to swarming against clusters of fungi has been recently uncovered. However, the molecular signals controlling the neutrophils' activities during the swarming process are just emerging. Here, we report that spleen tyrosine kinase (SYK) inhibitors severely impair neutrophil swarming responses, resulting in the complete loss of fungal restriction. These findings are enabled by a microscale platform to probe the biology of human neutrophils swarming against uniformly sized clusters of growing Candida albicans, a representative opportunistic fungal pathogen. We take advantage of the ability to monitor large arrays of swarms and quantify the effect of multiple chemical inhibitors on different phases of human neutrophil swarming. We show that inhibitors that interfere with PI3Ky signaling disrupt the regulation of the initiation of swarming, while the activation of JNK signaling is essential for the activation of biochemical antifungal functions. Furthermore, we reveal that granulocyte colony-stimulating factors (GCSF and GM-CSF) can partially rescue the antifungal functions of neutrophils exposed to SYK inhibitors. These findings advance our understanding of neutrophil swarming biology in humans and lay the foundation for novel therapeutics that may restore neutrophil function during immunosuppression. IMPORTANCE Neutrophils can amplify their destructive power through swarming, a crucial process against large targets that individual neutrophils cannot destroy. However, the molecular mechanisms controlling this process are just emerging. Here, we leveraged microscale tools to probe the biology of swarming against fungi. We used multiple chemical inhibitors and mapped SYK, PI3Ky, and JNK signaling roles during human neutrophil swarming against fungal clusters of Candida albicans. We also found that treating human neutrophils with GCSF and GM-CSF rescues some neutrophil antifungal function during SYK inhibition. These findings advance our understanding of swarming biology in humans while laying the foundation for developing therapeutics that enhance neutrophil function during immunosuppression.
Collapse
Affiliation(s)
- Alex Hopke
- BioMEMS Resource Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
| | - Adam L. Viens
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Natalie J. Alexander
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Seok Joon Mun
- BioMEMS Resource Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael K. Mansour
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel Irimia
- BioMEMS Resource Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Shriners Hospital for Children, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Hong S, He H, Fang P, Liu S, Chen C. Association of neutrophil-to-lymphocyte ratio and risk of cardiovascular and all-cause mortality in hypertension patients. Heliyon 2024; 10:e27517. [PMID: 38496832 PMCID: PMC10944217 DOI: 10.1016/j.heliyon.2024.e27517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024] Open
Abstract
Background and objective Hypertension affects over a billion people worldwide and is often associated with poor prognoses. The neutrophil-to-lymphocyte ratio (NLR) has become a significant marker, showing a connection to adverse outcomes in cardiovascular diseases (CVDs). The objective of this study is to examine the relationship between the NLR and outcomes in patients with hypertension. Methods The study included hypertensive individuals who were surveyed in the National Health and Nutrition Examination Survey (NHANES) from 2009 to 2018. Mortality status was determined using the data from National Death Index (NDI). To investigate the dose-response relationship, restricted cubic spline (RCS) models were used. This study employed adjusted cox proportional hazards regression models to compute hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs) for all-cause and cardiovascular mortality. The predictive accuracy of the NLR for survival outcomes was assessed utilizing time-dependent receiver operating characteristic (ROC) curve analysis. Results A total of 13,724 participants were included in the final analysis, including 7073 males and 6651 females. The cohort was stratified into higher (>2.0) and lower (≤2.0) NLR groups according to the median value. Over a median follow-up of 64 months, there were 1619 all-cause deaths and 522 cardiovascular deaths among participants. The RCS analysis indicated a non-linear relationship between NLR and the risk of mortality. The adjusted model showed that the group with a higher NLR had a significantly higher risk of all-cause (HR 1.47, 95% CI 1.22-1.77) and cardiovascular mortality (HR 2.08, 95% CI 1.52-2.86). ROC analysis showed that the area under the curves (AUCs) of 0.692, 0.662, 0.644, and 0.625 for predicting all-cause mortality, and 0.712, 0.692, 0.687, and 0.660 for cardiovascular mortality at 1, 3, 5, and 10 years. Conclusion Elevated NLR is associated with increased risk of cardiovascular and all-cause mortality, and NLR may independently predict outcomes in individuals with hypertension.
Collapse
Affiliation(s)
- Shaoqing Hong
- Corresponding author. Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.98 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China.
| | | | - Peng Fang
- Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.33 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China
| | - Shuai Liu
- Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.33 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China
| | - Changyi Chen
- Department of Cardiovascular Medicine, Huangshi Fifth Hospital, No.33 XiaLu Dadao, Xialu District, Huangshi, Hubei, 435005, China
| |
Collapse
|
3
|
Ramadan Q, Hazaymeh R, Zourob M. Immunity-on-a-Chip: Integration of Immune Components into the Scheme of Organ-on-a-Chip Systems. Adv Biol (Weinh) 2023; 7:e2200312. [PMID: 36866511 DOI: 10.1002/adbi.202200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/16/2023] [Indexed: 03/04/2023]
Abstract
Studying the immune system in vitro aims to understand how, when, and where the immune cells migrate/differentiate and respond to the various triggering events and the decision points along the immune response journey. It becomes evident that organ-on-a-chip (OOC) technology has a superior capability to recapitulate the cell-cell and tissue-tissue interaction in the body, with a great potential to provide tools for tracking the paracrine signaling with high spatial-temporal precision and implementing in situ real-time, non-destructive detection assays, therefore, enabling extraction of mechanistic information rather than phenotypic information. However, despite the rapid development in this technology, integration of the immune system into OOC devices stays among the least navigated tasks, with immune cells still the major missing components in the developed models. This is mainly due to the complexity of the immune system and the reductionist methodology of the OOC modules. Dedicated research in this field is demanded to establish the understanding of mechanism-based disease endotypes rather than phenotypes. Herein, we systemically present a synthesis of the state-of-the-art of immune-cantered OOC technology. We comprehensively outlined what is achieved and identified the technology gaps emphasizing the missing components required to establish immune-competent OOCs and bridge these gaps.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Riyadh, 11533, Kingdom of Saudi Arabia
| | - Rana Hazaymeh
- Almaarefa University, Diriyah, 13713, Kingdom of Saudi Arabia
| | | |
Collapse
|
4
|
Xiao Y, Cheng Y, Liu WJ, Liu K, Wang Y, Xu F, Wang DM, Yang Y. Effects of neutrophil fate on inflammation. Inflamm Res 2023; 72:2237-2248. [PMID: 37925664 DOI: 10.1007/s00011-023-01811-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/18/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
INTRODUCTION Neutrophils are important participants in the innate immune response. They rapidly and efficiently identify and clear infectious agents by expressing large numbers of membrane receptors. Upon tissue injury or pathogen invasion, neutrophils are the first immune cells to reach the site of injury and participate in the inflammatory response. MATERIALS AND METHODS A thorough search on PubMed related to neutrophil death or clearance pathways was performed. CONCLUSION Inflammatory response and tissue damage can be aggravated when neutrophils are not removed rapidly from the site of injury. Recent studies have shown that neutrophils can be cleared through a variety of pathways, including non-inflammatory and inflammatory death, as well as reverse migration. Non-inflammatory death pathways include apoptosis and autophagy. Inflammatory death pathways include necroptosis, pyroptosis and NETosis. This review highlights the basic properties of neutrophils and the impact of their clearance pathways on the inflammatory response.
Collapse
Affiliation(s)
- Yuan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yang Cheng
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wen-Jie Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Kun Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yan Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Feng Xu
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - De-Ming Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Yi Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
5
|
Van Bruggen S, Jarrot PA, Thomas E, Sheehy CE, Silva CMS, Hsu AY, Cunin P, Nigrovic PA, Gomes ER, Luo HR, Waterman CM, Wagner DD. NLRP3 is essential for neutrophil polarization and chemotaxis in response to leukotriene B4 gradient. Proc Natl Acad Sci U S A 2023; 120:e2303814120. [PMID: 37603754 PMCID: PMC10468616 DOI: 10.1073/pnas.2303814120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
Neutrophil recruitment to sites of infection and inflammation is an essential process in the early innate immune response. Upon activation, a subset of neutrophils rapidly assembles the multiprotein complex known as the NLRP3 inflammasome. The NLRP3 inflammasome forms at the microtubule organizing center, which promotes the formation of interleukin (IL)-1β and IL-18, essential cytokines in the immune response. We recently showed that mice deficient in NLRP3 (NLRP3-/-) have reduced neutrophil recruitment to the peritoneum in a model of thioglycolate-induced peritonitis. Here, we tested the hypothesis that this diminished recruitment could be, in part, the result of defects in neutrophil chemotaxis. We find that NLRP3-/- neutrophils show loss of cell polarization, as well as reduced directionality and velocity of migration toward increasing concentrations of leukotriene B4 (LTB4) in a chemotaxis assay in vitro, which was confirmed through intravital microscopy of neutrophil migration toward a laser-induced burn injury of the liver. Furthermore, pharmacologically blocking NLRP3 inflammasome assembly with MCC950 in vitro reduced directionality but preserved nondirectional movement, indicating that inflammasome assembly is specifically required for polarization and directional chemotaxis, but not cell motility per se. In support of this, pharmacological breakdown of the microtubule cytoskeleton via nocodazole treatment induced cell polarization and restored nondirectional cell migration in NLRP3-deficient neutrophils in the LTB4 gradient. Therefore, NLRP3 inflammasome assembly is required for establishment of cell polarity to guide the directional chemotactic migration of neutrophils.
Collapse
Affiliation(s)
- Stijn Van Bruggen
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
- Whitman Center, Marine Biological Laboratory, Chicago University, Woods Hole, MA02543
| | - Pierre-André Jarrot
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
| | - Eline Thomas
- Department of Life Science Technology, Imec, Leuven3001, Belgium
- Department of Biophysics, Katholieke Universiteit Leuven, Leuven3000, Belgium
| | - Casey E. Sheehy
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Camila M. S. Silva
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
| | - Alan Y. Hsu
- Department of Pathology, Harvard Medical School, Boston, MA02115
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Boston, MA02115
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Pierre Cunin
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Peter A. Nigrovic
- Division of Immunology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Edgar R. Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon1649-028, Portugal
| | - Hongbo R. Luo
- Department of Pathology, Harvard Medical School, Boston, MA02115
- Department of Pathology, Dana-Farber/Harvard Cancer Center, Boston, MA02115
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Clare M. Waterman
- Whitman Center, Marine Biological Laboratory, Chicago University, Woods Hole, MA02543
- Cell Biology and Physiology Center, National Heart, Lung, and Blood Institute of the NIH, Bethesda, MD20892
| | - Denisa D. Wagner
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA02115
- Department of Pediatrics, Harvard Medical School, Boston, MA02115
- Whitman Center, Marine Biological Laboratory, Chicago University, Woods Hole, MA02543
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA02115
| |
Collapse
|
6
|
Deng H, Li B, Shen Q, Zhang C, Kuang L, Chen R, Wang S, Ma Z, Li G. Mechanisms of diabetic foot ulceration: A review. J Diabetes 2023; 15:299-312. [PMID: 36891783 PMCID: PMC10101842 DOI: 10.1111/1753-0407.13372] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/23/2023] [Accepted: 02/16/2023] [Indexed: 03/10/2023] Open
Abstract
Diabetic foot ulcers (DFUs) are associated with complex pathogenic factors and are considered a serious complication of diabetes. The potential mechanisms underlying DFUs have been increasingly investigated. Previous studies have focused on the three aspects of diabetic peripheral vascular disease, neuropathy, and wound infections. With advances in technology, researchers have been gradually conducting studies using immune cells, endothelial cells, keratinocytes, and fibroblasts, as they are involved in wound healing. It has been reported that the upregulation or downregulation of molecular signaling pathways is essential for the healing of DFUs. With a recent increase in the awareness of epigenetics, its regulatory role in wound healing has become a much sought-after trend in the treatment of DFUs. This review focuses on four aspects involved in the pathogenesis of DFUs: physiological and pathological mechanisms, cellular mechanisms, molecular signaling pathway mechanisms, and epigenetics. Given the challenge in the treatment of DFUs, we are hopeful that our review will provide new ideas for peers.
Collapse
Affiliation(s)
- Haibo Deng
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Binghui Li
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Qian Shen
- School of Foreign StudiesZhongnan University of Economics and LawWuhanHubeiChina
| | - Chenchen Zhang
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Liwen Kuang
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Ran Chen
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - SiYuan Wang
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - ZhiQiang Ma
- Department of Wound Repair, Liyuan Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Gongchi Li
- Department of Hand Surgery, Union Hospital affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
7
|
Chowdhury O, Ghosh S, Das A, Liu H, Shang P, Stepicheva NA, Hose S, Sinha D, Chattopadhyay S. Sustained systemic inflammation increases autophagy and induces EMT/fibrotic changes in mouse liver cells: Protection by melatonin. Cell Signal 2023; 101:110521. [PMID: 36375715 DOI: 10.1016/j.cellsig.2022.110521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
The unending lifestyle stressors along with genetic predisposition, environmental factors and infections have pushed the immune system into a state of constant activity, leading to unresolved inflammation and increased vulnerability to chronic diseases. Liver fibrosis, an early-stage liver condition that increases the risk of developing liver diseases like cirrhosis and hepatocellular carcinoma, is among the various diseases linked to inflammation that dominate worldwide morbidity and mortality. We developed a mouse model with low-grade lipopolysaccharide (LPS) exposure that shows hepatic damage and a pro-inflammatory condition in the liver. We show that inflammation and oxidative changes increase autophagy in liver cells, a degradation process critical in maintaining cellular homeostasis. Our findings from in vivo and in vitro studies also show that induction of both inflammation and autophagy trigger epithelial-mesenchymal transition (EMT) and pro-fibrotic changes in hepatocytes. Inhibiting the inflammatory pathways with a naturally occurring NF-κB inhibitor and antioxidant, melatonin, could assuage the changes in autophagy and activation of EMT/fibrotic pathways in hepatocytes. Taken together, this study shows a pathway linking inflammation and autophagy which could be targeted for future drug development to delay the progression of liver fibrosis.
Collapse
Affiliation(s)
- Olivia Chowdhury
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ankur Das
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peng Shang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nadezda A Stepicheva
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sreya Chattopadhyay
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India; Centre for Research in Nanoscience and Nanotechnology (CRNN), University of Calcutta, JD-2, Salt Lake, Sector III, Kolkata 700098, India.
| |
Collapse
|
8
|
Development of Nanogel Loaded with Lidocaine for Wound-Healing: Illustration of Improved Drug Deposition and Skin Safety Analysis. Gels 2022; 8:gels8080466. [PMID: 35892725 PMCID: PMC9332171 DOI: 10.3390/gels8080466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 02/07/2023] Open
Abstract
A wound refers to a cut or blow that may result in primary or secondary infection or even death, if untreated. In the current study, we have explored the wound-healing properties of lidocaine nanogel, owing to its antioxidant and neutrophilic modulatory potential. Initially, the pre-formulation study was performed and then using central composite design (CCD) fabrication and the characterization of lidocaine-loaded nanoemulsion was carried out. After the preparation of a nanogel of lidocaine-loaded nanoemulsion, it was evaluated on various parameters, such as pH, spreadability, extrudability, drug content, in vitro drug release, dermatokinetic study and in vivo skin safety. Based on the pre-formulation study, the maximum solubility of lidocaine was found in oleic acid (324.41 ± 4.19 mg/mL) and in Tween 20 (192.05 ± 8.25 mg/mL), selected as a suitable emulsifier. The refractive index of the optimized nanoemulsion was found to be 1.35 ± 0.04, the electrokinetic potential was recorded as −15.47 ± 0.95 mV. The pH, spreadability and extrudability of nanogel was found to be 6.87 ± 0.51, 73.32 ± 4.59 gm.cm/sec and 107.41 ± 6.42 gm/cm2, respectively. The percentage of the cumulative drug content and drug release from nanogel was found to be 99.94 ± 1.70% and 93.00 ± 4.67%, respectively. Moreover, dermatokinetic study showed significantly (p < 0.0005) improved drug deposition and the in vivo skin safety study showed no sign of dermal erythematous lesion or any visible damage. Stability studies also testified the secureness of nanogel after storage in a prescribed environmental condition. Thus, this study provides substantial evidence for healing wounds effectively and the further evaluation of the in vivo model. The patent related to this work was published in the Indian Official Journal of the Patent Office (Issue number: 20/2022).
Collapse
|
9
|
Liu ML, Lyu X, Werth VP. Recent progress in the mechanistic understanding of NET formation in neutrophils. FEBS J 2022; 289:3954-3966. [PMID: 34042290 PMCID: PMC9107956 DOI: 10.1111/febs.16036] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/11/2021] [Accepted: 05/24/2021] [Indexed: 01/03/2023]
Abstract
Neutrophils are the most abundant circulating white blood cells and one of the major cell types of the innate immune system. Neutrophil extracellular traps (NETs) are a result of the extracellular release of nuclear chromatin from the ruptured nuclear envelope and plasma membrane. The externalized chromatin is an ancient defense weapon for animals to entrap and kill microorganisms in the extracellular milieu, thus protecting animals ranging from lower invertebrates to higher vertebrates. Although the externalized chromatin has the advantage of acting as anti-infective to protect against infections, extracellular chromatin might be problematic in higher vertebrate animals as they have an adaptive immune system that can trigger further immune or autoimmune responses. NETs and their associated nuclear and/or cytoplasmic components may induce sterile inflammation, immune, and autoimmune responses, leading to various human diseases. Though important in human pathophysiology, the cellular and molecular mechanisms of NET formation (also called NETosis) are not well understood. Given that nuclear chromatin forms the backbone of NETs, the nucleus is the root of the nuclear DNA extracellular traps. Thus, nuclear chromatin decondensation, along with the rupture of nuclear envelope and plasma membrane, is required for nuclear chromatin extracellular release and NET formation. So far, most of the literature focuses on certain signaling pathways, which are involved in NET formation but without explanation of cellular events and morphological changes described above. Here, we have summarized emerging evidence and discuss new mechanistic understanding, with our perspectives, in NET formation in neutrophils.
Collapse
Affiliation(s)
- Ming-Lin Liu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xing Lyu
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA,Department of Rheumatology and Immunology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Victoria P. Werth
- Corporal Michael J. Crescenz VAMC, Philadelphia, PA, 19104, USA,Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
10
|
Radovani B, Gudelj I. N-Glycosylation and Inflammation; the Not-So-Sweet Relation. Front Immunol 2022; 13:893365. [PMID: 35833138 PMCID: PMC9272703 DOI: 10.3389/fimmu.2022.893365] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammation is the main feature of many long-term inflammatory diseases such as autoimmune diseases, metabolic disorders, and cancer. There is a growing number of studies in which alterations of N-glycosylation have been observed in many pathophysiological conditions, yet studies of the underlying mechanisms that precede N-glycome changes are still sparse. Proinflammatory cytokines have been shown to alter the substrate synthesis pathways as well as the expression of glycosyltransferases required for the biosynthesis of N-glycans. The resulting N-glycosylation changes can further contribute to disease pathogenesis through modulation of various aspects of immune cell processes, including those relevant to pathogen recognition and fine-tuning the inflammatory response. This review summarizes our current knowledge of inflammation-induced N-glycosylation changes, with a particular focus on specific subsets of immune cells of innate and adaptive immunity and how these changes affect their effector functions, cell interactions, and signal transduction.
Collapse
Affiliation(s)
- Barbara Radovani
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Ivan Gudelj
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| |
Collapse
|
11
|
Leonova EI, Chirinskaite AV, Sopova JV. Atherosclerosis is a side effect of cellular senescence. RESEARCH RESULTS IN PHARMACOLOGY 2022. [DOI: 10.3897/rrpharmacology.8.81358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Atherosclerosis is a systemic autoimmune disease of the arterial wall characterized by chronic inflammation, high blood pressure, oxidative stress, and progressive loss of cell and organ function with aging. An imbalance of macrophage polarization is associated with many aging diseases, including atherosclerosis. The polarization toward the pro-inflammatory M1 macrophage is a major promoter of the atheroma formation. It is known that efferocytosis, or ingestion of apoptotic cells, is stimulated by M2 macrophage polarization. A failure of efferocytosis leads to the prolongation of chronic pathology in tissue. In addition, fat-laden macrophages contribute to the plague progression by transforming into foam cells in response to excess lipid deposition in arteries. In spite of the generally accepted theory that macrophages capture oxidized low-density lipoprotein by phagocytosis and become foam cells, we postulate that the main source of lipid accumulation in foam cells are senescent erythrocytes. Senescent erythrocytes lose their plasticity, which affects the rheological blood properties. It is known that their membrane contains high levels of cholesterol. There is evidence that senescent erythrocytes play a pathogenic role in the atheroma formation after breaking down during flowing through an artery bifurcation. Here we review the current knowledge on the impact of age-associated immune cells and red blood cells modifications on atherogenesis.
Graphical abstract:
Collapse
|
12
|
Maamar M, Artime A, Pariente E, Fierro P, Ruiz Y, Gutiérrez S, Tobalina M, Díaz-Salazar S, Ramos C, Olmos JM, Hernández JL. Post-COVID-19 syndrome, low-grade inflammation and inflammatory markers: a cross-sectional study. Curr Med Res Opin 2022; 38:901-909. [PMID: 35166141 PMCID: PMC8935459 DOI: 10.1080/03007995.2022.2042991] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Post-COVID syndrome (PCS) is a poorly known entity. An underlying chronic, low-grade inflammation (LGI) has been theorized as a pathophysiological mechanism. Available data on biomarkers in PCS show conflicting results. Our aim was to know whether subjects with PCS present higher levels of inflammatory markers, after a mild COVID-19. METHODS Analytical cross-sectional study. Cases of mild COVID-19 in a community setting were included. We collected epidemiological data (age, sex, BMI, smoking, comorbidities), variables of the acute COVID-19 (duration, symptoms), and data at 3 months after the acute phase (symptoms and laboratory test). Serum C-reactive protein (CRP), neutrophil and lymphocyte counts, neutrophil/lymphocyte ratio (NLR), lactate dehydrogenase, ferritin, fibrinogen, and D-dimer levels were analysed. LGI was defined as CRP >0.3 and <1.0 mg/dL. A subject was classified as PCS + if presented signs and symptoms >12 weeks after an infection consistent with COVID-19. Five composite indices (C1-C5) were developed, combining the upper ranges of biomarkers distributions. Multivariate analyses were performed. RESULTS We analysed 121 mild COVID-19 cases (mean age = 45.7 years, 56.2% women). Among the acute symptoms, women presented a higher frequency of fatigue (54.4% vs 30.2%; p = .008). PCS affected 35.8% of women and 20.8% of men (p = .07), and the most reported symptoms were fatigue (42.8%), anosmia (40%), ageusia (22.8%), dyspnea (17.1%) and myalgia (11.4%). Neutrophil count, NLR, CRP and fibrinogen showed the best correlations with PCS and were selected to develop the indices. In women PCS+, C1, C3 and C4 indices were more frequently met, while in men PCS+, C2, C5 and CRP were in the range of LGI. Anosmia, ageusia and fatigue were related to higher neutrophil counts, with sex differences. Fibrinogen levels were higher in persistent myalgia (510 ± 82 mg/dL vs 394 ± 87; p = .013). In multivariable analysis, a woman with a neutrophil count above the median, or with fibrinogen level or NLR in the highest tertile, had a 4-5-fold increased risk of prevalent PCS. A man with CRP in the range of LGI, or fibrinogen level or a neutrophil count in the highest tertile, had a 10-17-fold increased risk of prevalent PCS. CONCLUSIONS The data obtained in the present cross-sectional study seems to demonstrate a consistent association between PCS and upper ranges of the neutrophil count, NLR, fibrinogen, and CRP in the LGI range. Furthermore, composite indices appear useful in detecting relationships between slight elevations of biomarkers and PCS, and our study identifies relevant sex differences in symptoms and markers regarding the PCS.
Collapse
Affiliation(s)
- Meryam Maamar
- Emergency Service. Osakidetza, Servicio Vasco de Salud, Bilbao, País Vasco, Spain
| | - Arancha Artime
- El Llano - Primary Health Care Center, SESPA - Servicio Asturiano de Salud, Gijón Asturias, Spain
| | - Emilio Pariente
- Camargo Interior - Primary Health Care Center, Servicio Cántabro de Salud, Muriedas, Cantabria, Spain
- Depto. de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Cantabria, Spain
- CONTACT Emilio Pariente “Camargo Interior” Primary Care Center, Associate Professor, University of Cantabria, Avda Bilbao, s/n. 39600-Muriedas, Cantabria, Spain
| | - Patricia Fierro
- Camargo Interior - Primary Health Care Center, Servicio Cántabro de Salud, Muriedas, Cantabria, Spain
| | - Yolanda Ruiz
- Camargo Interior - Primary Health Care Center, Servicio Cántabro de Salud, Muriedas, Cantabria, Spain
| | - Silvia Gutiérrez
- Camargo Interior - Primary Health Care Center, Servicio Cántabro de Salud, Muriedas, Cantabria, Spain
| | - Marian Tobalina
- Camargo Interior - Primary Health Care Center, Servicio Cántabro de Salud, Muriedas, Cantabria, Spain
| | - Sara Díaz-Salazar
- Camargo Interior - Primary Health Care Center, Servicio Cántabro de Salud, Muriedas, Cantabria, Spain
| | - Carmen Ramos
- Depto. de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Cantabria, Spain
- Camargo Costa - Primary Health Care Center, Servicio Cántabro de Salud, Maliaño, Cantabria, Spain
| | - José M. Olmos
- Depto. de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Cantabria, Spain
- Servicio de Medicina Interna, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Valdecilla (IDIVAL), Santander, Cantabria, Spain
| | - José L. Hernández
- Depto. de Medicina y Psiquiatría, Universidad de Cantabria, Santander, Cantabria, Spain
- Servicio de Medicina Interna, Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Valdecilla (IDIVAL), Santander, Cantabria, Spain
| |
Collapse
|
13
|
Synergistic Antibacterial Effect of Zinc Oxide Nanoparticles and Polymorphonuclear Neutrophils. J Funct Biomater 2022; 13:jfb13020035. [PMID: 35466217 PMCID: PMC9036266 DOI: 10.3390/jfb13020035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Zinc oxide nanoparticles (ZnONPs) are inorganic nano-biomaterials with excellent antimicrobial properties. However, their effects on the anti-infection ability of the innate immune system remains poorly understood. The aim of the present study was to explore the potential immunomodulatory effects of ZnONPs on the innate immune system, represented by polymorphonuclear leukocytes (PMNs), and determine whether they can act synergistically to resist pathogen infections. In vitro experiment showed that ZnONPs not only exhibit obvious antibacterial activity at biocompatible concentrations but also enhance the antibacterial property of PMNs. In vivo experiments demonstrated the antibacterial effect of ZnONPs, accompanied by more infiltration of subcutaneous immune cells. Further ex vivo and in vitro experiments revealed that ZnONPs enhanced the migration of PMNs, promoted their bacterial phagocytosis efficiency, proinflammatory cytokine (TNF-α, IL-1β, and IL-6) expression, and reactive oxygen species (ROS) production. In summary, this study revealed potential synergistic effects of ZnONPs on PMNs to resist pathogen infection and the underlying mechanisms. The findings suggest that attempts should be made to fabricate and apply biomaterials in order to maximize their synergy with the innate immune system, thus promoting the host’s resistance to pathogen invasion.
Collapse
|
14
|
Seyres D, Cabassi A, Lambourne JJ, Burden F, Farrow S, McKinney H, Batista J, Kempster C, Pietzner M, Slingsby O, Cao TH, Quinn PA, Stefanucci L, Sims MC, Rehnstrom K, Adams CL, Frary A, Ergüener B, Kreuzhuber R, Mocciaro G, D’Amore S, Koulman A, Grassi L, Griffin JL, Ng LL, Park A, Savage DB, Langenberg C, Bock C, Downes K, Wareham NJ, Allison M, Vacca M, Kirk PDW, Frontini M. Transcriptional, epigenetic and metabolic signatures in cardiometabolic syndrome defined by extreme phenotypes. Clin Epigenetics 2022; 14:39. [PMID: 35279219 PMCID: PMC8917653 DOI: 10.1186/s13148-022-01257-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND This work is aimed at improving the understanding of cardiometabolic syndrome pathophysiology and its relationship with thrombosis by generating a multi-omic disease signature. METHODS/RESULTS We combined classic plasma biochemistry and plasma biomarkers with the transcriptional and epigenetic characterisation of cell types involved in thrombosis, obtained from two extreme phenotype groups (morbidly obese and lipodystrophy) and lean individuals to identify the molecular mechanisms at play, highlighting patterns of abnormal activation in innate immune phagocytic cells. Our analyses showed that extreme phenotype groups could be distinguished from lean individuals, and from each other, across all data layers. The characterisation of the same obese group, 6 months after bariatric surgery, revealed the loss of the abnormal activation of innate immune cells previously observed. However, rather than reverting to the gene expression landscape of lean individuals, this occurred via the establishment of novel gene expression landscapes. NETosis and its control mechanisms emerge amongst the pathways that show an improvement after surgical intervention. CONCLUSIONS We showed that the morbidly obese and lipodystrophy groups, despite some differences, shared a common cardiometabolic syndrome signature. We also showed that this could be used to discriminate, amongst the normal population, those individuals with a higher likelihood of presenting with the disease, even when not displaying the classic features.
Collapse
Affiliation(s)
- Denis Seyres
- National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK. .,Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK. .,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
| | - Alessandra Cabassi
- grid.5335.00000000121885934MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - John J. Lambourne
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Frances Burden
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Samantha Farrow
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Harriet McKinney
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Joana Batista
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carly Kempster
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Maik Pietzner
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Oliver Slingsby
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Thong Huy Cao
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Paulene A. Quinn
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Luca Stefanucci
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK ,British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew C. Sims
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK ,grid.454382.c0000 0004 7871 7212Oxford Haemophilia and Thrombosis Centre, Oxford University Hospitals NHS Foundation Trust, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Karola Rehnstrom
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Claire L. Adams
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ UK
| | - Amy Frary
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Bekir Ergüener
- grid.418729.10000 0004 0392 6802CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Roman Kreuzhuber
- grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.225360.00000 0000 9709 7726European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Gabriele Mocciaro
- grid.5335.00000000121885934Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA UK
| | - Simona D’Amore
- grid.24029.3d0000 0004 0383 8386Addenbrooke’s Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK ,grid.7644.10000 0001 0120 3326Department of Medicine, Aldo Moro University of Bari, Piazza Giulio Cesare 11, 70124 Bari, Italy ,National Cancer Research Center, IRCCS Istituto Tumori ‘Giovanni Paolo II’, Viale Orazio Flacco, 65, 70124 Bari, Italy
| | - Albert Koulman
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK ,grid.415055.00000 0004 0606 2472MRC Elsie Widdowson Laboratory, Cambridge, UK ,grid.5335.00000000121885934National Institute for Health Research Biomedical Research Centres Core Nutritional Biomarker Laboratory, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK ,grid.5335.00000000121885934National Institute for Health Research Biomedical Research Centres Core Metabolomics and Lipidomics Laboratory, Addenbrooke’s Hospital, University of Cambridge, Cambridge, UK
| | - Luigi Grassi
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.436365.10000 0000 8685 6563NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Julian L. Griffin
- grid.5335.00000000121885934Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA UK
| | - Leong Loke Ng
- grid.9918.90000 0004 1936 8411Department of Cardiovascular Sciences, Glenfield Hospital, University of Leicester, Leicester, UK ,grid.412925.90000 0004 0400 6581National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Adrian Park
- grid.24029.3d0000 0004 0383 8386Addenbrooke’s Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David B. Savage
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ UK
| | - Claudia Langenberg
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Christoph Bock
- grid.418729.10000 0004 0392 6802CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria ,grid.511293.d0000 0004 6104 8403Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria ,grid.22937.3d0000 0000 9259 8492Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Kate Downes
- grid.24029.3d0000 0004 0383 8386National Institute for Health Research BioResource, Cambridge University Hospitals, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.24029.3d0000 0004 0383 8386East Midlands and East of England Genomic Laboratory Hub, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Nicholas J. Wareham
- grid.5335.00000000121885934MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Michael Allison
- grid.24029.3d0000 0004 0383 8386Addenbrooke’s Hospital, NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Michele Vacca
- grid.5335.00000000121885934Metabolic Research Laboratories, Wellcome Trust-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ UK ,grid.5335.00000000121885934Department of Biochemistry and the Cambridge Systems Biology Centre, University of Cambridge, The Sanger Building, 80 Tennis Court Road, Cambridge, CB2 1GA UK
| | - Paul D. W. Kirk
- grid.5335.00000000121885934MRC Biostatistics Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK ,grid.5335.00000000121885934Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge Biomedical Campus, Puddicombe Way, Cambridge, CB2 0AW UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK. .,NHS Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK. .,British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge, UK. .,Institute of Biomedical & Clinical Science, College of Medicine and Health, University of Exeter Medical School, RILD Building, Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
15
|
Dahdah A, Johnson J, Gopalkrishna S, Jaggers RM, Webb D, Murphy AJ, Hanssen NMJ, Hanaoka BY, Nagareddy PR. Neutrophil Migratory Patterns: Implications for Cardiovascular Disease. Front Cell Dev Biol 2022; 10:795784. [PMID: 35309915 PMCID: PMC8924299 DOI: 10.3389/fcell.2022.795784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/18/2022] [Indexed: 12/31/2022] Open
Abstract
The body's inflammatory response involves a series of processes that are necessary for the immune system to mitigate threats from invading pathogens. Leukocyte migration is a crucial process in both homeostatic and inflammatory states. The mechanisms involved in immune cell recruitment to the site of inflammation are numerous and require several cascades and cues of activation. Immune cells have multiple origins and can be recruited from primary and secondary lymphoid, as well as reservoir organs within the body to generate an immune response to certain stimuli. However, no matter the origin, an important aspect of any inflammatory response is the web of networks that facilitates immune cell trafficking. The vasculature is an important organ for this trafficking, especially during an inflammatory response, mainly because it allows cells to migrate towards the source of insult/injury and serves as a reservoir for leukocytes and granulocytes under steady state conditions. One of the most active and vital leukocytes in the immune system's arsenal are neutrophils. Neutrophils exist under two forms in the vasculature: a marginated pool that is attached to the vessel walls, and a demarginated pool that freely circulates within the blood stream. In this review, we seek to present the current consensus on the mechanisms involved in leukocyte margination and demargination, with a focus on the role of neutrophil migration patterns during physio-pathological conditions, in particular diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Albert Dahdah
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jillian Johnson
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sreejit Gopalkrishna
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Robert M. Jaggers
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Darren Webb
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Andrew J. Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Nordin M. J. Hanssen
- Amsterdam Diabetes Centrum, Internal and Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Beatriz Y. Hanaoka
- Department of Internal Medicine, Division of Rheumatology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Prabhakara R. Nagareddy
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
16
|
Loewenstein F, Becker S, Kuehling J, Schrade H, Lechner M, Ringseis R, Eder K, Moritz A, Reiner G. Inflammation and necrosis syndrome is associated with alterations in blood and metabolism in pigs. BMC Vet Res 2022; 18:50. [PMID: 35045844 PMCID: PMC8767723 DOI: 10.1186/s12917-021-03107-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Swine inflammation and necrosis syndrome (SINS) can lead to significant clinical alterations at tail, ears, claws and other parts of the body in suckling piglets, weaners and fatteners. Clinical findings are associated with vasculitis, intima proliferation and thrombosis. The syndrome can be found in newborns, indicating a primarily endogenous aetiology. It has been hypothesized that SINS is triggered by gut-derived microbial-associated molecular patterns, causing derangements in liver metabolism and activity of peripheral white blood cells involving inflammation and blood haemostasis. In order to characterize these metabolic derangements of SINS for the first time, red and white blood counts, parameters of blood haemostasis, serum metabolites and acute phase proteins in the serum were analysed in 360 piglets, weaners and fatteners, each with significantly different SINS scores. RESULTS SINS scores and haematological/clinical chemical parameters were significantly associated (P < 0.05), especially in weaners and fatteners. Higher degrees of clinical SINS were associated with increased numbers of monocytes and neutrophils. Blood coagulation was altered in weaners and a thrombocytopenia was found in fatteners. Additionally, acute phase proteins, especially C-reactive protein and fibrinogen were increased in serum. Serum metabolites and serum liver enzymes were slightly altered. Aspartate transaminase levels overall exceeded physiological limit and increased in parallel with SINS scores in fatteners. CONCLUSION Clinical inflammation and necrosis at tail, ears, claws and other parts of the body were significantly associated with haematology and serum clinical chemistry, especially in weaners and fatteners. The involvement of inflammatory cells, blood coagulation, acute phase proteins and certain serum metabolites support the inflammatory-necrotising character of the syndrome and provide starting points for further studies to decipher its exact pathogenesis. The low to moderate variations seem less suitable for diagnostic use.
Collapse
Affiliation(s)
- Frederik Loewenstein
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
- LSZ Boxberg, Seehöfer Str. 50, 97944, Boxberg, Germany
| | - Sabrina Becker
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
| | - Josef Kuehling
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany
| | | | | | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus Liebig University Giessen, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Andreas Moritz
- Department of Veterinary Clinical Sciences, Clinic for Small Animals, Justus-Liebig-University, Frankfurter Strasse, 35392, Giessen, Germany
| | - Gerald Reiner
- Department of Veterinary Clinical Sciences, Clinic for Swine, Justus-Liebig-University, Frankfurter Strasse 112, 35392, Giessen, Germany.
| |
Collapse
|
17
|
Charles E, Dumont BL, Bonneau S, Neagoe PE, Villeneuve L, Räkel A, White M, Sirois MG. Angiopoietin 1 release from human neutrophils is independent from neutrophil extracellular traps (NETs). BMC Immunol 2021; 22:51. [PMID: 34344299 PMCID: PMC8336418 DOI: 10.1186/s12865-021-00442-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022] Open
Abstract
Background Neutrophils induce the synthesis and release of angiopoietin 1 (Ang1), a cytosolic growth factor involved in angiogenesis and capable of inducing several pro-inflammatory activities in neutrophils. Neutrophils also synthesize and release neutrophil extracellular traps (NETs), comprised from decondensed nuclear DNA filaments carrying proteins such as neutrophil elastase (NE), myeloperoxidase (MPO), proteinase 3 (PR3) and calprotectin (S100A8/S100A9), which together, contribute to the innate immune response against pathogens (e.g., bacteria). NETs are involved in various pathological conditions through pro-inflammatory, pro-thrombotic and endothelial dysfunction effects and have recently been found in heart failure (HF) and type 2 diabetes (T2DM) patients. The aim of the present study was to investigate the role of NETs on the synthesis and release of Ang1 by the neutrophils in patients with T2DM and HF with preserved ejection fraction (HFpEF) (stable or acute decompensated; ADHFpEF) with or without T2DM. Results Our data show that at basal level (PBS) and upon treatment with LPS, levels of NETs are slightly increased in patients suffering from T2DM, HFpEF ± T2DM and ADHF without (w/o) T2DM, whereas this increase was significant in ADHFpEF + T2DM patients compared to healthy control (HC) volunteers and ADHFpEF w/o T2DM. We also observed that treatments with PMA or A23187 increase the synthesis of Ang1 (from 150 to 250%) in HC and this effect is amplified in T2DM and in all cohorts of HF patients. Ang1 is completely released (100%) by neutrophils of all groups and does not bind to NETs as opposed to calprotectin. Conclusions Our study suggests that severely ill patients with HFpEF and diabetes synthesize and release a greater abundance of NETs while Ang1 exocytosis is independent of NETs synthesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00442-8.
Collapse
Affiliation(s)
- Elcha Charles
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada
| | - Benjamin L Dumont
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada
| | - Steven Bonneau
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada.,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada
| | - Paul-Eduard Neagoe
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada
| | - Agnès Räkel
- Department of Medicine, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, and Research Center-Centre Hospitalier de l'Université de Montréal (CHUM), Université de Montréal, Montreal, QC, Canada
| | - Michel White
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada. .,Department of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Martin G Sirois
- Research Center, Montreal Heart Institute, Université de Montréal, 5000 Belanger Street, Montreal, QC, H1T 1C8, Canada. .,Department of Pharmacology and Physiology , Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
18
|
Huang M, Ye K, Hu T, Liu K, You M, Wang L, Qin H. Silver Nanoparticles Attenuate the Antimicrobial Activity of the Innate Immune System by Inhibiting Neutrophil-Mediated Phagocytosis and Reactive Oxygen Species Production. Int J Nanomedicine 2021; 16:1345-1360. [PMID: 33633450 PMCID: PMC7901559 DOI: 10.2147/ijn.s292482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/15/2021] [Indexed: 11/23/2022] Open
Abstract
PURPOSE Despite the extensive development of antibacterial biomaterials, there are few reports on the effects of materials on the antibacterial ability of the immune system, and in particular of neutrophils. In this study, we observe differences between the in vivo and in vitro anti-infective efficacies of silver nanoparticles (AgNPs). The present study was designed to further explore the mechanism for this inconsistency using ex vivo models and in vitro experiments. METHODS AgNPs were synthesized using the polyol process and characterized by transmission electron microscopy and X-ray photoelectron spectroscopy. The antibacterial ability of AgNPs and neutrophils was tested by the spread-plate method. The infected air pouch model was prepared to detect the antimicrobial ability of AgNPs in vivo. Furthermore, blood-AgNPs-bacteria co-culture model and reactive oxygen species (ROS) measurement were used to evaluate the effect of AgNPs to neutrophil-mediated phagocytosis and ROS production. RESULTS The antibacterial experiments in vitro showed that AgNPs had superior antibacterial properties in cell compatible concentration. While, AgNPs had no significant antibacterial effect in vivo, and pathological section in AgNPs group indicated less neutrophil infiltration in inflammatory site than S. aureus group. Furthermore, AgNPs were found to reduce the phagocytosis of neutrophils and inhibit their ability to produce ROS and superoxide during ex vivo and in vitro experiments. CONCLUSION This study selects AgNPs as the representative of inorganic nano-biomaterials and reveals the phenomenon and the mechanism underlying the significant AgNPs-induced inhibition of the antibacterial ability of neutrophils, and may have a certain enlightening effect on the development of biomaterials in the future. In the fabrication of antibacterial biomaterials, however, attention should be paid to both cell and immune system safety to make the antibacterial properties of the biomaterials and innate immune system complement each other and jointly promote the host's ability to resist the invasion of pathogenic microorganisms.
Collapse
Affiliation(s)
- Moran Huang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Kai Ye
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Tu Hu
- Department of Musculoskeletal Surgery, Fudan University Shanghai Cancer Center, Shanghai, People’s Republic of China
| | - Kexin Liu
- Department of General Practice, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Mengzhen You
- Department of General Practice, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Lei Wang
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| | - Hui Qin
- Department of Orthopaedics, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, People’s Republic of China
| |
Collapse
|
19
|
Gjorevski N, Avignon B, Gérard R, Cabon L, Roth AB, Bscheider M, Moisan A. Neutrophilic infiltration in organ-on-a-chip model of tissue inflammation. LAB ON A CHIP 2020; 20:3365-3374. [PMID: 32761043 DOI: 10.1039/d0lc00417k] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The multiphasic etiology of tissue inflammation and the fundamental immunological differences between species render inflammatory pathologies difficult to recapitulate in animal models, and account for the paucity of therapies that are successfully translated from rodents to humans. Here, we present a human-relevant organ-on-a-chip platform for experimental inflammatory diseases. We created an immunocompetent in vitro gut model by incorporating intestinal epithelial and immune cells into microfluidic chambers that permit cell movement across an extracellular matrix (ECM) and fluidic channels. This is the first model that integrates a mucosal barrier, a three-dimensional ECM, resident and infiltrating immune cells, and simulates a functional crosstalk that ultimately triggers cellular processes representative of inflammation. Under homeostatic conditions, enterocytes form a tight epithelium and subepithelial macrophages are non-activated. Introduction of pro-inflammatory mediators triggers macrophage activation and inflammation-induced intestinal barrier leakiness. Neutrophils in a parallel, matrix-separated non-epithelial channel are attracted by such a pro-inflammatory microenvironment and migrate through the extracellular matrix, further exacerbating tissue inflammation and damage. With this model, we provide the foundations to recapitulate and investigate the onset of tissue inflammation in a controlled, human-relevant system.
Collapse
Affiliation(s)
- Nikolce Gjorevski
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
20
|
Predictors of hernia after loop ileostomy closure: a single-center retrospective review. Int J Colorectal Dis 2020; 35:1695-1702. [PMID: 32451647 DOI: 10.1007/s00384-020-03637-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2020] [Indexed: 02/04/2023]
Abstract
PURPOSE Our aims were to determine the rate of incisional hernia after closure of a loop ileostomy (LI) and to identify any perioperative risk factors that may be associated with hernia development. METHODS We performed an IRB-approved retrospective review that included consecutive patients who underwent LI closure from January 1, 2012, to December 31, 2014. The primary outcome was identification of hernia on physical exam or by abdominal imaging. A stepwise logistic regression analysis was used to determine predictors of ileostomy site hernia. RESULTS A total of 243 consecutive patients had LI closure during the study timeframe. The overall rate of hernia formation was 11.9% (29/243). The median time to discovery of the hernia was 16.4 (range, 2.2-55.9) months with a median follow-up time of 49 months. Although LI performed in conjunction with sigmoidectomy for diverticulitis consisted of only 19% of ileostomy cases, 62% of hernias were (18/29) identified in this cohort. Rates of hernia formation increased with respect to body mass index (underweight 0%; normal 4.6%; overweight 13.8%; obese 21%). The multivariable model identified increased BMI (underweight/normal weight versus overweight: OR 4.13, 95% CI 1.06-16.04; underweight/normal weight versus obese: OR 8.74, 95% CI 2.17-35.23) and surgical indication (reference variable: diverticulitis; rectal cancer: OR 0.21, 95% CI 0.06-0.76; Crohn's/ulcerative colitis: OR 0.06, 95% CI 0.01-0.53; other: OR 0.15, 95% CI 0.04-0.64) as significant predictors of ileostomy site hernia. CONCLUSION The development of a hernia at the site of LI closure was influenced mostly by BMI, surgical indication, and operative time.
Collapse
|
21
|
Clauder AK, Kordowski A, Bartsch YC, Köhl G, Lilienthal GM, Almeida LN, Lindemann T, Petry J, Rau CN, Gramalla-Schmitz A, Dühring L, Elbracht C, Kenno S, Tillmann J, Wuhrer M, Ludwig RJ, Ibrahim SM, Bieber K, Köhl J, Ehlers M, Manz RA. IgG Fc N-Glycosylation Translates MHCII Haplotype into Autoimmune Skin Disease. J Invest Dermatol 2020; 141:285-294. [PMID: 32653301 DOI: 10.1016/j.jid.2020.06.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/19/2020] [Accepted: 06/03/2020] [Indexed: 11/29/2022]
Abstract
The major histocompatibility complex haplotype represents the most prevalent genetic risk factor for the development of autoimmune diseases. However, the mechanisms by which major histocompatibility complex-associated genetic susceptibility translates into autoimmune disease are not fully understood. Epidermolysis bullosa acquisita is an autoimmune skin-blistering disease driven by autoantibodies to type VII collagen. Here, we investigated autoantigen-specific plasma cells, CD4+ T cells, and IgG fraction crystallizable glycosylation in murine epidermolysis bullosa acquisita in congenic mouse strains with the disease-permitting H2s or disease-nonpermitting H2b major histocompatibility complex II haplotypes. Mice with an H2s haplotype showed increased numbers of autoreactive CD4+ T cells and elevated IL-21 and IFN-γ production, associated with a higher frequency of IgG autoantibodies with an agalactosylated, proinflammatory N-glycan moiety. Mechanistically, we show that the altered antibody glycosylation leads to increased ROS release from neutrophils, the main drivers of autoimmune inflammation in this model. These results indicate that major histocompatibility complex II-associated susceptibility to autoimmune diseases acuminates in a proinflammatory IgG fraction crystallizable N-glycosylation pattern and provide a mechanistic link to increased ROS release by neutrophils.
Collapse
Affiliation(s)
- Ann-Katrin Clauder
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Anna Kordowski
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany; Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Yannic C Bartsch
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Gabriele Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Gina-Maria Lilienthal
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Larissa N Almeida
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Timo Lindemann
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Janina Petry
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Christina N Rau
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | | | - Lara Dühring
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Claudia Elbracht
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Samyr Kenno
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Jenny Tillmann
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Saleh M Ibrahim
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Marc Ehlers
- Laboratories of Immunology and Antibody Glycan Analysis, Institute for Nutritional Medicine, University of Lübeck and University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany.
| |
Collapse
|
22
|
Analysis of Biochemical Parameters in Children with Chronic Tonsillitis. CURRENT HEALTH SCIENCES JOURNAL 2020; 46:129-135. [PMID: 32874684 PMCID: PMC7445641 DOI: 10.12865/chsj.46.02.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/05/2020] [Indexed: 11/18/2022]
Abstract
Chronic tonsillitis are frequent in pediatric pathology with numerous involved risk factors and pathogenic mechanisms. In this study, epidemiological data and biochemical values addressed to inflammation and mineral, lipid and hepatic metabolism were analyzed for 37 children of school age with chronic tonsillitis. We found that in the majority of cases, chronic tonsillitis was associated with increasing number of blood circulating inflammatory cells, high values of transaminases, cholesterol, triglycerides and low values of procalcitonin, C-reactive protein, calcium, vitamin D and serum iron. The results indicated relations of the biochemical profile analyzed with risk factors and systemic mechanisms for initiation and maintenance of chronic tonsillitis, aspects that can be used to optimize the prognosis of chronic tonsillitis in children.
Collapse
|
23
|
Neutrophil swarming delays the growth of clusters of pathogenic fungi. Nat Commun 2020; 11:2031. [PMID: 32341348 PMCID: PMC7184738 DOI: 10.1038/s41467-020-15834-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Neutrophils employ several mechanisms to restrict fungi, including the action of enzymes such as myeloperoxidase (MPO) or NADPH oxidase, and the release of neutrophil extracellular traps (NETs). Moreover, they cooperate, forming “swarms” to attack fungi that are larger than individual neutrophils. Here, we designed an assay for studying how these mechanisms work together and contribute to neutrophil's ability to contain clusters of live Candida. We find that neutrophil swarming over Candida clusters delays germination through the action of MPO and NADPH oxidase, and restricts fungal growth through NET release within the swarm. In comparison with neutrophils from healthy subjects, those from patients with chronic granulomatous disease produce larger swarms against Candida, but their release of NETs is delayed, resulting in impaired control of fungal growth. We also show that granulocyte colony-stimulating factors (GCSF and GM-CSF) enhance swarming and neutrophil ability to restrict fungal growth, even during treatment with chemical inhibitors that disrupt neutrophil function. Neutrophils employ several mechanisms to control the growth of fungi, including enzymes, reactive oxygen species, extracellular traps, and formation of “swarms”. Here, Hopke et al. study how the different mechanisms work together, using an in vitro assay with human neutrophils and clusters of live Candida cells.
Collapse
|
24
|
The role of CXCR2 in acute inflammatory responses and its antagonists as anti-inflammatory therapeutics. Curr Opin Hematol 2020; 26:28-33. [PMID: 30407218 DOI: 10.1097/moh.0000000000000476] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW CXCR2 is key stimulant of immune cell migration and recruitment, especially of neutrophils. Alleviating excessive neutrophil accumulation and infiltration could prevent prolonged tissue damage in inflammatory disorders. This review focuses on recent advances in our understanding of the role of CXCR2 in regulating neutrophil migration and the use of CXCR2 antagonists for therapeutic benefit in inflammatory disorders. RECENT FINDINGS Recent studies have provided new insights into how CXCR2 signaling regulates hematopoietic cell mobilization and function in both health and disease. We also summarize several CXCR2 regulatory mechanisms during infection and inflammation such as via Wip1, T-bet, P-selectin glycoprotein ligand-1, granulocyte-colony-stimulating factor, and microbiome. Moreover, we provide an update of studies investigating CXCR2 blockade in the laboratory and in clinical trials. SUMMARY Neutrophil homeostasis, migration, and recruitment must be precisely regulated. The CXCR2 signaling pathway is a potential target for modifying neutrophil dynamics in inflammatory disorders. We discuss the recent clinical use of CXCR2 antagonists for controlling inflammation.
Collapse
|
25
|
Rossi B, Constantin G, Zenaro E. The emerging role of neutrophils in neurodegeneration. Immunobiology 2020; 225:151865. [DOI: 10.1016/j.imbio.2019.10.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/30/2019] [Indexed: 12/11/2022]
|
26
|
García-López JP, Vilos C, Feijóo CG. Zebrafish, a model to develop nanotherapeutics that control neutrophils response during inflammation. J Control Release 2019; 313:14-23. [PMID: 31622693 DOI: 10.1016/j.jconrel.2019.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 01/26/2023]
Abstract
Neutrophils are crucial modulators of the inflammation process, and their uncontrolled response worsens several chronic pathologies. The p38 mitogen-activated protein kinases (MAPKs) activity is critical for normal immune and inflammatory response through the regulation of pro-inflammatory cytokines synthesis. In this work, we study the effect of hybrid lipid-polymer nanoparticles loaded with the p38 MAPK inhibitor SB203580 in an acute and chronic inflammatory model in zebrafish containing a transgenic neutrophil cell line that constitutively expresses a green fluorescent protein. We identify the existence of at least two neutrophils subpopulation involved in the response during the acute inflammation triggered; a first-responder p38α-independent subset and a second-responder p38α-dependent subset. In the case of chronic inflammation, neutrophils recruited in the intestine only during the inflammation process, migrate in a p38α-dependent manner. Likewise, we establish that SB203580-loaded in NPs exerts their action during at least a double period than the inhibitor administers directly in both types of inflammation. Our results demonstrate the exceptional potential of the zebrafish as an inflammatory model for studying novel nanotherapeutics that selectively inhibit the neutrophils response, and to identify functional neutrophils subpopulations involved in the inflammation process.
Collapse
Affiliation(s)
- Juan P García-López
- Fish Immunology Laboratory, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile; Laboratory of Nanomedicine and Targeted Delivery, Center for Medical Research, School of Medicine, Universidad de Talca, 2 Norte 685, Talca 3460000, Chile
| | - Cristian Vilos
- Laboratory of Nanomedicine and Targeted Delivery, Center for Medical Research, School of Medicine, Universidad de Talca, 2 Norte 685, Talca 3460000, Chile; Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, 9170124, Santiago, Chile.
| | - Carmen G Feijóo
- Fish Immunology Laboratory, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
| |
Collapse
|
27
|
Oliveira AS, Nascimento JR, Trovão LO, Alves PCS, Maciel MCG, Silva LDM, Marques AA, Santos APSA, Silva LA, Nascimento FRF, Guerra RNM. The anti-inflammatory activity of Anacardium occidentale L. increases the lifespan of diabetic mice with lethal sepsis. JOURNAL OF ETHNOPHARMACOLOGY 2019; 236:345-353. [PMID: 30872173 DOI: 10.1016/j.jep.2019.03.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 02/14/2019] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Affiliation(s)
- Aluísio S Oliveira
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | - Johnny R Nascimento
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | - Liana O Trovão
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | - Patrícia C S Alves
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | | | - Luís Douglas M Silva
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | - André A Marques
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | - Ana Paula S A Santos
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | - Lucilene A Silva
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| | | | - Rosane N M Guerra
- Laboratory of Immunophysiology, Federal University of Maranhão, São Luís, Brazil.
| |
Collapse
|
28
|
Zhang C, Zhang L, Wu W, Gao F, Li RQ, Song W, Zhuang ZN, Liu CJ, Zhang XZ. Artificial Super Neutrophils for Inflammation Targeting and HClO Generation against Tumors and Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1901179. [PMID: 30924234 DOI: 10.1002/adma.201901179] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/13/2019] [Indexed: 05/22/2023]
Abstract
Neutrophils are powerful effector leukocytes that play an important role in innate immune systems for opposing tumor progression and ameliorating pathogen infections. Inspired by their distinct functions against tumors and infections, the artificial "super neutrophils" are proposed with excellent inflammation targeting and hypochlorous acid (HClO) generation characteristics for targeting and eliminating malignant tumor cells and pathogens. The "super neutrophils" are fabricated by embedding glucose oxidase (GOx) and chloroperoxidase (CPO) into zeolitic imidazolate framework-8 (ZIF-8) for HClO generation via enzymatic cascades, and then encapsulating them with the neutrophil membrane (NM) for inflammation targeting. In vitro and in vivo results indicate that these artificial "super neutrophils" can generate seven times higher reactive HClO than the natural neutrophils for eradicating tumors and infections. The "super neutrophils" demonstrated here with easy fabrication and good neutrophil-mimicking property exhibit great potential for biomedical applications.
Collapse
Affiliation(s)
- Chi Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Lu Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Wei Wu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Fan Gao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Run-Qing Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Wen Song
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Ze-Nan Zhuang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Chuan-Jun Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
29
|
Nachun D, Gao F, Isaacs C, Strawser C, Yang Z, Dokuru D, Van Berlo V, Sears R, Farmer J, Perlman S, Lynch DR, Coppola G. Peripheral blood gene expression reveals an inflammatory transcriptomic signature in Friedreich's ataxia patients. Hum Mol Genet 2019; 27:2965-2977. [PMID: 29790959 PMCID: PMC6097013 DOI: 10.1093/hmg/ddy198] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/17/2018] [Indexed: 12/15/2022] Open
Abstract
Transcriptional changes in Friedreich's ataxia (FRDA), a rare and debilitating recessive Mendelian neurodegenerative disorder, have been studied in affected but inaccessible tissues-such as dorsal root ganglia, sensory neurons and cerebellum-in animal models or small patient series. However, transcriptional changes induced by FRDA in peripheral blood, a readily accessible tissue, have not been characterized in a large sample. We used differential expression, association with disability stage, network analysis and enrichment analysis to characterize the peripheral blood transcriptome and identify genes that were differentially expressed in FRDA patients (n = 418) compared with both heterozygous expansion carriers (n = 228) and controls (n = 93 739 individuals in total), or were associated with disease progression, resulting in a disease signature for FRDA. We identified a transcriptional signature strongly enriched for an inflammatory innate immune response. Future studies should seek to further characterize the role of peripheral inflammation in FRDA pathology and determine its relevance to overall disease progression.
Collapse
Affiliation(s)
- Daniel Nachun
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Fuying Gao
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Charles Isaacs
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Zhongan Yang
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Deepika Dokuru
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Victoria Van Berlo
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Renee Sears
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Susan Perlman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - David R Lynch
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Giovanni Coppola
- Department of Psychiatry and Semel Institute, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
30
|
Tsutsui M, Hirase R, Miyamura S, Nagayasu K, Nakagawa T, Mori Y, Shirakawa H, Kaneko S. TRPM2 Exacerbates Central Nervous System Inflammation in Experimental Autoimmune Encephalomyelitis by Increasing Production of CXCL2 Chemokines. J Neurosci 2018; 38:8484-8495. [PMID: 30201769 PMCID: PMC6596171 DOI: 10.1523/jneurosci.2203-17.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 08/11/2018] [Accepted: 08/20/2018] [Indexed: 11/21/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the CNS characterized by demyelination and axonal injury. Current therapies that mainly target lymphocytes do not fully meet clinical need due to the risk of severe side effects and lack of efficacy against progressive MS. Evidence suggests that MS is associated with CNS inflammation, although the underlying molecular mechanism is poorly understood. Transient receptor potential melastatin 2 (TRPM2), a Ca2+-permeable nonselective cation channel, is expressed at high levels in the brain and by immune cells, including monocyte lineage cells. Here, we show that TRPM2 plays a pathological role in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Knockout (KO) or pharmacological inhibition of TRPM2 inhibited progression of EAE and TRPM2-KO mice showed lower activation of Iba1-immunopositive monocyte lineage cells and neutrophil infiltration of the CNS than WT mice. Moreover, CXCL2 production in TRPM2-KO mice was significantly reduced at day 14, although the severity of EAE was the same as that in WT mice at that time point. In addition, we used BM chimeric mice to show that TRPM2 expressed by CNS-infiltrating macrophages contributes to progression of EAE. Because CXCL2 induces migration of neutrophils, these results indicate that reduced expression of CXCL2 in the CNS suppresses neutrophil infiltration and slows progression of EAE in TRPM2-KO mice. Together, the results suggest that TRPM2 plays an important role in progression of EAE pathology and shed light on its putative role as a therapeutic target for MS.SIGNIFICANCE STATEMENT Current therapies for multiple sclerosis (MS), which mainly target lymphocytes, carry the risk of severe side effects and lack efficacy against the progressive form of the disease. Here, we found that the transient receptor potential melastatin 2 (TRPM2) channel, which is abundantly expressed in CNS-infiltrating macrophages, plays a crucial role in development of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. EAE progression was suppressed by Knockout (KO) or pharmacological inhibition of TRPM2; this was attributed to a reduction in CXCL2 chemokine production by CNS-infiltrating macrophages in TRPM2-KO mice, resulting in suppression of neutrophil infiltration into the CNS. These results reveal an important role of TRPM2 in the pathogenesis of EAE and shed light on its potential as a therapeutic target.
Collapse
Affiliation(s)
- Masato Tsutsui
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Ryo Hirase
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Sakie Miyamura
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Takayuki Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Sakyo-ku, Kyoto 606-8507, Japan, and
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura Campus, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan,
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
Ali M, Jasmin S, Fariduddin M, Alam SMK, Arslan MI, Biswas SK. Neutrophil elastase and myeloperoxidase mRNA expression in overweight and obese subjects. Mol Biol Rep 2018; 45:1245-1252. [PMID: 30056589 DOI: 10.1007/s11033-018-4279-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 07/23/2018] [Indexed: 12/19/2022]
Abstract
Neutrophil elastase and myeloperoxidase enzymes have been implicated in high-fat diet-induced obesity, insulin resistance (IR) and atherosclerosis in animal models. The aim of the present study was to explore neutrophil elastase and myeloperoxidase mRNA expressions in the peripheral blood leukocytes (PBL) in overweight and obese subjects, and to correlate those mRNA expressions with BMI, IR and cardiovascular biomarkers. In this cross-sectional study, 74 apparently healthy subjects including 22 lean, 27 overweight and 25 obese subjects were recruited. Cardiovascular and metabolic biomarkers were evaluated from fasting blood samples. The mRNA levels of neutrophil elastase and myeloperoxidase genes in the PBL were quantified by real-time PCR. Compared to lean group, the overweight and obese groups showed significant upregulation of both neutrophil elastase (p < 0.001) and myeloperoxidase (p < 0.03) mRNA expressions in the PBL. But no difference was found between overweight and obese groups. The neutrophil elastase and myeloperoxidase mRNA levels showed significant positive correlation with BMI, serum triglyceride, atherogenic index of plasma and 10-year risk of developing cardiovascular disease. But no correlation was found with glucose, insulin or IR. It was concluded that the neutrophil elastase and myeloperoxidase genes are up-regulated in both overweight and obese subjects and are associated with BMI and markers of cardiovascular disease.
Collapse
Affiliation(s)
- Mohammad Ali
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka - 1000, Bangladesh
| | - Shahana Jasmin
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka - 1000, Bangladesh
| | - Mohammad Fariduddin
- Department of Endocrinology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Dhaka, Bangladesh
| | - Sheikh M K Alam
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka - 1000, Bangladesh
| | - M I Arslan
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka - 1000, Bangladesh
| | - Subrata K Biswas
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujib Medical University (BSMMU), Shahbag, Dhaka - 1000, Bangladesh.
| |
Collapse
|
32
|
Franchin M, Freires IA, Lazarini JG, Nani BD, da Cunha MG, Colón DF, de Alencar SM, Rosalen PL. The use of Brazilian propolis for discovery and development of novel anti-inflammatory drugs. Eur J Med Chem 2018; 153:49-55. [DOI: 10.1016/j.ejmech.2017.06.050] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/07/2017] [Accepted: 06/23/2017] [Indexed: 01/13/2023]
|
33
|
Guo X, Fang X, He G, Zaman MH, Fei X, Qiao W, Deng G. The role of neutrophils in skin damage induced by tissue-deposited lupus IgG. Immunology 2018; 154:604-612. [PMID: 29450882 PMCID: PMC6050218 DOI: 10.1111/imm.12908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/17/2018] [Accepted: 02/05/2018] [Indexed: 01/13/2023] Open
Abstract
Skin injury is the second most common clinical manifestation in patients with systemic lupus erythematosus (SLE). Neutrophils are crucial effector cells in the immune system but the significance of neutrophils in the pathogenesis of SLE is not clear. This study is to explore the role of neutrophils in the skin damage of SLE. We used lupus-prone mice and a C57BL/6 mouse model of lupus serum IgG-induced skin inflammation to investigate the role of neutrophils in skin damage of SLE. We found that a few neutrophils infiltrated the inflammatory sites of skin in lupus-prone mice and the lupus-IgG-induced skin damage mouse model. Depletion of neutrophils did not affect the development of skin inflammation caused by lupus IgG, and lupus IgG can induce apoptosis of neutrophils. The apoptosis of neutrophils induced by lupus IgG is related to FcγRIII and Fas/Fas ligand pathways. Our study indicates that neutrophils are not major contributors in the skin damage caused by tissue-deposited lupus IgG but death of neutrophils caused by lupus IgG may provide a resource of a large amount of autoantigens in SLE.
Collapse
Affiliation(s)
- Xuanxuan Guo
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
| | - Xiang Fang
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
| | - Guodan He
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
| | - Muhammad Haidar Zaman
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
| | - Xibin Fei
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
| | - Wei Qiao
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
| | - Guo‐Min Deng
- Key Laboratory of Antibody Technology of National Health and Family Planning CommissionNanjing Medical UniversityNanjingChina
- First Affiliated HospitalNanjing Medical UniversityNanjingChina
- State Key Laboratory of Reproductive MedicineNanjing Medical UniversityNanjingChina
| |
Collapse
|
34
|
Honokiol suppresses formyl peptide-induced human neutrophil activation by blocking formyl peptide receptor 1. Sci Rep 2017; 7:6718. [PMID: 28751674 PMCID: PMC5532207 DOI: 10.1038/s41598-017-07131-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/23/2017] [Indexed: 11/24/2022] Open
Abstract
Formyl peptide receptor 1 (FPR1) mediates bacterial and mitochondrial N-formyl peptides-induced neutrophil activation. Therefore, FPR1 is an important therapeutic target for drugs to treat septic or sterile inflammatory diseases. Honokiol, a major bioactive compound of Magnoliaceae plants, possesses several anti-inflammatory activities. Here, we show that honokiol exhibits an inhibitory effect on FPR1 binding in human neutrophils. Honokiol inhibited superoxide anion generation, reactive oxygen species formation, and elastase release in bacterial or mitochondrial N-formyl peptides (FPR1 agonists)-activated human neutrophils. Adhesion of FPR1-induced human neutrophils to cerebral endothelial cells was also reduced by honokiol. The receptor-binding results revealed that honokiol repressed FPR1-specific ligand N-formyl-Nle-Leu-Phe-Nle-Tyr-Lys-fluorescein binding to FPR1 in human neutrophils, neutrophil-like THP-1 cells, and hFPR1-transfected HEK293 cells. However, honokiol did not inhibit FPR2-specific ligand binding to FPR2 in human neutrophils. Furthermore, honokiol inhibited FPR1 agonist-induced calcium mobilization as well as phosphorylation of p38 MAPK, ERK, and JNK in human neutrophils. In conclusion, our data demonstrate that honokiol may have therapeutic potential for treating FPR1-mediated inflammatory diseases.
Collapse
|
35
|
El-Benna J, Hurtado-Nedelec M, Marzaioli V, Marie JC, Gougerot-Pocidalo MA, Dang PMC. Priming of the neutrophil respiratory burst: role in host defense and inflammation. Immunol Rev 2017; 273:180-93. [PMID: 27558335 DOI: 10.1111/imr.12447] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neutrophils are the major circulating white blood cells in humans. They play an essential role in host defense against pathogens. In healthy individuals, circulating neutrophils are in a dormant state with very low efficiency of capture and arrest on the quiescent endothelium. Upon infection and subsequent release of pro-inflammatory mediators, the vascular endothelium signals to circulating neutrophils to roll, adhere, and cross the endothelial barrier. Neutrophils migrate toward the infection site along a gradient of chemo-attractants, then recognize and engulf the pathogen. To kill this pathogen entrapped inside the vacuole, neutrophils produce and release high quantities of antibacterial peptides, proteases, and reactive oxygen species (ROS). The robust ROS production is also called 'the respiratory burst', and the NADPH oxidase or NOX2 is the enzyme responsible for the production of superoxide anion, leading to other ROS. In vitro, several soluble and particulate agonists induce neutrophil ROS production. This process can be enhanced by prior neutrophil treatment with 'priming' agents, which alone do not induce a respiratory burst. In this review, we will describe the priming process and discuss the beneficial role of controlled neutrophil priming in host defense and the detrimental effect of excessive neutrophil priming in inflammatory diseases.
Collapse
Affiliation(s)
- Jamel El-Benna
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France
| | - Margarita Hurtado-Nedelec
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France.,Département d'Immunologie et d'Hématologie, UF Dysfonctionnements Immunitaires, HUPNVS, Hôpital Bichat, Paris, France
| | - Viviana Marzaioli
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France
| | - Jean-Claude Marie
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France.,Département d'Immunologie et d'Hématologie, UF Dysfonctionnements Immunitaires, HUPNVS, Hôpital Bichat, Paris, France
| | - Marie-Anne Gougerot-Pocidalo
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France.,Département d'Immunologie et d'Hématologie, UF Dysfonctionnements Immunitaires, HUPNVS, Hôpital Bichat, Paris, France
| | - Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Sorbonne Paris Cité, Laboratoire d'Excellence Inflamex, DHU FIRE, Faculté de Médecine, Site Xavier Bichat, Université Paris Diderot, Paris, France
| |
Collapse
|
36
|
Neutrophil migration in infection and wound repair: going forward in reverse. Nat Rev Immunol 2017; 16:378-91. [PMID: 27231052 DOI: 10.1038/nri.2016.49] [Citation(s) in RCA: 697] [Impact Index Per Article: 87.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neutrophil migration and its role during inflammation has been the focus of increased interest in the past decade. Advances in live imaging and the use of new model systems have helped to uncover the behaviour of neutrophils in injured and infected tissues. Although neutrophils were considered to be short-lived effector cells that undergo apoptosis in damaged tissues, recent evidence suggests that neutrophil behaviour is more complex and, in some settings, neutrophils might leave sites of tissue injury and migrate back into the vasculature. The role of reverse migration and its contribution to resolution of inflammation remains unclear. In this Review, we discuss the different cues within tissues that mediate neutrophil forward and reverse migration in response to injury or infection and the implications of these mechanisms to human disease.
Collapse
|
37
|
Gomes BS, Neto BPS, Lopes EM, Cunha FVM, Araújo AR, Wanderley CWS, Wong DVT, Júnior RCPL, Ribeiro RA, Sousa DP, Venes R Medeiros J, Oliveira RCM, Oliveira FA. Anti-inflammatory effect of the monoterpene myrtenol is dependent on the direct modulation of neutrophil migration and oxidative stress. Chem Biol Interact 2017; 273:73-81. [PMID: 28559105 DOI: 10.1016/j.cbi.2017.05.019] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 05/15/2017] [Accepted: 05/19/2017] [Indexed: 12/27/2022]
Abstract
Myrtenol is a bicyclic monoterpene with anti-inflammatory properties. However, the mechanisms involved are partially unknown. Here, we investigated the effect of myrtenol during experimental chronic arthritis and the possible modulating activity of oxidative stress and neutrophil migration. Complete Freund's Adjuvant (CFA)-sensitized rats were treated with vehicle (1 mL/kg, po), myrtenol (12.5, 25 or 50 mg/kg, po), indomethacin (10 mg/kg, po) or dexamethasone (0.4 mg/kg) followed by intra-articular injection of CFA (0.5 mg/mL, 50 μL per joint). Then, paw edema and articular incapacitation (paw elevation time) were evaluated for 14 days. On the last day, a blood concentration superoxide dismutase (SOD) and nitrite was determined. In another experimental setting, human neutrophils were incubated with vehicle (sterile saline, 1 mL) or myrtenol (10-100 ng/mL) and the in vitro chemotaxis to N-formylmethionine-leucyl-phenylalanine (fMLP) (10-7 M/well) was evaluated. In addition, antiinflammatory effect of myrtenol was investigated in carrageenan-induced peritonitis. We found that CFA induced a prominent paw swelling and incapacitation of the joint, which were significantly prevented by myrtenol (P < 0.05). In addition, blood accumulation nitrite was attenuated by myrtenol when compared with vehicle-treated CFA group (P < 0.05). Furthermore, plasma levels of SOD were significantly increased by myrtenol versus vehicle-treated CFA group (P < 0.05). Moreover, fMLP-triggered neutrophil chemotaxis and carrageenan-induced peritonitis were markedly prevented by myrtenol (P < 0.05). Therefore, myrtenol showed anti-inflammatory and antinociceptive effects on experimental chronic arthritis, which seems to be related to the direct modulation of neutrophil migration and antioxidant activity.
Collapse
Affiliation(s)
- Bruno S Gomes
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil.
| | - Benedito P S Neto
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Everton M Lopes
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Francisco V M Cunha
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Alyne R Araújo
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Carlos W S Wanderley
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Deysi V T Wong
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Roberto César P L Júnior
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Ronaldo A Ribeiro
- Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Damião P Sousa
- Department of Pharmaceutical Sciences, Federal University of Parnaíba, João Pessoa, Paraíba, Brazil
| | - Jand Venes R Medeiros
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Rita C M Oliveira
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| | - Francisco A Oliveira
- Medicinal Plants Research Center, Federal University of Piauí, Teresina, Piauí, Brazil
| |
Collapse
|
38
|
Pedraza-Zamora CP, Delgado-Domínguez J, Zamora-Chimal J, Becker I. Th17 cells and neutrophils: Close collaborators in chronicLeishmania mexicanainfections leading to disease severity. Parasite Immunol 2017; 39. [DOI: 10.1111/pim.12420] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 02/03/2017] [Indexed: 12/21/2022]
Affiliation(s)
- C. P. Pedraza-Zamora
- Facultad de Medicina; Unidad de Investigación en Medicina Experimental; Universidad Nacional Autónoma de México (UNAM); Hospital General de México; Ciudad de México México
| | - J. Delgado-Domínguez
- Facultad de Medicina; Unidad de Investigación en Medicina Experimental; Universidad Nacional Autónoma de México (UNAM); Hospital General de México; Ciudad de México México
| | - J. Zamora-Chimal
- Facultad de Medicina; Unidad de Investigación en Medicina Experimental; Universidad Nacional Autónoma de México (UNAM); Hospital General de México; Ciudad de México México
| | - I. Becker
- Facultad de Medicina; Unidad de Investigación en Medicina Experimental; Universidad Nacional Autónoma de México (UNAM); Hospital General de México; Ciudad de México México
| |
Collapse
|
39
|
Pietronigro EC, Della Bianca V, Zenaro E, Constantin G. NETosis in Alzheimer's Disease. Front Immunol 2017; 8:211. [PMID: 28303140 PMCID: PMC5332471 DOI: 10.3389/fimmu.2017.00211] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 02/15/2017] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder characterized by the progressive deterioration of cognitive functions. Its neuropathological features include amyloid-β (Aβ) accumulation, the formation of neurofibrillary tangles, and the loss of neurons and synapses. Neuroinflammation is a well-established feature of AD pathogenesis, and a better understanding of its mechanisms could facilitate the development of new therapeutic approaches. Recent studies in transgenic mouse models of AD have shown that neutrophils adhere to blood vessels and migrate inside the parenchyma. Moreover, studies in human AD subjects have also shown that neutrophils adhere and spread inside brain vessels and invade the parenchyma, suggesting these cells play a role in AD pathogenesis. Indeed, neutrophil depletion and the therapeutic inhibition of neutrophil trafficking, achieved by blocking LFA-1 integrin in AD mouse models, significantly reduced memory loss and the neuropathological features of AD. We observed that neutrophils release neutrophil extracellular traps (NETs) inside blood vessels and in the parenchyma of AD mice, potentially harming the blood–brain barrier and neural cells. Furthermore, confocal microscopy confirmed the presence of NETs inside the cortical vessels and parenchyma of subjects with AD, providing more evidence that neutrophils and NETs play a role in AD-related tissue destruction. The discovery of NETs inside the AD brain suggests that these formations may exacerbate neuro-inflammatory processes, promoting vascular and parenchymal damage during AD. The inhibition of NET formation has achieved therapeutic benefits in several models of chronic inflammatory diseases, including autoimmune diseases affecting the brain. Therefore, the targeting of NETs may delay AD pathogenesis and offer a novel approach for the treatment of this increasingly prevalent disease.
Collapse
Affiliation(s)
| | - Vittorina Della Bianca
- Department of Medicine, Section of General Pathology, University of Verona , Verona , Italy
| | - Elena Zenaro
- Department of Medicine, Section of General Pathology, University of Verona , Verona , Italy
| | - Gabriela Constantin
- Department of Medicine, Section of General Pathology, University of Verona , Verona , Italy
| |
Collapse
|
40
|
Severe Hemorrhagic Shock Induces Acute Activation and Expansion of IL-8+/IL-10+ Neutrophils with Enhanced Oxidative Reactivity in Non-Human Primates. Shock 2016; 46:129-36. [DOI: 10.1097/shk.0000000000000643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
41
|
Le Blon D, Hoornaert C, Detrez JR, Bevers S, Daans J, Goossens H, De Vos WH, Berneman Z, Ponsaerts P. Immune remodelling of stromal cell grafts in the central nervous system: therapeutic inflammation or (harmless) side-effect? J Tissue Eng Regen Med 2016; 11:2846-2852. [PMID: 27320821 DOI: 10.1002/term.2188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 02/03/2016] [Accepted: 03/14/2016] [Indexed: 12/13/2022]
Abstract
Over the past two decades, several cell types with fibroblast-like morphology, including mesenchymal stem/stromal cells, but also other adult, embryonic and extra-embryonic fibroblast-like cells, have been brought forward in the search for cellular therapies to treat severe brain injuries and/or diseases. Although current views in regenerative medicine are highly focused on the immune modulating and regenerative properties of stromal cell transplantation in vivo, many open questions remain regarding their true mode of action. In this perspective, this study integrates insights gathered over the past 10 years to formulate a unifying model of the cellular events that accompany fibroblast-like cell grafting in the rodent brain. Cellular interactions are discussed step-by-step, starting from the day of implantation up to 10 days after transplantation. During the short period that precedes stable settlement of autologous/syngeneic stromal cell grafts, there is a complex interplay between hypoxia-mediated cell death of grafted cells, neutrophil invasion, microglia and macrophage recruitment, astrocyte activation and neo-angiogenesis within the stromal cell graft site. Consequently, it is speculated that regenerative processes following cell therapeutic intervention in the CNS are not only modulated by soluble factors secreted by grafted stromal cells (bystander hypothesis), but also by in vivo inflammatory processes following stromal cell grafting. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Debbie Le Blon
- Laboratory of Experimental Haematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Chloé Hoornaert
- Laboratory of Experimental Haematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Jan R Detrez
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium.,Cell Systems and Cellular Imaging, Ghent University, Ghent, Belgium
| | - Sanne Bevers
- Laboratory of Experimental Haematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Haematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium.,Cell Systems and Cellular Imaging, Ghent University, Ghent, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Haematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Haematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
42
|
Powell DR, Huttenlocher A. Neutrophils in the Tumor Microenvironment. Trends Immunol 2015; 37:41-52. [PMID: 26700397 DOI: 10.1016/j.it.2015.11.008] [Citation(s) in RCA: 419] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/13/2023]
Abstract
Neutrophils are the first responders to sites of acute tissue damage and infection. Recent studies suggest that in addition to neutrophil apoptosis, resolution of neutrophil inflammation at wounds can be mediated by reverse migration from tissues and transmigration back into the vasculature. In settings of chronic inflammation, neutrophils persist in tissues, and this persistence has been associated with cancer progression. However, the role of neutrophils in the tumor microenvironment remains controversial, with evidence for both pro- and anti-tumor roles. Here we review the mechanisms that regulate neutrophil recruitment and resolution at sites of tissue damage, with a specific focus on the tumor microenvironment. We discuss the current understanding as to how neutrophils alter the tumor microenvironment to support or hinder cancer progression, and in this context outline gaps in understanding and important areas of inquiry.
Collapse
Affiliation(s)
- Davalyn R Powell
- Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
43
|
IL-10 mediates plasmacytosis-associated immunodeficiency by inhibiting complement-mediated neutrophil migration. J Allergy Clin Immunol 2015; 137:1487-1497.e6. [PMID: 26653800 DOI: 10.1016/j.jaci.2015.10.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 10/01/2015] [Accepted: 10/06/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Plasmacytosis (ie, an expansion of plasma cell populations to much greater than the homeostatic level) occurs in the context of various immune disorders and plasma cell neoplasia. This condition is often associated with immunodeficiency that causes increased susceptibility to severe infections. Yet a causative link between plasmacytosis and immunodeficiency has not been established. OBJECTIVE Because recent studies have identified plasma cells as a relevant source of the immunosuppressive cytokine IL-10, we sought to investigate the role of IL-10 during conditions of polyclonal and neoplastic plasmacytosis for the regulation of immunity and its effect on inflammation and immunodeficiency. METHODS We used flow cytometry, IL-10 reporter (Vert-X) and B cell-specific IL-10 knockout mice, migration assays, and antibody-mediated IL-10 receptor blockade to study plasmacytosis-associated IL-10 expression and its effect on inflammation and Streptococcus pneumoniae infection in mice. ELISA was used to quantify IL-10 levels in patients with myeloma. RESULTS IL-10 production was a common feature of normal and neoplastic plasma cells in mice, and IL-10 levels increased with myeloma progression in patients. IL-10 directly inhibited neutrophil migration toward the anaphylatoxin C5a and suppressed neutrophil-dependent inflammation in a murine model of autoimmune disease. MOPC.315.BM murine myeloma leads to an increased incidence of bacterial infection in the airways, which was reversed after IL-10 receptor blockade. CONCLUSION We provide evidence that plasmacytosis-associated overexpression of IL-10 inhibits neutrophil migration and neutrophil-mediated inflammation but also promotes immunodeficiency.
Collapse
|
44
|
Plesnila N. The immune system in traumatic brain injury. Curr Opin Pharmacol 2015; 26:110-7. [PMID: 26613129 DOI: 10.1016/j.coph.2015.10.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 01/21/2023]
Abstract
Traumatic brain injury (TBI) is the major cause of death in children and young adults and one of the major reasons for long-term disability worldwide, however, no specific clinical treatment option could be established so far. This is surprising since it is well known that following the initial mechanical damage to the brain a plethora of delayed processes are activated which ultimately result in additional brain damage. Among these secondary mechanisms, acute and chronic activation of the innate and adaptive immune system is increasingly believed to play an important role for the pathogenesis of TBI. Understanding these processes may results in new, clinically applicable therapeutic options for TBI patients.
Collapse
Affiliation(s)
- Nikolaus Plesnila
- Institute for Stroke and Dementia Research and Munich Cluster of System Neurology (Synergy), University of Munich Medical Center, Munich, Germany.
| |
Collapse
|
45
|
ROS production, intracellular HSP70 levels and their relationship in human neutrophils: effects of age. Oncotarget 2015; 5:11800-12. [PMID: 25514461 PMCID: PMC4322963 DOI: 10.18632/oncotarget.2856] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/02/2014] [Indexed: 12/16/2022] Open
Abstract
ROS production and intracellular HSP70 levels were measured in human neutrophils for three age groups: young (20-59 years), elders (60-89 years) and nonagenarians (90 years and older). Elders showed higher levels of spontaneous intracellular ROS content compared with young and nonagenarian groups, which had similar intracellular ROS levels. Zymosan-induced (non-spontaneous) extracellular ROS levels were also similar for young and nonagenarians but were lower in elders. However, spontaneous extracellular ROS production increased continuously with age. Correlation analysis revealed positive relationships between HSP70 levels and zymosan-stimulated ROS production in the elder group. This was consistent with a promoting role for HSP70 in ROS-associated neutrophils response to pathogens. No positive correlation between ROS production and intracellular HSP70 levels was found for groups of young people and nonagenarians. In contrast, significant negative correlations of some ROS and HSP70 characteriscics were found for neutrophils from young people and nonagenarians. The observed difference in ROS and HSP70 correlations in elders and nonagenarians might be associated with an increased risk of mortality in older individuals less than 90 years old.
Collapse
|
46
|
Dorgham K, Amoura Z, Parizot C, Arnaud L, Frances C, Pionneau C, Devilliers H, Pinto S, Zoorob R, Miyara M, Larsen M, Yssel H, Gorochov G, Mathian A. Ultraviolet light converts propranolol, a nonselective β-blocker and potential lupus-inducing drug, into a proinflammatory AhR ligand. Eur J Immunol 2015; 45:3174-87. [DOI: 10.1002/eji.201445144] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 06/30/2015] [Accepted: 09/07/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Karim Dorgham
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
| | - Zahir Amoura
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; institut E3M; Service de médecine interne 2; Centre de Référence National pour le Lupus et le Syndrome des Antiphospholipides; Paris France
| | - Christophe Parizot
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; Département d'immunologie; Paris France
| | - Laurent Arnaud
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; institut E3M; Service de médecine interne 2; Centre de Référence National pour le Lupus et le Syndrome des Antiphospholipides; Paris France
| | | | - Cédric Pionneau
- Sorbonne Universités, UPMC, Inserm UMS-29 Omique, Plateforme P3S, F-75013; Paris France
- Centre Hospitalier Universitaire de Dijon; Service de médecine interne 2 et centre d'investigation clinique; Dijon France
| | - Hervé Devilliers
- Centre Hospitalier Universitaire de Dijon; Service de médecine interne 2 et centre d'investigation clinique; Dijon France
| | - Sandra Pinto
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
| | - Rima Zoorob
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
| | - Makoto Miyara
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; institut E3M; Service de médecine interne 2; Centre de Référence National pour le Lupus et le Syndrome des Antiphospholipides; Paris France
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; Département d'immunologie; Paris France
| | - Martin Larsen
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
| | - Hans Yssel
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
| | - Guy Gorochov
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; Département d'immunologie; Paris France
| | - Alexis Mathian
- Sorbonne Universités; UPMC Univ Paris 06, Inserm UMRS1135; Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 83 Bd de l'hôpital; F-75013, Paris France
- AP-HP; Groupement Hospitalier Pitié-Salpêtrière; institut E3M; Service de médecine interne 2; Centre de Référence National pour le Lupus et le Syndrome des Antiphospholipides; Paris France
| |
Collapse
|
47
|
Huang X, Li J, Dorta-Estremera S, Di Domizio J, Anthony SM, Watowich SS, Popkin D, Liu Z, Brohawn P, Yao Y, Schluns KS, Lanier LL, Cao W. Neutrophils Regulate Humoral Autoimmunity by Restricting Interferon-γ Production via the Generation of Reactive Oxygen Species. Cell Rep 2015; 12:1120-32. [PMID: 26257170 DOI: 10.1016/j.celrep.2015.07.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 06/12/2015] [Accepted: 07/09/2015] [Indexed: 02/07/2023] Open
Abstract
Here, we examine the mechanism by which plasmacytoid dendritic cells (pDCs) and type I interferons promote humoral autoimmunity. In an amyloid-induced experimental autoimmune model, neutrophil depletion enhanced anti-nuclear antibody development, which correlated with heightened IFN-γ production by natural killer (NK) cells. IFN-α/β produced by pDCs activated NK cells via IL-15 induction. Neutrophils released reactive oxygen species (ROS), which negatively modulated the levels of IL-15, thereby inhibiting IFN-γ production. Mice deficient in NADPH oxidase 2 produced increased amounts of IFN-γ and developed augmented titers of autoantibodies. Both the pDC-IFN-α/β pathway and IFN-γ were indispensable in stimulating humoral autoimmunity. Male NZB/W F1 mice expressed higher levels of superoxide than their female lupus-prone siblings, and depletion of neutrophils resulted in spontaneous NK cell and autoimmune B cell activation. Our findings suggest a regulatory role for neutrophils in vivo and highlight the importance of an NK-IFN-γ axis downstream of the pDC-IFN-α/β pathway in systemic autoimmunity.
Collapse
Affiliation(s)
- Xinfang Huang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Jingjing Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephanie Dorta-Estremera
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Jeremy Di Domizio
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott M Anthony
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Daniel Popkin
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zheng Liu
- MedImmune, LLC, Gaithersburg, MD 20878, USA
| | | | - Yihong Yao
- MedImmune, LLC, Gaithersburg, MD 20878, USA
| | - Kimberly S Schluns
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology and Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Wei Cao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
48
|
Abstract
INTRODUCTION Every year recreational and professional sports cause thousands of lesions to muscle, ligament and tendon. Critical progress in biological interventions could meet a pressing health need to help athletes resume their activity levels. AREAS COVERED We perform a narrative review on platelet-rich plasma (PRP) therapies and muscle injuries. The field is eminently translational, thus besides clinical data we summarize experimental studies that bring meaningful biological insights on PRP effects. Some concepts regarding healing mechanisms are reviewed including innate immune response, myogenesis and fibrosis. It is commonly thought that PRPs are not uniform and cannot be assessed against each other, thus current PRP classifications are addressed. PRP effects also depend on the characteristics of the host tissue; therefore we focus on clinical muscle injury classifications. Controversial clinical findings are attributed to both the variability in PRP formulations and variability in the application protocols, so we discuss current clinical data in this basis. EXPERT OPINION Currently, there is little clinical evidence to support the use of PRP in skeletal muscle injuries. The future of PRP therapies relies not only in finding the best products, most appropriate indications and application protocols, but also in conceiving combination products. Moreover, as our understanding of healing mechanisms progresses, off-the-shelf allogenic PRP products could be part of the solution for sport injuries.
Collapse
Affiliation(s)
- Isabel Andia
- Cruces University Hospital, BioCruces Health Research Institute, Regenerative Medicine Laboratory , Barakaldo 48903 , Spain
| | | |
Collapse
|
49
|
Edwards JV, Prevost NT, French AD, Concha M, Condon BD. Kinetic and structural analysis of fluorescent peptides on cotton cellulose nanocrystals as elastase sensors. Carbohydr Polym 2015; 116:278-85. [DOI: 10.1016/j.carbpol.2014.04.067] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/18/2014] [Accepted: 04/20/2014] [Indexed: 11/26/2022]
|
50
|
Hwang TL, Aljuffali IA, Lin CF, Chang YT, Fang JY. Cationic additives in nanosystems activate cytotoxicity and inflammatory response of human neutrophils: lipid nanoparticles versus polymeric nanoparticles. Int J Nanomedicine 2015; 10:371-85. [PMID: 25609950 PMCID: PMC4294622 DOI: 10.2147/ijn.s73017] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
This report compares the effect of lipid and polymeric nanoparticles upon human neutrophils in the presence of cationic surfactants. Nanostructured lipid carriers and poly(lactic-co-glycolic) acid nanoparticles were manufactured as lipid and polymeric systems, respectively. Some cytotoxic and proinflammatory mediators such as lactate dehydrogenase (LDH), elastase, O2•−, and intracellular Ca2+ were examined. The nanoparticles showed a size of 170–225 nm. Incorporation of cetyltrimethylammonium bromide or soyaethyl morpholinium ethosulfate, the cationic surfactant, converted zeta potential from a negative to a positive charge. Nanoparticles without cationic surfactants revealed a negligible change on immune and inflammatory responses. Cationic surfactants in both nanoparticulate and free forms induced cell death and the release of mediators. Lipid nanoparticles generally demonstrated a greater response compared to polymeric nanoparticles. The neutrophil morphology observed by electron microscopy confirmed this trend. Cetyltrimethylammonium bromide as the coating material showed more significant activation of neutrophils than soyaethyl morpholinium ethosulfate. Confocal microscope imaging displayed a limited internalization of nanoparticles into neutrophils. It is proposed that cationic nanoparticles interact with the cell membrane, triggering membrane disruption and the following Ca2+ influx. The elevation of intracellular Ca2+ induces degranulation and oxidative stress. The consequence of these effects is cytotoxicity and cell death. Caution should be taken when selecting feasible nanoparticulate formulations and cationic additives for consideration of applicability and toxicity.
Collapse
Affiliation(s)
- Tsong-Long Hwang
- Cell Pharmacology Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan ; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Ibrahim A Aljuffali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Chwan-Fwu Lin
- Department of Cosmetic Science, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan ; Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan
| | - Yuan-Ting Chang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan ; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan ; Chang Gung Memorial Hospital, Kweishan, Taoyuan, Taiwan
| |
Collapse
|