1
|
Chu E, Mychasiuk R, Green TRF, Zamani A, Dill LK, Sharma R, Raftery AL, Tsantikos E, Hibbs ML, Semple BD. Regulation of microglial responses after pediatric traumatic brain injury: exploring the role of SHIP-1. Front Neurosci 2023; 17:1276495. [PMID: 37901420 PMCID: PMC10603304 DOI: 10.3389/fnins.2023.1276495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Severe traumatic brain injury (TBI) is the world's leading cause of permanent neurological disability in children. TBI-induced neurological deficits may be driven by neuroinflammation post-injury. Abnormal activity of SH2 domain-containing inositol 5' phosphatase-1 (SHIP-1) has been associated with dysregulated immunological responses, but the role of SHIP-1 in the brain remains unclear. The current study investigated the immunoregulatory role of SHIP-1 in a mouse model of moderate-severe pediatric TBI. Methods SHIP-1+/- and SHIP-1-/- mice underwent experimental TBI or sham surgery at post-natal day 21. Brain gene expression was examined across a time course, and immunofluorescence staining was evaluated to determine cellular immune responses, alongside peripheral serum cytokine levels by immunoassays. Brain tissue volume loss was measured using volumetric analysis, and behavior changes both acutely and chronically post-injury. Results Acutely, inflammatory gene expression was elevated in the injured cortex alongside increased IBA-1 expression and altered microglial morphology; but to a similar extent in SHIP-1-/- mice and littermate SHIP-1+/- control mice. Similarly, the infiltration and activation of CD68-positive macrophages, and reactivity of GFAP-positive astrocytes, was increased after TBI but comparable between genotypes. TBI increased anxiety-like behavior acutely, whereas SHIP-1 deficiency alone reduced general locomotor activity. Chronically, at 12-weeks post-TBI, SHIP-1-/- mice exhibited reduced body weight and increased circulating cytokines. Pro-inflammatory gene expression in the injured hippocampus was also elevated in SHIP-1-/- mice; however, GFAP immunoreactivity at the injury site in TBI mice was lower. TBI induced a comparable loss of cortical and hippocampal tissue in both genotypes, while SHIP-1-/- mice showed reduced general activity and impaired working memory, independent of TBI. Conclusion Together, evidence does not support SHIP-1 as an essential regulator of brain microglial morphology, brain immune responses, or the extent of tissue damage after moderate-severe pediatric TBI in mice. However, our data suggest that reduced SHIP-1 activity induces a greater inflammatory response in the hippocampus chronically post-TBI, warranting further investigation.
Collapse
Affiliation(s)
- Erskine Chu
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Deparment of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Tabitha R. F. Green
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, United States
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Larissa K. Dill
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Alfred Health, Prahran, VIC, Australia
| | - Rishabh Sharma
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - April L. Raftery
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Deparment of Neurology, Alfred Health, Prahran, VIC, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
2
|
Horner E, Lord JM, Hazeldine J. The immune suppressive properties of damage associated molecular patterns in the setting of sterile traumatic injury. Front Immunol 2023; 14:1239683. [PMID: 37662933 PMCID: PMC10469493 DOI: 10.3389/fimmu.2023.1239683] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Associated with the development of hospital-acquired infections, major traumatic injury results in an immediate and persistent state of systemic immunosuppression, yet the underlying mechanisms are poorly understood. Detected in the circulation in the minutes, days and weeks following injury, damage associated molecular patterns (DAMPs) are a heterogeneous collection of proteins, lipids and DNA renowned for initiating the systemic inflammatory response syndrome. Suggesting additional immunomodulatory roles in the post-trauma immune response, data are emerging implicating DAMPs as potential mediators of post-trauma immune suppression. Discussing the results of in vitro, in vivo and ex vivo studies, the purpose of this review is to summarise the emerging immune tolerising properties of cytosolic, nuclear and mitochondrial-derived DAMPs. Direct inhibition of neutrophil antimicrobial activities, the induction of endotoxin tolerance in monocytes and macrophages, and the recruitment, activation and expansion of myeloid derived suppressor cells and regulatory T cells are examples of some of the immune suppressive properties assigned to DAMPs so far. Crucially, with studies identifying the molecular mechanisms by which DAMPs promote immune suppression, therapeutic strategies that prevent and/or reverse DAMP-induced immunosuppression have been proposed. Approaches currently under consideration include the use of synthetic polymers, or the delivery of plasma proteins, to scavenge circulating DAMPs, or to treat critically-injured patients with antagonists of DAMP receptors. However, as DAMPs share signalling pathways with pathogen associated molecular patterns, and pro-inflammatory responses are essential for tissue regeneration, these approaches need to be carefully considered in order to ensure that modulating DAMP levels and/or their interaction with immune cells does not negatively impact upon anti-microbial defence and the physiological responses of tissue repair and wound healing.
Collapse
Affiliation(s)
- Emily Horner
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Janet M. Lord
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| | - Jon Hazeldine
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham, Birmingham, United Kingdom
| |
Collapse
|
3
|
Zhou H, Liu C, Hu F, Shen C, Shen B, He W, Du J. Increased levels of circulating granulocytic myeloid‑derived suppressor cells in lumbar disc herniation. Exp Ther Med 2023; 26:367. [PMID: 37408862 PMCID: PMC10318602 DOI: 10.3892/etm.2023.12066] [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: 11/11/2022] [Accepted: 03/10/2023] [Indexed: 07/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) expand when the body undergoes inflammatory diseases and chronic diseases. However, its role in intervertebral disc degeneration remains unclear. The present study aimed to characterize specific subsets of MDSCs as potential indicators of disease progression in patients with lumbar disc herniation (LDH). The Gene Expression Omnibus (GEO) database was used to analyze the changes in granulocyte MDSCs (G-MDSCs). Peripheral blood samples were collected from 40 patients with LDH and 15 healthy controls, and flow cytometry was used to characterize different subsets of MDSCs. All subjects underwent lumbar spine magnetic resonance imaging. Then, t-distributed stochastic neighborhood embedding and FlowSOM were used to analyze the data obtained by CytoFlex. The correlation between circulating MDSCs and the clinicopathological stage of LDH was then further analyzed. The GEO database predicted that G-MDSCs were highly expressed in patients with LDH. The frequency of circulating G-MDSCs increased with Pfirrmann stage III and IV, while the percentage of mononuclear MDSCs (M-MDSCs) only increased. Patient age and sex did not correlate with the frequency of circulating G-MDSCs and M-MDSCs. The results of the computer algorithm analysis were consistent with those of our manual gating. The present study showed that the occurrence of LDH led to changes in the MDSC subpopulation in the circulating peripheral blood of patients, and the frequency of circulating G-MDSCs in patients with clinical stage III and IV LDH increased with the degree of degeneration. The determination of G-MDSCs can be used as an auxiliary examination item for LDH.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Chang Liu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Fangfang Hu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Chunlin Shen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Bing Shen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wei He
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Juan Du
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Comprehensive Central Laboratory, School of Medicine, The Second Affiliated Hospital, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, P.R. China
| |
Collapse
|
4
|
Janakiram NB, Valerio MS, Goldman SM, Dearth CL. The Role of the Inflammatory Response in Mediating Functional Recovery Following Composite Tissue Injuries. Int J Mol Sci 2021; 22:ijms222413552. [PMID: 34948349 PMCID: PMC8705789 DOI: 10.3390/ijms222413552] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 01/08/2023] Open
Abstract
Composite tissue injuries (CTI) are common among US Military Service members during combat operations, and carry a high potential of morbidity. Furthermore, CTI are often complicated due to an altered wound healing response, resulting in part from a dysregulation of the innate and adaptive immune responses. Unlike normal wound healing, in CTI, disruptions occur in innate immune responses, altering neutrophil functions, macrophage activation and polarization, further impacting the functions of T regulatory cells. Additionally, the biological underpinnings of these unfavorable wound healing conditions are multifactorial, including various processes, such as: ischemia, hypoxia, low nutrient levels, and altered cell metabolic pathways, among others, all of which are thought to trigger anergy in immune cells and destabilize adaptive immune responses. As a result, impaired wound healing is common in CTI. Herein, we review the altered innate and adaptive immune cells and their metabolic status and responses following CTI, and discuss the role a multi-pronged immunomodulatory approach may play in facilitating improved outcomes for afflicted patients.
Collapse
Affiliation(s)
- Naveena B. Janakiram
- Research & Surveillance Division, DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD 20889, USA; (N.B.J.); (M.S.V.); (S.M.G.)
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Michael S. Valerio
- Research & Surveillance Division, DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD 20889, USA; (N.B.J.); (M.S.V.); (S.M.G.)
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Stephen M. Goldman
- Research & Surveillance Division, DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD 20889, USA; (N.B.J.); (M.S.V.); (S.M.G.)
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Christopher L. Dearth
- Research & Surveillance Division, DoD-VA Extremity Trauma and Amputation Center of Excellence, Bethesda, MD 20889, USA; (N.B.J.); (M.S.V.); (S.M.G.)
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
- Correspondence: ; Tel.: +1-(301)-319-2461
| |
Collapse
|
5
|
Finlay LDB, Conway Morris A, Deane AM, Wood AJT. Neutrophil kinetics and function after major trauma: A systematic review. World J Crit Care Med 2021; 10:260-277. [PMID: 34616661 PMCID: PMC8462018 DOI: 10.5492/wjccm.v10.i5.260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/18/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune dysfunction following major traumatic injury is complex and strongly associated with significant morbidity and mortality through the development of multiple organ dysfunction syndrome (MODS), persistent inflammation, immunosuppression, and catabolism syndrome and sepsis. Neutrophils are thought to be a pivotal mediator in the development of immune dysfunction.
AIM To provide a review with a systematic approach of the recent literature describing neutrophil kinetics and functional changes after major trauma in humans and discuss hypotheses as to the mechanisms of the observed neutrophil dysfunction in this setting.
METHODS Medline, Embase and PubMed were searched on January 15, 2021. Papers were screened by two reviewers and those included had their reference list hand searched for additional papers of interest. Inclusion criteria were adults > 18 years old, with an injury severity score > 12 requiring admission to an intensive care unit. Papers that analysed major trauma patients as a subgroup were included.
RESULTS Of 107 papers screened, 48 were included in the review. Data were heterogeneous and most studies had a moderate to significant risk of bias owing to their observational nature and small sample sizes. Key findings included a persistently elevated neutrophil count, stereotyped alterations in cell-surface markers of activation, and the elaboration of heterogeneous and immunosuppressive populations of cells in the circulation. Some of these changes correlate with clinical outcomes such as MODS and secondary infection. Neutrophil phenotype remains a promising avenue for the development of predictive markers for immune dysfunction.
CONCLUSION Understanding of neutrophil phenotypes after traumatic injury is expanding. A greater emphasis on incorporating functional and clinically significant markers, greater uniformity in study design and assessment of extravasated neutrophils may facilitate risk stratification in patients affected by major trauma.
Collapse
Affiliation(s)
- Liam DB Finlay
- Melbourne Medical School, University of Melbourne, Melbourne 3052, Victoria, Australia
| | - Andrew Conway Morris
- Department of Medicine, University of Cambridge, Cambridge 01223, United Kingdom
| | - Adam M Deane
- Centre for Integrated Critical Care, University of Melbourne, Parkville 3052, Victoria, Australia
- Intensive Care Unit, Royal Melbourne Hospital, Parkville 3052, Victoria, Australia
| | - Alexander JT Wood
- Centre for Integrated Critical Care, University of Melbourne, Parkville 3052, Victoria, Australia
- Intensive Care Unit, Royal Melbourne Hospital, Parkville 3052, Victoria, Australia
| |
Collapse
|
6
|
Dean MJ, Ochoa JB, Sanchez-Pino MD, Zabaleta J, Garai J, Del Valle L, Wyczechowska D, Baiamonte LB, Philbrook P, Majumder R, Vander Heide RS, Dunkenberger L, Thylur RP, Nossaman B, Roberts WM, Chapple AG, Wu J, Hicks C, Collins J, Luke B, Johnson R, Koul HK, Rees CA, Morris CR, Garcia-Diaz J, Ochoa AC. Severe COVID-19 Is Characterized by an Impaired Type I Interferon Response and Elevated Levels of Arginase Producing Granulocytic Myeloid Derived Suppressor Cells. Front Immunol 2021; 12:695972. [PMID: 34341659 PMCID: PMC8324422 DOI: 10.3389/fimmu.2021.695972] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 ranges from asymptomatic in 35% of cases to severe in 20% of patients. Differences in the type and degree of inflammation appear to determine the severity of the disease. Recent reports show an increase in circulating monocytic-myeloid-derived suppressor cells (M-MDSC) in severe COVID 19 that deplete arginine but are not associated with respiratory complications. Our data shows that differences in the type, function and transcriptome of granulocytic-MDSC (G-MDSC) may in part explain the severity COVID-19, in particular the association with pulmonary complications. Large infiltrates by Arginase 1+ G-MDSC (Arg+G-MDSC), expressing NOX-1 and NOX-2 (important for production of reactive oxygen species) were found in the lungs of patients who died from COVID-19 complications. Increased circulating Arg+G-MDSC depleted arginine, which impaired T cell receptor and endothelial cell function. Transcriptomic signatures of G-MDSC from patients with different stages of COVID-19, revealed that asymptomatic patients had increased expression of pathways and genes associated with type I interferon (IFN), while patients with severe COVID-19 had increased expression of genes associated with arginase production, and granulocyte degranulation and function. These results suggest that asymptomatic patients develop a protective type I IFN response, while patients with severe COVID-19 have an increased inflammatory response that depletes arginine, impairs T cell and endothelial cell function, and causes extensive pulmonary damage. Therefore, inhibition of arginase-1 and/or replenishment of arginine may be important in preventing/treating severe COVID-19.
Collapse
Affiliation(s)
- Matthew J. Dean
- Louisiana State University Cancer Center, New Orleans, LA, United States
| | - Juan B. Ochoa
- Department of Surgery, Ochsner Medical Center, New Orleans, LA, United States
| | - Maria Dulfary Sanchez-Pino
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Jovanny Zabaleta
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Pediatrics, LSU Health, New Orleans, LA, United States
| | - Jone Garai
- Louisiana State University Cancer Center, New Orleans, LA, United States
| | - Luis Del Valle
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Pathology LSU Health, New Orleans, LA, United States
| | | | | | - Phaethon Philbrook
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Rinku Majumder
- Department of Biochemistry, LSU Health, New Orleans, LA, United States
| | | | - Logan Dunkenberger
- Louisiana State University Cancer Center, New Orleans, LA, United States
| | | | - Bobby Nossaman
- Department of Surgery, Ochsner Medical Center, New Orleans, LA, United States
| | - W. Mark Roberts
- Department of Internal Medicine, Ochsner Medical Center, New Orleans, LA, United States
| | - Andrew G. Chapple
- Louisiana State University Cancer Center, New Orleans, LA, United States
- School of Public Health, LSU Health, New Orleans, LA, United States
| | - Jiande Wu
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Chindo Hicks
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Jack Collins
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Brian Luke
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Randall Johnson
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Hari K. Koul
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Biochemistry, LSU Health, New Orleans, LA, United States
| | - Chris A. Rees
- Division of Emergency Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Claudia R. Morris
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Julia Garcia-Diaz
- Tissue Biorepository, Ochsner Medical Center, New Orleans, LA, United States
| | - Augusto C. Ochoa
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Pediatrics, LSU Health, New Orleans, LA, United States
| |
Collapse
|
7
|
de Boniface J, Mao Y, Schmidt-Mende J, Kiessling R, Poschke I. Expression patterns of the immunomodulatory enzyme arginase 1 in blood, lymph nodes and tumor tissue of early-stage breast cancer patients. Oncoimmunology 2021; 1:1305-1312. [PMID: 23243594 PMCID: PMC3518503 DOI: 10.4161/onci.21678] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Arginase 1 (ARG1) is an important enzyme in amino acid metabolism that also exerts immunoregulatory function. High ARG1 expression, which is associated with cell cycle arrest and functional unresponsiveness in T cells, has been observed after trauma, infections and in cancer patients. We studied ARG1 expression in early-stage breast cancer patients (stage 1, n = 20; stage 2, n = 23) by multi-parametric flow cytometry and immunohistochemistry. Despite a low tumor burden, ARG1 expression was significantly increased in blood-derived myeloid cells of breast cancer patients compared with healthy controls. The ARG1hi myeloid population in the blood of cancer patients contained a high frequency of CD14+ cells and was, therefore, distinct from the granulocytic ARG1+ population observed in control individuals. Expression of ARG1 in patient blood cells correlated with tumor grade and was significantly reduced after surgical tumor removal. ARG1+ myeloid cells could also be detected in tumors and tumor-draining lymph nodes, where ARG1 expression levels exceeded those measured in the blood. We conclude that even patients with early-stage breast cancer exhibit tumor-related changes of ARG1 expression. The level of ARG1-mediated immunomodulation at this early stage remains to be determined. However, high ARG1 expression is likely to interfere with antitumor T-cell responses and immunotherapeutic interventions, making ARG1 or its downstream effector interesting therapeutic targets.
Collapse
Affiliation(s)
- Jana de Boniface
- Institution of Molecular Medicine and Surgery; Karolinska Institutet; Stockholm, Sweden ; Department of Breast and Endocrine Surgery; Karolinska University Hospital; Stockholm, Sweden
| | | | | | | | | |
Collapse
|
8
|
Zhang F, Liu F, Yu S, Zhang G, Li J, Sun X. Protective Effect of Curcumin on Bone Trauma in a Rat Model via Expansion of Myeloid Derived Suppressor Cells. Med Sci Monit 2020; 26:e924724. [PMID: 33184252 PMCID: PMC7670833 DOI: 10.12659/msm.924724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Bone fracture, a common injury to bones leads to various biophysiological changes and pathological responses in the body. The current study investigated curcumin for treatment of bone fracture in a rat model of bone trauma, and evaluated the related mechanism. MATERIAL AND METHODS The rats were separated randomly into 3 groups; sham, model, and curcumin treatment groups. The fracture rat model was established by transverse osteotomy in the right femur bone at the mid-shaft. The osteoblast count was determined using hematoxylin and eosin staining. Vascular endothelial growth factor (VEGF) and proliferating cell nuclear antigen (PCNA) expression were measured by western blotting. RESULTS The rpS6-phosphorylation was suppressed and light chain 3 (LC3II) expression elevated in the curcumin treated group of the fracture rat model. In the curcumin-treated group, mineralization of fracture calluses was markedly higher on day 14 of fracture. The formation of osteoblasts was observed at a greater rate in the curcumin treated group compared to the model rat group. Treatment of rats with curcumin significantly (P<0.05) promoted expression of PCNA and VEGF. The decrease in CD11b+/Gr-1+ cell expansion in rats with bone trauma was alleviated significantly by curcumin treatment. A marked increase in arginase-1 expression in rats with bone trauma was caused by curcumin treatment. CONCLUSIONS In summary, curcumin activates autophagy and inhibits mTOR activation in bone tissues of rats with trauma. The curcumin promoted myeloid-derived suppressor cell (MDSC) proliferation and increased expansion of MDSCs in a rat model of trauma. Therefore, curcumin may have beneficial effect in patients with bone trauma and should be evaluated further for development of treatment.
Collapse
Affiliation(s)
- Futian Zhang
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Fu Liu
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Shaofen Yu
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Guihong Zhang
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Jie Li
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| | - Xinjun Sun
- Department of Orthopaedic Trauma, The 80th Group Army Hospital of The People's Liberation Army of China, Weifang, Shandong, China (mainland)
| |
Collapse
|
9
|
Hassani M, Hellebrekers P, Chen N, van Aalst C, Bongers S, Hietbrink F, Koenderman L, Vrisekoop N. On the origin of low-density neutrophils. J Leukoc Biol 2020; 107:809-818. [PMID: 32170882 PMCID: PMC7318192 DOI: 10.1002/jlb.5hr0120-459r] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/31/2020] [Accepted: 02/17/2020] [Indexed: 01/01/2023] Open
Abstract
Here we elaborate on the origin of low(er)-density neutrophils (LDNs) to better understand the variation found in literature. Supplemented with original data, we test the hypothesis that buoyant density of neutrophils is characterized by a spectrum that as a whole shifts to a lower density after activation. Both the 20% highest density (HDNs) and 20% lowest density (LDNs) neutrophils from healthy donors were isolated by Percoll of different densities. Using this method we found that LDNs were significantly better in T-cell suppression and bacterial containment than their 20% highest density counterparts. We found no statistically relevant differences in neutrophil survival or bacterial phagocytosis. Stimulation of healthy donor neutrophils with N-formyl-methionyl-leucyl-phenylalanine induced LDNs co-segregating with peripheral blood mononuclear cells after Ficoll separation. These in vitro induced LDNs showed increased activation markers compared to HDNs and were comparable to the activation markers found on the LDN fraction seen in patients with chronic inflammatory conditions such as present in cancer patients. This all fits with the hypothesis that the density of neutrophils is distributed in a spectrum partially coupled to maturation. Additionally a shift in this spectrum can be induced by in vitro stimulation or by activation in disease.
Collapse
Affiliation(s)
- Marwan Hassani
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
| | - Pien Hellebrekers
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
- Department of SurgeryUniversity Medical Center UtrechtThe Netherlands
| | - Na Chen
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
| | - Corneli van Aalst
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
| | - Suus Bongers
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
- Department of SurgeryUniversity Medical Center UtrechtThe Netherlands
| | - Falco Hietbrink
- Department of SurgeryUniversity Medical Center UtrechtThe Netherlands
| | - Leo Koenderman
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
| | - Nienke Vrisekoop
- Department of Respiratory Medicine and Center for Translational ImmunologyUniversity Medical Center UtrechtThe Netherlands
| |
Collapse
|
10
|
Mortaz E, Zadian SS, Shahir M, Folkerts G, Garssen J, Mumby S, Adcock IM. Does Neutrophil Phenotype Predict the Survival of Trauma Patients? Front Immunol 2019; 10:2122. [PMID: 31552051 PMCID: PMC6743367 DOI: 10.3389/fimmu.2019.02122] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/23/2019] [Indexed: 12/14/2022] Open
Abstract
According to the World Health Organization (WHO), trauma is responsible for 10% of deaths and 16% of disabilities worldwide. This is considerably higher than those for malaria, tuberculosis, and HIV/AIDS combined. While the human suffering and death caused by injury is well-recognized, injury has a significant medical care cost. Better prediction of the state of trauma patients in the days immediately after trauma may reduce costs. Traumatic injuries to multiple organs can cause dysfunction in all systems of the body especially the immune system placing patients at high risk of infections and inflammatory complications which are often fatal. Neutrophils are the most abundant leukocyte in the human circulation and are crucial for the prevention of microbial disease. Significant changes in neutrophil functions such as enhanced chemotaxis, Neutrophil extracellular trap (NET)-induced cell death (NETosis), and phagocytosis occur early after injury followed by prolonged functional defects such as phagocytosis, killing mechanisms, and receptor expression. Analysis of these changes may improve the prediction of the patient's condition over time. We provide a comprehensive and up-to-date review of the literature investigating the effect of trauma on neutrophil phenotype with an underlying goal of using this knowledge to examine the predictive potential of neutrophil alterations on secondary complications in patients with traumatic injuries. We conclude that alterations in neutrophil surface markers and functions may be potential biomarkers that predict the outcome of trauma patients.
Collapse
Affiliation(s)
- Esmaeil Mortaz
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Sajjad Zadian
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehri Shahir
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Gert Folkerts
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands.,Nutricia Research Centre for Specialized Nutrition, Utrecht, Netherlands
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
11
|
Hesselink L, Spijkerman R, van Wessem KJP, Koenderman L, Leenen LPH, Huber-Lang M, Hietbrink F. Neutrophil heterogeneity and its role in infectious complications after severe trauma. World J Emerg Surg 2019; 14:24. [PMID: 31164913 PMCID: PMC6542247 DOI: 10.1186/s13017-019-0244-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Background Trauma leads to a complex inflammatory cascade that induces both immune activation and a refractory immune state in parallel. Although both components are deemed necessary for recovery, the balance is tight and easily lost. Losing the balance can lead to life-threatening infectious complications as well as long-term immunosuppression with recurrent infections. Neutrophils are known to play a key role in these processes. Therefore, this review focuses on neutrophil characteristics and function after trauma and how these features can be used to identify trauma patients at risk for infectious complications. Results Distinct neutrophil subtypes exist that play their own role in the recovery and/or development of infectious complications after trauma. Furthermore, the refractory immune state is related to the risk of infectious complications. These findings change the initial concepts of the immune response after trauma and give rise to new biomarkers for monitoring and predicting inflammatory complications in severely injured patients. Conclusion For early recognition of patients at risk, the immune system should be monitored. Several neutrophil biomarkers show promising results and analysis of these markers has become accessible to such extent that they can be used for point-of-care decision making after trauma.
Collapse
Affiliation(s)
- Lillian Hesselink
- Department of Trauma Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology and Department of Respiratory Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Roy Spijkerman
- Department of Trauma Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
- Laboratory of Translational Immunology and Department of Respiratory Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Leo Koenderman
- Laboratory of Translational Immunology and Department of Respiratory Medicine, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Luke P. H. Leenen
- Department of Trauma Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital of Ulm, Ulm, Germany
| | - Falco Hietbrink
- Department of Trauma Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
12
|
Cassetta L, Baekkevold ES, Brandau S, Bujko A, Cassatella MA, Dorhoi A, Krieg C, Lin A, Loré K, Marini O, Pollard JW, Roussel M, Scapini P, Umansky V, Adema GJ. Deciphering myeloid-derived suppressor cells: isolation and markers in humans, mice and non-human primates. Cancer Immunol Immunother 2019; 68:687-697. [PMID: 30684003 PMCID: PMC6447515 DOI: 10.1007/s00262-019-02302-2] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022]
Abstract
In cancer, infection and inflammation, the immune system's function can be dysregulated. Instead of fighting disease, immune cells may increase pathology and suppress host-protective immune responses. Myeloid cells show high plasticity and adapt to changing conditions and pathological challenges. Despite their relevance in disease pathophysiology, the identity, heterogeneity and biology of myeloid cells is still poorly understood. We will focus on phenotypical and functional markers of one of the key myeloid regulatory subtypes, the myeloid derived suppressor cells (MDSC), in humans, mice and non-human primates. Technical issues regarding the isolation of the cells from tissues and blood, timing and sample handling of MDSC will be detailed. Localization of MDSC in a tissue context is of crucial importance and immunohistochemistry approaches for this purpose are discussed. A minimal antibody panel for MDSC research is provided as part of the Mye-EUNITER COST action. Strategies for the identification of additional markers applying state of the art technologies such as mass cytometry will be highlighted. Such marker sets can be used to study MDSC phenotypes across tissues, diseases as well as species and will be crucial to accelerate MDSC research in health and disease.
Collapse
Affiliation(s)
- Luca Cassetta
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, EH16 4TJ, Edinburgh, UK.
| | - Espen S Baekkevold
- Centre for Immune Regulation, Department of Pathology, University of Oslo, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Sven Brandau
- West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anna Bujko
- Centre for Immune Regulation, Department of Pathology, University of Oslo, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Marco A Cassatella
- Division of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Anca Dorhoi
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany.,Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Carsten Krieg
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, USA
| | - Ang Lin
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Department of Medicine Solna, Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Olivia Marini
- Division of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, EH16 4TJ, Edinburgh, UK
| | - Mikael Roussel
- Centre Hospitalier Universitaire, Pôle Biologie, INSERM, UMR U1236, Université Rennes 1, EFS Bretagne, Rennes, France
| | - Patrizia Scapini
- Division of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Gosse J Adema
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Geert Grooteplein 28, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
13
|
Neutrophils in Tissue Trauma of the Skin, Bone, and Lung: Two Sides of the Same Coin. J Immunol Res 2018; 2018:8173983. [PMID: 29850639 PMCID: PMC5937416 DOI: 10.1155/2018/8173983] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 03/21/2018] [Indexed: 12/12/2022] Open
Abstract
Following severe tissue injury, patients are exposed to various danger- and microbe-associated molecular patterns, which provoke a strong activation of the neutrophil defense system. Neutrophils trigger and modulate the initial posttraumatic inflammatory response and contribute critically to subsequent repair processes. However, severe trauma can affect central neutrophil functions, including circulation half-life, chemokinesis, phagocytosis, cytokine release, and respiratory burst. Alterations in neutrophil biology may contribute to trauma-associated complications, including immune suppression, sepsis, multiorgan dysfunction, and disturbed tissue regeneration. Furthermore, there is evidence that neutrophil actions depend on the quality of the initial stimulus, including trauma localization and severity, the micromilieu in the affected tissue, and the patient's overall inflammatory status. In the present review, we describe the effects of severe trauma on the neutrophil phenotype and dysfunction and the consequences for tissue repair. We particularly concentrate on the role of neutrophils in wound healing, lung injury, and bone fractures, because these are the most frequently affected tissues in severely injured patients.
Collapse
|
14
|
MDSCs are induced after experimental blunt chest trauma and subsequently alter antigen-specific T cell responses. Sci Rep 2017; 7:12808. [PMID: 28993671 PMCID: PMC5634472 DOI: 10.1038/s41598-017-13019-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 09/12/2017] [Indexed: 01/27/2023] Open
Abstract
Severe blunt chest trauma (TxT) induces a strong inflammatory response with posttraumatic immune suppression pointing to an impaired adaptive immune response. Since CD11b+Gr-1+-expressing myeloid-derived suppressor cells (MDSCs) are induced after inflammation and suppress T cell responses, MDSC induction and their impact on T cell functions was analysed in an experimental TxT model. MDSCs were induced preferentially in the lung until 24 hours after TxT. Although MDSC numbers were only faintly increased in the spleen, splenic MDSCs isolated after TxT strongly inhibited alloantigen-induced T cell proliferation in vitro. Suppressive activity correlated with increased expression of arginase-1 and iNOS. MDSCs also prevented antigen-induced T cell expansion in vivo, since staphylococcus enterotoxin B (SEB)-induced proliferation of vβ8+ T cells was impaired in TxT mice in the presence of CD11b+Gr-1+ cells. Surprisingly, MDSCs were not involved in shifting T cells into Th2 cells, characterized by the secretion of cytokines impairing cell-mediated immunity and promoting immunosuppression. Instead, the presence of CD11b+Gr-1+ cells was required for efficient IL-2, IFN-γ and TNFα production after antigenic stimulation, indicating, that elevation of MDSCs early after traumatic injuries might contribute to restrict the initial inflammatory response by alleviating T cell expansion, however, without impeding Th1 functions.
Collapse
|
15
|
Aghaeepour N, Kin C, Ganio EA, Jensen KP, Gaudilliere DK, Tingle M, Tsai A, Lancero HL, Choisy B, McNeil LS, Okada R, Shelton AA, Nolan GP, Angst MS, Gaudilliere BL. Deep Immune Profiling of an Arginine-Enriched Nutritional Intervention in Patients Undergoing Surgery. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:2171-2180. [PMID: 28794234 PMCID: PMC5807249 DOI: 10.4049/jimmunol.1700421] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/11/2017] [Indexed: 01/08/2023]
Abstract
Application of high-content immune profiling technologies has enormous potential to advance medicine. Whether these technologies reveal pertinent biology when implemented in interventional clinical trials is an important question. The beneficial effects of preoperative arginine-enriched dietary supplements (AES) are highly context specific, as they reduce infection rates in elective surgery, but possibly increase morbidity in critically ill patients. This study combined single-cell mass cytometry with the multiplex analysis of relevant plasma cytokines to comprehensively profile the immune-modifying effects of this much-debated intervention in patients undergoing surgery. An elastic net algorithm applied to the high-dimensional mass cytometry dataset identified a cross-validated model consisting of 20 interrelated immune features that separated patients assigned to AES from controls. The model revealed wide-ranging effects of AES on innate and adaptive immune compartments. Notably, AES increased STAT1 and STAT3 signaling responses in lymphoid cell subsets after surgery, consistent with enhanced adaptive mechanisms that may protect against postsurgical infection. Unexpectedly, AES also increased ERK and P38 MAPK signaling responses in monocytic myeloid-derived suppressor cells, which was paired with their pronounced expansion. These results provide novel mechanistic arguments as to why AES may exert context-specific beneficial or adverse effects in patients with critical illness. This study lays out an analytical framework to distill high-dimensional datasets gathered in an interventional clinical trial into a fairly simple model that converges with known biology and provides insight into novel and clinically relevant cellular mechanisms.
Collapse
Affiliation(s)
- Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Cindy Kin
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94121
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Kent P Jensen
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94121; and
| | - Dyani K Gaudilliere
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94121
| | - Martha Tingle
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Amy Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Hope L Lancero
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Benjamin Choisy
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Leslie S McNeil
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Robin Okada
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Andrew A Shelton
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94121
| | - Garry P Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA 94121
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121
| | - Brice L Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94121;
| |
Collapse
|
16
|
Xu J, Guardado J, Hoffman R, Xu H, Namas R, Vodovotz Y, Xu L, Ramadan M, Brown J, Turnquist HR, Billiar TR. IL33-mediated ILC2 activation and neutrophil IL5 production in the lung response after severe trauma: A reverse translation study from a human cohort to a mouse trauma model. PLoS Med 2017; 14:e1002365. [PMID: 28742815 PMCID: PMC5526517 DOI: 10.1371/journal.pmed.1002365] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 06/20/2017] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The immunosuppression and immune dysregulation that follows severe injury includes type 2 immune responses manifested by elevations in interleukin (IL) 4, IL5, and IL13 early after injury. We hypothesized that IL33, an alarmin released early after tissue injury and a known regulator of type 2 immunity, contributes to the early type 2 immune responses after systemic injury. METHODS AND FINDINGS Blunt trauma patients admitted to the trauma intensive care unit of a level I trauma center were enrolled in an observational study that included frequent blood sampling. Dynamic changes in IL33 and soluble suppression of tumorigenicity 2 (sST2) levels were measured in the plasma and correlated with levels of the type 2 cytokines and nosocomial infection. Based on the observations in humans, mechanistic experiments were designed in a mouse model of resuscitated hemorrhagic shock and tissue trauma (HS/T). These experiments utilized wild-type C57BL/6 mice, IL33-/- mice, B6.C3(Cg)-Rorasg/sg mice deficient in group 2 innate lymphoid cells (ILC2), and C57BL/6 wild-type mice treated with anti-IL5 antibody. Severely injured human blunt trauma patients (n = 472, average injury severity score [ISS] = 20.2) exhibited elevations in plasma IL33 levels upon admission and over time that correlated positively with increases in IL4, IL5, and IL13 (P < 0.0001). sST2 levels also increased after injury but in a delayed manner compared with IL33. The increases in IL33 and sST2 were significantly greater in patients that developed nosocomial infection and organ dysfunction than similarly injured patients that did not (P < 0.05). Mechanistic studies were carried out in a mouse model of HS/T that recapitulated the early increase in IL33 and delayed increase in sST2 in the plasma (P < 0.005). These studies identified a pathway where IL33 induces ILC2 activation in the lung within hours of HS/T. ILC2 IL5 up-regulation induces further IL5 expression by CXCR2+ lung neutrophils, culminating in early lung injury. The major limitations of this study are the descriptive nature of the human study component and the impact of the potential differences between human and mouse immune responses to polytrauma. Also, the studies performed did not permit us to make conclusions about the impact of IL33 on pulmonary function. CONCLUSIONS These results suggest that IL33 may initiate early detrimental type 2 immune responses after trauma through ILC2 regulation of neutrophil IL5 production. This IL33-ILC2-IL5-neutrophil axis defines a novel regulatory role for ILC2 in acute lung injury that could be targeted in trauma patients prone to early lung dysfunction.
Collapse
Affiliation(s)
- Jing Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- State Key Laboratory of Trauma, Burns and Combined Injury, Second Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jesse Guardado
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Rosemary Hoffman
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Hui Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P. R. China
| | - Rami Namas
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Yoram Vodovotz
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Li Xu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Emergency Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Mostafa Ramadan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Joshua Brown
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Heth R. Turnquist
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (TRB); (HRT)
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (TRB); (HRT)
| |
Collapse
|
17
|
Hazeldine J, Lord JM, Belli A. Traumatic Brain Injury and Peripheral Immune Suppression: Primer and Prospectus. Front Neurol 2015; 6:235. [PMID: 26594196 PMCID: PMC4633482 DOI: 10.3389/fneur.2015.00235] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022] Open
Abstract
Nosocomial infections are a common occurrence in patients following traumatic brain injury (TBI) and are associated with an increased risk of mortality, longer length of hospital stay, and poor neurological outcome. Systemic immune suppression arising as a direct result of injury to the central nervous system (CNS) is considered to be primarily responsible for this increased incidence of infection, a view strengthened by recent studies that have reported novel changes in the composition and function of the innate and adaptive arms of the immune system post-TBI. However, our knowledge of the mechanisms that underlie TBI-induced immune suppression is equivocal at best. Here, after summarizing our current understanding of the impact of TBI on peripheral immunity and discussing CNS-mediated regulation of immune function, we propose roles for a series of novel mechanisms in driving the immune suppression that is observed post-TBI. These mechanisms, which have never been considered before in the context of TBI-induced immune paresis, include the CNS-driven emergence into the circulation of myeloid-derived suppressor cells and suppressive neutrophil subsets, and the release from injured tissue of nuclear and mitochondria-derived damage associated molecular patterns. Moreover, in an effort to further our understanding of the mechanisms that underlie TBI-induced changes in immunity, we pose throughout the review a series of questions, which if answered would address a number of key issues, such as establishing whether manipulating peripheral immune function has potential as a future therapeutic strategy by which to treat and/or prevent infections in the hospitalized TBI patient.
Collapse
Affiliation(s)
- Jon Hazeldine
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham , Birmingham , UK ; Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Janet M Lord
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham , Birmingham , UK ; Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| | - Antonio Belli
- NIHR Surgical Reconstruction and Microbiology Research Centre, Queen Elizabeth Hospital Birmingham , Birmingham , UK ; Institute of Inflammation and Ageing, University of Birmingham , Birmingham , UK
| |
Collapse
|
18
|
Darcy CJ, Minigo G, Piera KA, Davis JS, McNeil YR, Chen Y, Volkheimer AD, Weinberg JB, Anstey NM, Woodberry T. Neutrophils with myeloid derived suppressor function deplete arginine and constrain T cell function in septic shock patients. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2014; 18:R163. [PMID: 25084831 PMCID: PMC4261583 DOI: 10.1186/cc14003] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 06/18/2014] [Indexed: 01/05/2023]
Abstract
Introduction Impaired T cell function in sepsis is associated with poor outcome, but the mechanisms are unclear. In cancer, arginase-expressing myeloid derived suppressor cells (MDSCs) deplete arginine, impair T cell receptor CD3 zeta-chain expression and T cell function and are linked to poor clinical outcome, but their role during acute human infectious disease and in particular sepsis remains unknown. Hypoarginemia is prevalent in sepsis. This study aimed to determine whether neutrophils that co-purify with PBMC express arginase, and if arginine depletion constrains T cell CD3 zeta-chain expression and function in human sepsis. Methods Using flow cytometry, cell culture, HPLC, arginase activity and mRNA detection, our study examined whether neutrophils, with reduced buoyant density isolated in the Ficoll interface, metabolise L-arginine and suppress T cell proliferation in sepsis. A total of 35 sepsis patients (23 with septic shock) and 12 hospital controls in a tertiary referral hospital in tropical Australia were evaluated. Results Only sepsis patients had interphase neutrophils, neutrophils co-purifying with mononuclear cells (≤1.077 specific gravity). The percentage of interphase neutrophils in sepsis was proportional to sepsis severity and correlated with plasma IL-6 concentrations. Ex vivo, sepsis-derived interphase neutrophils expressed arginase, metabolised culture L-arginine and suppressed T cell proliferation and CD3 zeta-chain expression. In vivo, in septic shock there was a longitudinal inverse association between interphase neutrophil number and CD3 zeta-chain expression. Depletion or inhibition of interphase neutrophils in vitro restored zeta-chain expression and T cell function. Conclusions For the first time during an acute human infection, interphase neutrophils that express arginase were found to circulate in sepsis, in proportion to disease severity. These neutrophil-MDSCs impair T cell CD3 zeta-chain expression and T cell function via L-arginine metabolism, and likely contribute to the T cell dysfunction seen in sepsis. Modulation of neutrophil-MDSC or their downstream effects warrant consideration as targets for novel adjunctive therapies in sepsis. Electronic supplementary material The online version of this article (doi:10.1186/cc14003) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Ssemaganda A, Kindinger L, Bergin P, Nielsen L, Mpendo J, Ssetaala A, Kiwanuka N, Munder M, Teoh TG, Kropf P, Müller I. Characterization of neutrophil subsets in healthy human pregnancies. PLoS One 2014; 9:e85696. [PMID: 24551035 PMCID: PMC3923728 DOI: 10.1371/journal.pone.0085696] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/06/2013] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that in successful pregnancies increased arginase activity is a mechanism that contributes to the suppression of the maternal immune system. We identified the main type of arginase-expressing cells as a population of activated low-density granulocytes (LDGs) in peripheral blood mononuclear cells and in term placentae. In the present study, we analyzed the phenotype of LDGs and compared it to the phenotype of normal density granulocytes (NDGs) in maternal peripheral blood, placental biopsies and cord blood. Our data reveal that only LDGs but no NDGs could be detected in placental biopsies. Phenotypically, NDGs and LDGs from both maternal and cord blood expressed different levels of maturation, activation and degranulation markers. NDGs from the maternal and cord blood were phenotypically similar, while maternal, cord and placental LDGs showed different expression levels of CD66b. LDGs present in cord blood expressed higher levels of arginase compared to maternal and placental LDGs. In summary, our results show that in maternal and cord blood, two phenotypically different populations of neutrophils can be identified, whereas in term placentae, only activated neutrophils are present.
Collapse
Affiliation(s)
- Aloysius Ssemaganda
- Uganda Virus Research Institute - International AIDS Vaccine Initiative, Entebbe, Uganda
| | - Lindsay Kindinger
- Department of Obstetrics and Gynecology, St. Mary's Hospital, London, United Kingdom
| | - Philip Bergin
- International AIDS Vaccine Initiative Human Immunology Laboratory, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Leslie Nielsen
- Uganda Virus Research Institute - International AIDS Vaccine Initiative, Entebbe, Uganda
| | - Juliet Mpendo
- Uganda Virus Research Institute - International AIDS Vaccine Initiative, Entebbe, Uganda
| | - Ali Ssetaala
- Uganda Virus Research Institute - International AIDS Vaccine Initiative, Entebbe, Uganda
| | - Noah Kiwanuka
- Uganda Virus Research Institute - International AIDS Vaccine Initiative, Entebbe, Uganda ; School of Public Health, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Markus Munder
- Third Department of Medicine, Hematology, Oncology and Pneumology, University Medical Center, Mainz, Germany
| | - Tiong Ghee Teoh
- Department of Obstetrics and Gynecology, St. Mary's Hospital, London, United Kingdom
| | - Pascale Kropf
- Section of Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ingrid Müller
- Section of Immunology, Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
20
|
Koestler DC, Christensen B, Karagas MR, Marsit CJ, Langevin SM, Kelsey KT, Wiencke JK, Houseman EA. Blood-based profiles of DNA methylation predict the underlying distribution of cell types: a validation analysis. Epigenetics 2013; 8:816-26. [PMID: 23903776 DOI: 10.4161/epi.25430] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The potential influence of underlying differences in relative leukocyte distributions in studies involving blood-based profiling of DNA methylation is well recognized and has prompted development of a set of statistical methods for inferring changes in the distribution of white blood cells using DNA methylation signatures. However, the extent to which this methodology can accurately predict cell-type proportions based on blood-derived DNA methylation data in a large-scale epigenome-wide association study (EWAS) has yet to be examined. We used publicly available data deposited in the Gene Expression Omnibus (GEO) database (accession number GSE37008), which consisted of both blood-derived epigenome-wide DNA methylation data assayed using the Illumina Infinium HumanMethylation27 BeadArray and complete blood cell (CBC) counts among a community cohort of 94 non-diseased individuals. Constrained projection (CP) was used to obtain predictions of the proportions of lymphocytes, monocytes and granulocytes for each of the study samples based on their DNA methylation signatures. Our findings demonstrated high consistency between the average CBC-derived and predicted percentage of monocytes and lymphocytes (17.9% and 17.6% for monocytes and 82.1% and 81.4% for lymphocytes), with root mean squared error (rMSE) of 5% and 6%, for monocytes and lymphocytes, respectively. Similarly, there was moderate-high correlation between the CP-predicted and CBC-derived percentages of monocytes and lymphocytes (0.60 and 0.61, respectively), and these results were robust to the number of leukocyte differentially methylated regions (L-DMRs) used for CP prediction. These results serve as further validation of the CP approach and highlight the promise of this technique for EWAS where DNA methylation is profiled using whole-blood genomic DNA.
Collapse
Affiliation(s)
- Devin C Koestler
- Department of Community and Family Medicine; Geisel School of Medicine at Dartmouth College; Lebanon, NH USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci 2013; 70:3813-27. [PMID: 23423530 PMCID: PMC3781313 DOI: 10.1007/s00018-013-1286-4] [Citation(s) in RCA: 296] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 01/14/2013] [Accepted: 01/30/2013] [Indexed: 02/06/2023]
Abstract
Neutrophils are essential effector cells in the host defense against invading pathogens. Recently, novel neutrophil functions have emerged in addition to their classical anti-microbial role. One of these functions is the suppression of T cell responses. In this respect, neutrophils share similarities with granulocytic myeloid-derived suppressor cells (G-MDSCs). In this review, we will discuss the similarities and differences between neutrophils and G-MDSCs. Various types of G-MDSCs have been described, ranging from immature to mature cells shaping the immune response by different immune suppressive mechanisms. However, all types of G-MDSCs share distinct features of neutrophils, such as surface markers and morphology. We propose that G-MDSCs are heterogeneous and represent novel phenotypes of neutrophils, capable of suppressing the immune response. In this review, we will attempt to clarify the differences and similarities between neutrophils and G-MDSCs and attempt to facilitate further research.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Many physiologic and pathophysiologic processes are modulated by arginine availability, which can be regulated by arginase. An understanding of the conditions that result in elevated arginase activity as well as the consequences of arginine deficiency is essential for design of effective nutritional support for disease. This review will emphasize recent findings regarding effects of plasma arginase and arginine deficiencies in disease. RECENT FINDINGS Elevations in plasma arginase, derived primarily from hemolysis of red blood cells or liver damage, that are associated with arginine deficiency have been identified in an increasing number of diseases and conditions. Arginine insufficiency not only can activate a stress kinase pathway that impairs function of T lymphocytes but it also can inhibit the mitogen-activated protein kinase signaling pathway required for macrophage production of cytokines in response to bacterial endotoxin/lipopolysaccharide. SUMMARY There are at least two broad categories of arginine deficiency syndromes, involving either T-cell dysfunction or endothelial dysfunction, depending on the disease context in which arginine deficiency occurs. There is limited information regarding the safety and efficacy of supplementation with arginine or its precursor citrulline in ameliorating arginine deficiency in specific diseases, indicating the need for further studies.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
23
|
Pribis JP, Zhu X, Vodovotz Y, Ochoa JB. Systemic arginine depletion after a murine model of surgery or trauma. JPEN J Parenter Enteral Nutr 2011; 36:53-9. [PMID: 22179519 DOI: 10.1177/0148607111414579] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Arginine metabolism and availability after surgery or trauma (ST) is an important modulator of immune responses. Arginine levels are significantly depleted in human trauma patients. Diets containing arginine administered to surgery patients have restored immune function. We hypothesized an arginase-dependent depletion of arginine in a murine model of ST. In addition, we hypothesized a systemic arginase release in human trauma patients. METHODS Male mice were anesthetized and a laparotomy with bowel manipulation was used as a model of ST. Plasma was collected after ST for analysis of arginase activity and arginine, ornithine, and citrulline. Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in plasma were measured after ST. Also, arginase activity was determined in human plasma from 4 healthy controls and 8 trauma patients. RESULTS Arginase activity increased maximally at 2-4 hours after ST, and arginine was significantly reduced after ST. Citrulline was significantly decreased at 8 and 12 hours after ST. Plasma AST and ALT did not significantly vary from control mice after ST. In addition, on day 1 after intensive care unit admission, human trauma patients exhibited a significant increase in arginase activity. CONCLUSIONS The biological consequences of arginine depletion remain incompletely understood. These data are consistent with data showing that patients given arginine-containing diets experience reduced morbidity. Understanding of arginine metabolism after ST may lead to therapies aimed at improving clinical outcome after ST.
Collapse
Affiliation(s)
- John P Pribis
- Department of Surgery, University of Pittsburgh Medical Center, 200 Lothrop St., Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
24
|
Langouche L, Marques MB, Ingels C, Gunst J, Derde S, Vander Perre S, D'Hoore A, Van den Berghe G. Critical illness induces alternative activation of M2 macrophages in adipose tissue. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2011; 15:R245. [PMID: 22018099 PMCID: PMC3334796 DOI: 10.1186/cc10503] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 08/09/2011] [Accepted: 10/21/2011] [Indexed: 12/13/2022]
Abstract
Introduction We recently reported macrophage accumulation in adipose tissue of critically ill patients. Classically activated macrophage accumulation in adipose tissue is a known feature of obesity, where it is linked with increasing insulin resistance. However, the characteristics of adipose tissue macrophage accumulation in critical illness remain unknown. Methods We studied macrophage markers with immunostaining and gene expression in visceral and subcutaneous adipose tissue from healthy control subjects (n = 20) and non-surviving prolonged critically ill patients (n = 61). For comparison, also subcutaneous in vivo adipose tissue biopsies were studied from 15 prolonged critically ill patients. Results Subcutaneous and visceral adipose tissue biopsies from non-surviving prolonged critically ill patients displayed a large increase in macrophage staining. This staining corresponded with elevated gene expression of "alternatively activated" M2 macrophage markers arginase-1, IL-10 and CD163 and low levels of the "classically activated" M1 macrophage markers tumor necrosis factor (TNF)-α and inducible nitric-oxide synthase (iNOS). Immunostaining for CD163 confirmed positive M2 macrophage staining in both visceral and subcutaneous adipose tissue biopsies from critically ill patients. Surprisingly, circulating levels and tissue gene expression of the alternative M2 activators IL-4 and IL-13 were low and not different from controls. In contrast, adipose tissue protein levels of peroxisome proliferator-activated receptor-γ (PPARγ), a nuclear receptor required for M2 differentiation and acting downstream of IL-4, was markedly elevated in illness. In subcutaneous abdominal adipose tissue biopsies from surviving critically ill patients, we could confirm positive macrophage staining with CD68 and CD163. We also could confirm elevated arginase-1 gene expression and elevated PPARγ protein levels. Conclusions Unlike obesity, critical illness evokes adipose tissue accumulation of alternatively activated M2 macrophages, which have local anti-inflammatory and insulin sensitizing features. This M2 macrophage accumulation may contribute to the previously observed protective metabolic activity of adipose tissue during critical illness.
Collapse
Affiliation(s)
- Lies Langouche
- Laboratory and Department of Intensive Care Medicine, K.U.Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|