1
|
Heiss J, Grün K, Singerer I, Tempel L, Matasci M, Jung C, Pfeil A, Schulze PC, Neri D, Franz M. Expression of Inflammatory Genes in Murine Lungs in a Model of Experimental Pulmonary Hypertension: Effects of an Antibody-Based Targeted Delivery of Interleukin-9. Adv Respir Med 2024; 92:27-35. [PMID: 38247549 PMCID: PMC10801467 DOI: 10.3390/arm92010005] [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: 11/11/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Pathogenesis of pulmonary hypertension (PH) is a multifactorial process driven by inflammation and pulmonary vascular remodeling. To target these two aspects of PH, we recently tested a novel treatment: Interleukin-9 (IL9) fused to F8, an antibody that binds to the extra-domain A of fibronectin (EDA+ Fn). As EDA+ Fn is not found in healthy adult tissue but is expressed during PH, IL9 is delivered specifically to the tissue affected by PH. We found that F8IL9 reduced pulmonary vascular remodeling and attenuated PH compared with sham-treated mice. PURPOSE To evaluate possible F8IL9 effects on PH-associated inflammatory processes, we analysed the expression of genes involved in pulmonary immune responses. METHODS We applied the monocrotaline (MCT) model of PH in mice (n = 44). Animals were divided into five experimental groups: sham-induced animals without PH (control, n = 4), MCT-induced PH without treatment (PH, n = 8), dual endothelin receptor antagonist treatment (dual ERA, n = 8), F8IL9 treatment (n = 12, 2 formats with n = 6 each), or with KSFIL9 treatment (KSFIL9, n = 12, 2 formats with n = 6 each, KSF: control antibody with irrelevant antigen specificity). After 28 days, a RT-PCR gene expression analysis of inflammatory response (84 genes) was performed in the lung. RESULTS Compared with the controls, 19 genes exhibited relevant (+2.5-fold) upregulation in the PH group without treatment. Gene expression levels in F8IL9-treated lung tissue were reduced compared to the PH group without treatment. This was the case especially for CCL20, CXCL5, C-reactive protein, pentraxin related (CRPPR), and Kininogen-1 (KNG1). CONCLUSION In accordance with the hypothesis stated above, F8IL9 treatment diminished the upregulation of some genes associated with inflammation in a PH animal model. Therefore, we hypothesize that IL9-based immunocytokine treatment will likely modulate various inflammatory pathways.
Collapse
Affiliation(s)
- Judith Heiss
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (J.H.); (K.G.); (I.S.); (L.T.); (P.C.S.)
- Else Kröner Graduate School for Medical Students “JSAM”, Jena University Hospital, 07747 Jena, Germany
| | - Katja Grün
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (J.H.); (K.G.); (I.S.); (L.T.); (P.C.S.)
| | - Isabell Singerer
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (J.H.); (K.G.); (I.S.); (L.T.); (P.C.S.)
| | - Laura Tempel
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (J.H.); (K.G.); (I.S.); (L.T.); (P.C.S.)
| | | | - Christian Jung
- Medical Faculty, Division of Cardiology, Pulmonology and Vascular Medicine, Heinrich-Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Alexander Pfeil
- Department of Internal Medicine III, University Hospital Jena, 07747 Jena, Germany;
| | - P. Christian Schulze
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (J.H.); (K.G.); (I.S.); (L.T.); (P.C.S.)
| | - Dario Neri
- Philochem AG, 8112 Otelfingen, Switzerland; (M.M.)
| | - Marcus Franz
- Department of Internal Medicine I, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany; (J.H.); (K.G.); (I.S.); (L.T.); (P.C.S.)
| |
Collapse
|
2
|
Zheng L, Zhang Z, Song K, Xu X, Tong Y, Wei J, Jiang L. Potential biomarkers for inflammatory response in acute lung injury. Open Med (Wars) 2022; 17:1066-1076. [PMID: 35795000 PMCID: PMC9186513 DOI: 10.1515/med-2022-0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 03/24/2022] [Accepted: 04/27/2022] [Indexed: 11/15/2022] Open
Abstract
Acute lung injury (ALI) is a severe respiratory disorder occurring in critical care medicine, with high rates of mortality and morbidity. This study aims to screen the potential biomarkers for ALI. Microarray data of lung tissues from lung-specific geranylgeranyl pyrophosphate synthase large subunit 1 knockout and wild-type mice treated with lipopolysaccharide were downloaded. Differentially expressed genes (DEGs) between ALI and wild-type mice were screened. Functional analysis and the protein-protein interaction (PPI) modules were analyzed. Finally, a miRNA-transcription factor (TF)-target regulation network was constructed. Totally, 421 DEGs between ALI and wild-type mice were identified. The upregulated DEGs were mainly enriched in the peroxisome proliferator-activated receptor signaling pathway, and fatty acid metabolic process, while downregulated DEGs were related to cytokine-cytokine receptor interaction and regulation of cytokine production. Cxcl5, Cxcl9, Ccr5, and Cxcr4 were key nodes in the PPI network. In addition, three miRNAs (miR505, miR23A, and miR23B) and three TFs (PU1, CEBPA, and CEBPB) were key molecules in the miRNA-TF-target network. Nine genes including ADRA2A, P2RY12, ADORA1, CXCR1, and CXCR4 were predicted as potential druggable genes. As a conclusion, ADRA2A, P2RY12, ADORA1, CXCL5, CXCL9, CXCR1, and CXCR4 might be novel markers and potential druggable genes in ALI by regulating inflammatory response.
Collapse
Affiliation(s)
- Lanzhi Zheng
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310006 Zhejiang Province, China
| | - Zhuoyi Zhang
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Youdian Road 54#, Shangcheng District, Hangzhou City, 310006 Zhejiang Province, China
| | - Kang Song
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310006 Zhejiang Province, China
| | - Xiaoyang Xu
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310006 Zhejiang Province, China
| | - Yixin Tong
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310006 Zhejiang Province, China
| | - Jinling Wei
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310006 Zhejiang Province, China
| | - Lu Jiang
- Emergency Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, 310006 Zhejiang Province, China
| |
Collapse
|
3
|
Qiao Q, Liu X, Yang T, Cui K, Kong L, Yang C, Zhang Z. Nanomedicine for acute respiratory distress syndrome: The latest application, targeting strategy, and rational design. Acta Pharm Sin B 2021; 11:3060-3091. [PMID: 33977080 PMCID: PMC8102084 DOI: 10.1016/j.apsb.2021.04.023] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 01/08/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by the severe inflammation and destruction of the lung air-blood barrier, leading to irreversible and substantial respiratory function damage. Patients with coronavirus disease 2019 (COVID-19) have been encountered with a high risk of ARDS, underscoring the urgency for exploiting effective therapy. However, proper medications for ARDS are still lacking due to poor pharmacokinetics, non-specific side effects, inability to surmount pulmonary barrier, and inadequate management of heterogeneity. The increased lung permeability in the pathological environment of ARDS may contribute to nanoparticle-mediated passive targeting delivery. Nanomedicine has demonstrated unique advantages in solving the dilemma of ARDS drug therapy, which can address the shortcomings and limitations of traditional anti-inflammatory or antioxidant drug treatment. Through passive, active, or physicochemical targeting, nanocarriers can interact with lung epithelium/endothelium and inflammatory cells to reverse abnormal changes and restore homeostasis of the pulmonary environment, thereby showing good therapeutic activity and reduced toxicity. This article reviews the latest applications of nanomedicine in pre-clinical ARDS therapy, highlights the strategies for targeted treatment of lung inflammation, presents the innovative drug delivery systems, and provides inspiration for strengthening the therapeutic effect of nanomedicine-based treatment.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- AEC II, alveolar type II epithelial cells
- AM, alveolar macrophages
- ARDS, acute respiratory distress syndrome
- Acute lung injury
- Acute respiratory distress syndrome
- Anti-inflammatory therapy
- BALF, bronchoalveolar lavage fluid
- BSA, bovine serum albumin
- CD, cyclodextrin
- CLP, cecal ligation and perforation
- COVID-19
- COVID-19, coronavirus disease 2019
- DOPE, phosphatidylethanolamine
- DOTAP, 1-diolefin-3-trimethylaminopropane
- DOX, doxorubicin
- DPPC, dipalmitoylphosphatidylcholine
- Drug delivery
- ECM, extracellular matrix
- ELVIS, extravasation through leaky vasculature and subsequent inflammatory cell-mediated sequestration
- EPCs, endothelial progenitor cells
- EPR, enhanced permeability and retention
- EVs, extracellular vesicles
- EphA2, ephrin type-A receptor 2
- Esbp, E-selectin-binding peptide
- FcgR, Fcγ receptor
- GNP, peptide-gold nanoparticle
- H2O2, hydrogen peroxide
- HO-1, heme oxygenase-1
- ICAM-1, intercellular adhesion molecule-1
- IKK, IκB kinase
- IL, interleukin
- LPS, lipopolysaccharide
- MERS, Middle East respiratory syndrome
- MPMVECs, mouse pulmonary microvascular endothelial cells
- MPO, myeloperoxidase
- MSC, mesenchymal stem cells
- NAC, N-acetylcysteine
- NE, neutrophil elastase
- NETs, neutrophil extracellular traps
- NF-κB, nuclear factor-κB
- Nanomedicine
- PC, phosphatidylcholine
- PCB, poly(carboxybetaine)
- PDA, polydopamine
- PDE4, phosphodiesterase 4
- PECAM-1, platelet-endothelial cell adhesion molecule
- PEG, poly(ethylene glycol)
- PEI, polyetherimide
- PEVs, platelet-derived extracellular vesicles
- PLGA, poly(lactic-co-glycolic acid)
- PS-PEG, poly(styrene-b-ethylene glycol)
- Pathophysiologic feature
- RBC, red blood cells
- RBD, receptor-binding domains
- ROS, reactive oxygen species
- S1PLyase, sphingosine-1-phosphate lyase
- SARS, severe acute respiratory syndrome
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- SDC1, syndecan-1
- SORT, selective organ targeting
- SP, surfactant protein
- Se, selenium
- Siglec, sialic acid-binding immunoglobulin-like lectin
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor-α
- TPP, triphenylphosphonium cation
- Targeting strategy
- YSA, YSAYPDSVPMMS
- cRGD, cyclic arginine glycine-d-aspartic acid
- iNOS, inducible nitric oxide synthase
- rSPANb, anti-rat SP-A nanobody
- scFv, single chain variable fragments
Collapse
Affiliation(s)
- Qi Qiao
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiong Liu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ting Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Kexin Cui
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
4
|
Club Cell Protein 16 Attenuates CD16 brightCD62 dim Immunosuppressive Neutrophils in Damaged Tissue upon Posttraumatic Sepsis-Induced Lung Injury. J Immunol Res 2021; 2021:6647753. [PMID: 33575362 PMCID: PMC7861919 DOI: 10.1155/2021/6647753] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/24/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Background Recently, identification of immunosuppressive polymorphonuclear leukocytes (PMNL) that were traditionally described as proinflammatory cells emerged in the field of posttraumatic immunity. To understand their local and remote distribution after trauma, PMNL-subsets and the impact of immunomodulatory Club Cell protein (CC)16 that correlates with pulmonary complications were assessed. Methods C57BL/6N mice were divided into three groups, receiving isolated blunt chest trauma (TxT), undergoing TxT followed by cecal ligation and puncture (CLP, TxT + CLP) after 24 h, or sham undergoing analgosedation (n = 18/group). Further, each group was subdivided into three groups receiving either no treatment (ctrl) or intratracheal neutralization of CC16 by application of anti-CC16-antibody or application of an unspecific IgG control antibody (n = 6/group). Treatment was set at the time point after TxT. Analyses followed 6 h post-CLP. PMNL were characterized via expression of CD11b, CD16, CD45, CD62L, and Ly6G by flow cytometry in bone marrow (BM), blood, spleen, lung, liver, and bronchoalveolar and peritoneal lavage fluid (BALF and PL). Apoptosis was assessed by activated (cleaved) caspase-3. Results from untreated ctrl and IgG-treated mice were statistically comparable between all corresponding sham, TxT, and TxT + CLP groups. Results Immature (CD16dimCD62Lbright) PMNL increased significantly in BM, circulation, and spleen after TxT vs. sham and were significantly attenuated in the lungs, BALF, PL, and liver. Classical-shaped (CD16brightCD62Lbright) PMNL increased after TxT vs. sham in peripheral tissue and were significantly attenuated in circulation, proposing a trauma-induced migration of mature or peripheral differentiation of circulating immature PMNL. Immunosuppressive (CD16brightCD62Ldim) PMNL decreased significantly in the lungs and spleen, while they systemically increased after TxT vs. sham. CLP in the TxT + CLP group reduced immunosuppressive PMNL in PL and increased their circulatory rate vs. isolated TxT, showing local reduction in affected tissue and their increase in nonaffected tissue. CC16 neutralization enhanced the fraction of immunosuppressive PMNL following TxT vs. sham and decreased caspase-3 in the lungs post-CLP in the TxT + CLP group, while apoptotic cells in the liver diminished post-TxT. Posttraumatic CC16 neutralization promotes the subset of immunosuppressive PMNL and antagonizes their posttraumatic distribution. Conclusion Since CC16 affects both the distribution of PMNL subsets and apoptosis in tissues after trauma, it may constitute as a novel target to beneficially shape the posttraumatic tissue microenvironment and homeostasis to improving outcomes.
Collapse
|
5
|
Relja B, Yang B, Bundkirchen K, Xu B, Köhler K, Neunaber C. Different experimental multiple trauma models induce comparable inflammation and organ injury. Sci Rep 2020; 10:20185. [PMID: 33214576 PMCID: PMC7678855 DOI: 10.1038/s41598-020-76499-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 10/05/2020] [Indexed: 01/04/2023] Open
Abstract
Multiple injuries appear to be a decisive factor for experimental polytrauma. Therefore, our aim was to compare the inflammatory response and organ damage of five different monotrauma with three multiple trauma models. For this, mice were randomly assigned to 10 groups: Healthy control (Ctrl), Sham, hemorrhagic shock (HS), thoracic trauma (TxT), osteotomy with external fixation (Fx), bilateral soft tissue trauma (bsTT) or laparotomy (Lap); polytrauma I (PT I, TxT + HS + Fx), PT II (TxT + HS + Fx + Lap) and one multi-trauma group (MT, TxT + HS + bsTT + Lap). The inflammatory response and organ damage were quantified at 6 h by analyses of IL-6, IL-1β, IL-10, CXCL1, SAA1, HMGB1 and organ injury. Systemic IL-6 increased in all mono and multiple trauma groups, while CXCL1 increased only in HS, PT I, PT II and MT vs. control. Local inflammatory response was most prominent in HS, PT I, PT II and MT in the liver. Infiltration of inflammatory cells into lung and liver was significant in all multiple trauma groups vs. controls. Hepatic and pulmonary injury was prominent in HS, PT I, PT II and MT groups. These experimental multiple trauma models closely mimic the early post-traumatic inflammatory response in human. Though, the choice of read-out parameters is very important for therapeutic immune modulatory approaches.
Collapse
Affiliation(s)
- Borna Relja
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Bing Yang
- Trauma Department, Hannover Medical School, Hannover, Germany
| | | | - Baolin Xu
- Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany.,Trauma Department, Hannover Medical School, Hannover, Germany.,Department of Trauma, Hand and Reconstructive Surgery, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kernt Köhler
- Institute of Veterinary Pathology, Justus Liebig University, Giessen, Germany
| | | |
Collapse
|
6
|
Vollrath JT, Stoermann P, Becker N, Wutzler S, Hildebrand F, Marzi I, Relja B. Early local neutralization of CC16 in sepsis‑induced ALI following blunt chest trauma leads to delayed mortality without benefitting overall survival. Int J Mol Med 2020; 46:2207-2215. [PMID: 33125155 PMCID: PMC7595659 DOI: 10.3892/ijmm.2020.4767] [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: 03/18/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022] Open
Abstract
Blunt thoracic trauma (TxT) is a common injury pattern in polytraumatized patients. When combined with a secondary trigger, TxT often results in acute lung injury (ALI), which negatively affects outcomes. Recent findings suggest that ALI is caused by both local and systemic inflammatory reactions. Club cell protein (CC)16 is an anti‑inflammatory peptide associated with lung injury following TxT. Recently, the anti‑inflammatory properties of endogenous CC16 in a murine model of TxT with subsequent cecal‑ligation and puncture (CLP) as the secondary hit were demonstrated by our group. The present study aimed to determine whether CC16 neutralization improves survival following 'double‑hit'‑induced ALI. For this purpose, a total of 120 C57BL/6N mice were subjected to TxT, followed by CLP after 24 h. Sham‑operated animals underwent anesthesia without the induction of TxT + CLP. CC16 neutralization was performed by providing a CC16 antibody intratracheally following TxT (early) or following CLP (late). Survival was assessed in 48 animals for 6 days after CLP. Sacrifice was performed 6 or 24 h post‑CLP to evaluate the anti‑inflammatory effect of CC16. The results revealed that CC16 neutralization enhanced pro‑inflammatory CXCL1 levels, thereby confirming the anti‑inflammatory characteristics of CC16 in this model. Early CC16 neutralization immediately following TxT significantly prolonged survival within 60 h; however, the survival rate did not change until 6 days post‑trauma. Late CC16 neutralization did not provide any survival benefits. On the whole, the present study demonstrated that neutralizing CC16 confirmed its anti‑inflammatory potential in this double‑hit ALI model. Early CC16 neutralization prolonged survival within 60 h; however, no survival benefits were observed after 6 days post‑CLP in any group.
Collapse
Affiliation(s)
- Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Philipp Stoermann
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Nils Becker
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Sebastian Wutzler
- Department of Trauma, Hand and Orthopedic Surgery, Helios Horst Schmidt Clinic, D‑65199 Wiesbaden, Germany
| | - Frank Hildebrand
- Department of Trauma Surgery, RWTH University, D‑52062 Aachen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, D‑60590 Frankfurt, Germany
| |
Collapse
|
7
|
Horst K, Hildebrand F. Focus on chest trauma: implications from clinical and experimental studies. Eur J Trauma Emerg Surg 2020; 46:1-2. [PMID: 32067054 PMCID: PMC7026208 DOI: 10.1007/s00068-020-01310-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Klemens Horst
- Department of Trauma and Reconstructive Surgery, Uniklinik RWTH Aachen, Aachen, Germany
| | - Frank Hildebrand
- Department of Trauma and Reconstructive Surgery, Uniklinik RWTH Aachen, Aachen, Germany.
| |
Collapse
|
8
|
Janicova A, Becker N, Xu B, Wutzler S, Vollrath JT, Hildebrand F, Ehnert S, Marzi I, Störmann P, Relja B. Endogenous Uteroglobin as Intrinsic Anti-inflammatory Signal Modulates Monocyte and Macrophage Subsets Distribution Upon Sepsis Induced Lung Injury. Front Immunol 2019; 10:2276. [PMID: 31632392 PMCID: PMC6779999 DOI: 10.3389/fimmu.2019.02276] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022] Open
Abstract
Sepsis is a serious clinical condition which can cause life-threatening organ dysfunction, and has limited therapeutic options. The paradigm of limiting excessive inflammation and promoting anti-inflammatory responses is a simplified concept. Yet, the absence of intrinsic anti-inflammatory signaling at the early stage of an infection can lead to an exaggerated activation of immune cells, including monocytes and macrophages. There is emerging evidence that endogenous molecules control those mechanisms. Here we aimed to identify and describe the dynamic changes in monocyte and macrophage subsets and lung damage in CL57BL/6N mice undergoing blunt chest trauma with subsequent cecal ligation and puncture. We showed that early an increase in systemic and activated Ly6C+CD11b+CD45+Ly6G- monocytes was paralleled by their increased emigration into lungs. The ratio of pro-inflammatory Ly6ChighCD11b+CD45+Ly6G- to patrolling Ly6ClowCD11b+CD45+Ly6G- monocytes significantly increased in blood, lungs and bronchoalveolar lavage fluid (BALF) suggesting an early transition to inflammatory phenotypes during early sepsis development. Similar to monocytes, the level of pro-inflammatory Ly6ChighCD45+F4/80+ macrophages increased in lungs and BALF, while tissue repairing Ly6ClowCD45+F4/80+ macrophages declined in BALF. Levels of inflammatory mediators TNF-α and MCP-1 in blood and RAGE in lungs and BALF were elevated, and besides their boosting of inflammation via the recruitment of cells, they may promote monocyte and macrophage polarization, respectively, toward the pro-inflammatory phenotype. Neutralization of uteroglobin increased pro-inflammatory cytokine levels, activation of inflammatory phenotypes and their recruitment to lungs; concurrent with increased pulmonary damage in septic mice. In in vitro experiments, the influence of uteroglobin on monocyte functions including migratory behavior, TGF-β1 expression, cytotoxicity and viability were proven. These results highlight an important role of endogenous uteroglobin as intrinsic anti-inflammatory signal upon sepsis-induced early lung injury, which modules the early monocyte/macrophages driven inflammation. Short Summary Blunt chest injury is the third largest cause of death following major trauma, and ongoing excessive pro-inflammatory immune response entails high risk for the development of secondary complications, such as sepsis, with limited therapeutic options. In murine double hit trauma consisting of thoracic trauma and subsequent cecal ligation and puncture, we investigated the cytokine profile, pulmonary epithelial integrity and phenotypic shift of patrolling Ly6ClowCD11b+CD45+Ly6G- monocytes and Ly6ClowCD45+F4/80+ macrophages to pro-inflammatory Ly6ChighCD11b+CD45+Ly6G- monocytes and Ly6ChighCD45+F4/80+ cells in blood, lungs and bronchoalveolar lavage fluid (BALF). Pro-inflammatory mediators and phenotypes were elevated and uteroglobin neutralization led to further increase. Enhanced total protein levels in BALF suggests leakage of respiratory epithelium. In vitro, uteroglobin inhibited the migratory capacity of monocytes and the TGF-β1 expression without affecting the viability. These results highlight an important role of endogenous uteroglobin as an intrinsic anti-inflammatory signal upon sepsis-induced early lung injury, which modulates the early monocyte/macrophages driven inflammation.
Collapse
Affiliation(s)
- Andrea Janicova
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany.,Department of Aquatic Ecotoxicology, Goethe University, Frankfurt, Germany.,Department of Radiology and Nuclear Medicine, Experimental Radiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Nils Becker
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Baolin Xu
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Sebastian Wutzler
- Orthopedic and Trauma Surgery, Helios Horst Schmidt Clinic, Wiesbaden, Germany
| | - Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | | | - Sabrina Ehnert
- Department of Trauma and Reconstructive Surgery, Siegfried Weller Research Institute, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Ingo Marzi
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Philipp Störmann
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, Frankfurt, Germany
| |
Collapse
|
9
|
A nationwide fluidics biobank of polytraumatized patients: implemented by the Network "Trauma Research" (NTF) as an expansion to the TraumaRegister DGU ® of the German Trauma Society (DGU). Eur J Trauma Emerg Surg 2019; 46:499-504. [PMID: 31324937 PMCID: PMC7280175 DOI: 10.1007/s00068-019-01193-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/15/2019] [Indexed: 11/24/2022]
Abstract
To decrypt the complexity of the posttraumatic immune responses and to potentially identify novel research pathways for exploration, large-scale multi-center projects including not only in vivo and in vitro modeling, but also temporal sample and material collection along with clinical data capture from multiply injured patients is of utmost importance. To meet this gap, a nationwide biobank for fluidic samples from polytraumatized patients was initiated in 2013 by the task force Network “Trauma Research” (Netzwerk Traumaforschung, NTF) of the German Trauma Society (Deutsche Gesellschaft für Unfallchirurgie e.V., DGU). The NTF-Biobank completes the clinical NTF-Biobank Database and complements the TR-DGU with temporal biological samples from multiply injured patients. The concept behind the idea of the NTF-Biobank was to create a robust interface for meaningful innovative basic, translational and clinical research. For the first time, an integrated platform to prospectively evaluate and monitor candidate biomarkers and/or potential therapeutic targets in biological specimens of quality-controlled and documented patients is introduced, allowing reduction in variability of measurements with high impact due to its large sample size. Thus, the project was introduced to systemically evaluate and monitor multiply injured patients for their (patho-)physiological sequalae together with their clinical treatment strategies applied for overall outcome improval.
Collapse
|
10
|
Early Local Inhibition of Club Cell Protein 16 Following Chest Trauma Reduces Late Sepsis-Induced Acute Lung Injury. J Clin Med 2019; 8:jcm8060896. [PMID: 31234556 PMCID: PMC6616892 DOI: 10.3390/jcm8060896] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
Blunt thoracic trauma (TxT) deteriorates clinical post-injury outcomes. Ongoing inflammatory changes promote the development of post-traumatic complications, frequently causing Acute Lung Injury (ALI). Club Cell Protein (CC)16, a pulmonary anti-inflammatory protein, correlates with lung damage following TxT. Whether CC16-neutralization influences the inflammatory course during ALI is elusive. Ninety-six male CL57BL/6N mice underwent a double hit model of TxT and cecal ligation puncture (CLP, 24 h post-TxT). Shams underwent surgical procedures. CC16 was neutralized by the intratracheal application of an anti-CC16-antibody, either after TxT (early) or following CLP (late). Euthanasia was performed at 6 or 24 h post-CLP. Systemic and pulmonary levels of IL-6, IL-1β, and CXCL5 were determined, the neutrophils were quantified in the bronchoalveolar lavage fluid, and histomorphological lung damage was assessed. ALI induced a significant systemic IL-6 increase among all groups, while the local inflammatory response was most prominent after 24 h in the double-hit groups as compared to the shams. Significantly increased neutrophilic infiltration upon double hit was paralleled with the enhanced lung damage in all groups as compared to the sham, after 6 and 24 h. Neutralization of CC16 did not change the systemic inflammation. However, early CC16-neutralization increased the neutrophilic infiltration and lung injury at 6 h post-CLP, while 24 h later, the lung injury was reduced. Late CC16-neutralization increased neutrophilic infiltration, 24 h post-CLP, and was concurrent with an enhanced lung injury. The data confirmed the anti-inflammatory potential of endogenous CC16 in the murine double-hit model of ALI.
Collapse
|