1
|
Guo K, Yin Y, Zheng L, Wu Z, Rao X, Zhu W, Zhou B, Liu L, Liu D. Integration of microbiomics, metabolomics, and transcriptomics reveals the therapeutic mechanism underlying Fuzheng-Qushi decoction for the treatment of lipopolysaccharide-induced lung injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118584. [PMID: 39019418 DOI: 10.1016/j.jep.2024.118584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/12/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fuzheng-Qushi decoction (FZQS) is a practical Chinese herbal formula for relieving cough and fever. Therefore, the action and specific molecular mechanism of FZQS in the treatment of lung injury with cough and fever as the main symptoms need to be further investigated. AIMS OF THE STUDY To elucidate the protective effects of FZQS against lung injury in mice and reveal its potential targets and key biological pathways for the treatment of lung injury based on transcriptomics, microbiomics, and untargeted metabolomics analyses. MATERIALS AND METHODS Lipopolysaccharide (LPS) was used to induce a mouse model of lung injury, followed by the administration of FZQS. ELISA was used to detect IL-1β, IL-6, IL-17A, IL-4, IL-10, and TNF-α, in mouse lung tissues. Macrophage polarization and neutrophil activation were measured by flow cytometry. RNA sequencing (RNA-seq) was applied to screen for differentially expressed genes (DEGs) in lung tissues. RT-qPCR and Western blot assays were utilized to validate key DEGs and target proteins in lung tissues. 16S rRNA sequencing was employed to characterize the gut microbiota of mice. Metabolites in the gut were analyzed using untargeted metabolomics. RESULTS FZQS treatment significantly ameliorated lung histopathological damage, decreased pro-inflammatory cytokine levels, and increased anti-inflammatory cytokine levels. M1 macrophage levels in the peripheral blood decreased, M2 macrophage levels increased, and activated neutrophils were inhibited in mice with LPS-induced lung injury. Importantly, transcriptomic analysis showed that FZQS downregulated macrophage and neutrophil activation and migration and adhesion pathways by reversing 51 DEGs, which was further confirmed by RT-qPCR and Western blot analysis. In addition, FZQS modulated the dysbiosis of the gut microbiota by reversing the abundance of Corynebacterium, Facklamia, Staphylococcus, Paenalcaligenes, Lachnoclostridium, norank_f_Muribaculaceae, and unclassified_f_Lachnospiraceae. Meanwhile, metabolomics analysis revealed that FZQS significantly regulated tryptophan metabolism by reducing the levels of 3-Indoleacetonitrile and 5-Hydroxykynurenine. CONCLUSION FZQS effectively ameliorated LPS-induced lung injury by inhibiting the activation, migration, and adhesion of macrophages and neutrophils and modulating gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Kaien Guo
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Yuting Yin
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Linxin Zheng
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Zenan Wu
- Department of Postgraduate, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Xiaoyong Rao
- National Engineering Center for Manufacturing Technology of Solid Preparations of Traditional Chinese Medicine Manufacturing Technology, Nanchang, 330004, Jiangxi Province, China
| | - Weifeng Zhu
- Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China
| | - Bugao Zhou
- Department of Research, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Liangji Liu
- Affiliated Hospital, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| | - Duanyong Liu
- Formula-pattern Research Center, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China; School of Nursing, Jiangxi University of Chinese Medicine, Nanchang, 330004, Jiangxi Province, China.
| |
Collapse
|
2
|
Aydın Ö, Apaydın Yıldırım B. Determination of systemic inflammation response index (SIRI), systemic inflammatory index (SII), HMGB1, Mx1 and TNF levels in neonatal calf diarrhea with systemic inflammatory response syndrome. Vet Immunol Immunopathol 2024; 275:110815. [PMID: 39153273 DOI: 10.1016/j.vetimm.2024.110815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/10/2024] [Accepted: 08/11/2024] [Indexed: 08/19/2024]
Abstract
The objective of this study was to examine the values of MX dynamin-like GTPase 1 (Mx1), high mobility group box-1 (HMGB1), systemic inflammatory response index (SIRI), systemic inflammatory index (SII), tumor necrosis factor (TNF), and other hematological indices in calves with systemic inflammatory response syndrome (SIRS). The study material was divided into two groups: the SIRS group (comprising 13 calves) and the control group (comprising 10 calves). The independent samples t-test and Mann-Whitney U test were employed for normally distributed and non-normally distributed data, respectively. The relationship between the two groups was determined using Spearman correlation coefficient analysis. Significant differences were identified between the SIRS group and the control group with regard to white blood cell (WBC; P < 0.05), neutrophil (NEU; P < 0.01), and neutrophil-to-lymphocyte ratio (NLR; P < 0.001) values, in addition to SIRI (P < 0.05), SII (P < 0.01) values. Furthermore, HMGB1 (P < 0.001), Mx1 (P < 0.05), and TNF values (P < 0.001) demonstrated notable disparities between the two groups. As a result of this study, it was concluded that there were significant increases in inflammatory hematological indices, as well as in the levels of HMGB1, Mx1, and TNF, in calves with SIRS.
Collapse
Affiliation(s)
- Ömer Aydın
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Betül Apaydın Yıldırım
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
3
|
Fujimoto R, Oda S, Matsumoto H, Nakao S, Shimizu K, Okuzaki D, Oda J. Longitudinal whole blood transcriptome analysis of a septic shock patient with secondary hemophagocytic syndrome. QJM 2024; 117:470-471. [PMID: 38460187 DOI: 10.1093/qjmed/hcae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Indexed: 03/11/2024] Open
Affiliation(s)
- Risei Fujimoto
- Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - S Oda
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - H Matsumoto
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita City, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, 2-8 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - S Nakao
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - K Shimizu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - D Okuzaki
- Center for Infectious Disease Education and Research, Osaka University, 2-8 Yamadaoka, Suita City, Osaka 565-0871, Japan
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita City, Osaka 565-0871, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, 3-1 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - J Oda
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, 2-15 Yamadaoka, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Zhong T, Chen S, Deng K, Guan J, Zhang J, Lu F, Shichen M, Lv R, Liu Z, Liu Y, Chang P, Liu Z. Magnesium alleviates extracellular histone-induced apoptosis and defective bacterial phagocytosis in macrophages by regulating intracellular calcium signal. Int Immunopharmacol 2024; 132:111870. [PMID: 38547771 DOI: 10.1016/j.intimp.2024.111870] [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: 01/04/2024] [Revised: 02/18/2024] [Accepted: 03/12/2024] [Indexed: 05/01/2024]
Abstract
Extracellular histones have been determined as important mediators of sepsis, which induce excessive inflammatory responses in macrophages and impair innate immunity. Magnesium (Mg2+), one of the essential nutrients of the human body, contributes to the proper regulation of immune function. However, no reports indicate whether extracellular histones affect survival and bacterial phagocytosis in macrophages and whether Mg2+ is protective against histone-induced macrophage damage. Our clinical data revealed a negative correlation between circulating histone and monocyte levels in septic patients, and in vitro experiments confirmed that histones induced mitochondria-associated apoptosis and defective bacterial phagocytosis in macrophages. Interestingly, our clinical data also indicated an association between lower serum Mg2+ levels and reduced monocyte levels in septic patients. Moreover, in vitro experiments demonstrated that Mg2+ attenuated histone-induced apoptosis and defective bacterial phagocytosis in macrophages through the PLC/IP3R/STIM-mediated calcium signaling pathway. Importantly, further animal experiments proved that Mg2+ significantly improved survival and attenuated histone-mediated lung injury and macrophage damage in histone-stimulated mice. Additionally, in a cecal ligation and puncture (CLP) + histone-induced injury mouse model, Mg2+ inhibited histone-mediated apoptosis and defective phagocytosis in macrophages and further reduced bacterial load. Overall, these results suggest that Mg2+ supplementation may be a promising treatment for extracellular histone-mediated macrophage damage in sepsis.
Collapse
Affiliation(s)
- Tao Zhong
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Sainan Chen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ke Deng
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianbin Guan
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaqi Zhang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Furong Lu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Maoyou Shichen
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ronggui Lv
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Zhifeng Liu
- Department of Medicine Intensive Care Units, General Hospital of Southern Theatre Command of PLA, Guangzhou, Guangdong, China.
| | - Yong Liu
- Department of Intensive Care Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Ping Chang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhanguo Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
5
|
Valverde A, Naqvi RA, Naqvi AR. Non-coding RNA LINC01010 regulates macrophage polarization and innate immune functions by modulating NFκB signaling pathway. J Cell Physiol 2024; 239:e31225. [PMID: 38403999 PMCID: PMC11096022 DOI: 10.1002/jcp.31225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/27/2024]
Abstract
Innate immune response is regulated by tissue resident or infiltrating immune cells such as macrophages (Mφ) that play critical role in tissue development, homeostasis, and repair of damaged tissue. However, the epigenetic mechanisms that regulate Mφ plasticity and innate immune functions are not well understood. Long non-coding RNA (lncRNA) are among the most abundant class of transcriptome but their function in myeloid cell biology is less explored. In this study, we deciphered the regulatory role of previously uncharacterized lncRNAs in Mφ polarization and innate immune responses. Two lncRNAs showed notable changes in their levels during M1 and M2 Mφ differentiation. Our findings indicate that LINC01010 expression increased and AC007032 expression decreased significantly. LINC01010 exhibit myeloid cell-specificity, while AC007032.1 is ubiquitous and expressed in both myeloid and lymphoid (T cells, B cells and NK cells) cells. Expression of these lncRNAs is dysregulated in periodontal disease (PD), a microbial biofilm-induced immune disease, and responsive to lipopolysaccharide (LPS) from different oral and non-oral bacteria. Knockdown of LINC01010 but not AC007032.1 reduced the surface expression of Mφ differentiation markers CD206 and CD68, and M1Mφ polarization markers MHCII and CD32. Furthermore, LINC01010 RNAi attenuated bacterial phagocytosis, antigen processing and cytokine secretion suggesting its key function in innate immunity. Mechanistically, LINC01010 knockdown Mφ treated with Escherichia coli LPS exhibit significantly reduced expression of multiple nuclear factor kappa B pathway genes. Together, our data highlight functional role of a PD-associated lncRNA LINC01010 in shaping macrophage differentiation, polarization, and innate immune activation.
Collapse
Affiliation(s)
- Araceli Valverde
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois, United States
| | - Raza Ali Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois, United States
| | - Afsar R. Naqvi
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, Illinois, United States
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
| |
Collapse
|
6
|
Lin XM, Zhang LF, Wang YT, Huang T, Lin XF, Hong XY, Zheng HJ, Xie RC, Ma JF. Application of neutrophil-to-lymphocyte-to-monocyte ratio in predicting mortality risk in adult patients with septic shock: A retrospective cohort study conducted at a single center. Heliyon 2024; 10:e28809. [PMID: 38596065 PMCID: PMC11002270 DOI: 10.1016/j.heliyon.2024.e28809] [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: 12/04/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/11/2024] Open
Abstract
Background Sepsis is a life-threatening condition characterized by an aberrant host response to infection, resulting in multi-organ dysfunction. The application of currently available prognostic indicators for sepsis in primary hospitals is challenging. In this retrospective study, we established a novel index, the neutrophil-to-lymphocyte-to-monocyte ratio (NLMR), based on routine blood examination upon admission, and assessed its prognostic value for early mortality risk in adult patients with septic shock. Methods This study included clinical data from adult patients with septic shock who were admitted to the hospital between January 1, 2018, and December 31, 2022. Training and validation sets were constructed, and patients were categorized into "survival" and "death" groups based on their survival status within the 28-day hospitalization period. Baseline data, including demographic characteristics and comorbidities, and laboratory results, such as complete blood count parameters, were collected for analysis. The Sequential Organ Failure Assessment (SOFA) and Acute Physiology and Chronic Health Evaluation II (APACHE II) scores were documented.The NLMR was determined through the utilization of multivariate binary logistic regression analysis, leading to the development of a risk model aimed at predicting early mortality in adult patients suffering from septic shock. Results Overall, 112 adult patients with septic shock were enrolled in this study, with 84 and 28 patients in the training and validation sets, respectively. Multivariate binary logistic analysis revealed that the neutrophil, lymphocyte, and monocyte counts independently contributed to the mortality risk (odds ratios = 1.22, 0.08, and 0.16, respectively). The NLMR demonstrated an area under the receiver operating characteristic curve (ROC-AUC) of 0.83 for internal validation in the training set and 0.97 for external validation in the validation set. Both overall model quality values were significantly high at 0.74 and 0.91, respectively (P < 0.05). NLMR exhibited a higher ROC-AUC value of 0.88 than quick SOFA (ROC-AUC = 0.71), SOFA (ROC-AUC = 0.83), and APACHE II (ROC-AUC = 0.78). Conclusion NLMR may be a potential marker for predicting the risk of early death in adult patients with septic shock, warranting further exploration and verification.
Collapse
Affiliation(s)
- Xiao-ming Lin
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Lian-fang Zhang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Yu-ting Wang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Ting Huang
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Xue-feng Lin
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Xiang-yu Hong
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Hong-jun Zheng
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Rong-cheng Xie
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
| | - Jie-fei Ma
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, Fujian province, PR China
- Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| |
Collapse
|
7
|
Quin C, DeJong EN, Cook EK, Luo YZ, Vlasschaert C, Sadh S, McNaughton AJ, Buttigieg MM, Breznik JA, Kennedy AE, Zhao K, Mewburn J, Dunham-Snary KJ, Hindmarch CC, Bick AG, Archer SL, Rauh MJ, Bowdish DM. Neutrophil-mediated innate immune resistance to bacterial pneumonia is dependent on Tet2 function. J Clin Invest 2024; 134:e171002. [PMID: 38573824 PMCID: PMC11142737 DOI: 10.1172/jci171002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 03/27/2024] [Indexed: 04/06/2024] Open
Abstract
Individuals with clonal hematopoiesis of indeterminate potential (CHIP) are at increased risk of aging related health conditions and all-cause mortality, but whether CHIP affects risk of infection is much less clear. Using UK Biobank data, we revealed a positive association between CHIP and incident pneumonia in 438,421 individuals. We show that inflammation enhanced pneumonia risk, as CHIP carriers with a hypomorphic IL6 receptor polymorphism were protected. To better characterize the pathways of susceptibility, we challenged hematopoietic Tet Methylcytosine Dioxygenase 2-knockout (Tet2-/-) and floxed control mice (Tet2fl/fl) with Streptococcus pneumoniae. As with human CHIP carriers, Tet2-/- mice had hematopoietic abnormalities resulting in the expansion of inflammatory monocytes and neutrophils in peripheral blood. Yet, these cells were insufficient in defending against S. pneumoniae and resulted in increased pathology, impaired bacterial clearance, and higher mortality in Tet2-/- mice. We delineated the transcriptional landscape of Tet2-/- neutrophils and found that, while inflammation-related pathways were upregulated in Tet2-/- neutrophils, migration and motility pathways were compromised. Using live-imaging techniques, we demonstrated impairments in motility, pathogen uptake, and neutrophil extracellular trap (NET) formation by Tet2-/- neutrophils. Collectively, we show that CHIP is a risk factor for bacterial pneumonia related to innate immune impairments.
Collapse
Affiliation(s)
- Candice Quin
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Erica N. DeJong
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada
| | - Elina K. Cook
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences
| | - Yi Zhen Luo
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences
| | | | - Sanathan Sadh
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences
| | | | - Marco M. Buttigieg
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences
| | - Jessica A. Breznik
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada
| | - Allison E. Kennedy
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada
| | - Kevin Zhao
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada
| | | | | | - Charles C.T. Hindmarch
- Department of Medicine
- Queen’s CardioPulmonary Unit, Queen’s University, Kingston, Ontario, Canada
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Stephen L. Archer
- Department of Medicine
- Queen’s CardioPulmonary Unit, Queen’s University, Kingston, Ontario, Canada
| | - Michael J. Rauh
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences
| | - Dawn M.E. Bowdish
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare, Hamilton, Ontario, Canada
| |
Collapse
|
8
|
Miao S, Chang Z, Gu B, Jiang J, Pei F, Liu Y, Zhou Y, Liu Z, Si X, Guan X, Wu J. GENERATION OF TOLEROGENIC DENDRITIC CELLS UNDER THE PERSISTENT INFLAMMATION STIMULATION. Shock 2024; 61:454-464. [PMID: 38412105 DOI: 10.1097/shk.0000000000002318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
ABSTRACT Immunosuppression, commonly accompanied by persistent inflammation, is a key feature in the later phase of sepsis. However, the pathophysiological mechanisms underlying this phenomenon remain unclear. Dendritic cells (DCs), specifically tolerogenic DCs (tolDCs), play a crucial role in this process by regulating immune responses through inducing T cell anergy and releasing anti-inflammatory cytokines. Nevertheless, the existing cell models are inadequate for investigating tolDCs during the immunosuppressive phase of sepsis. Therefore, this study aimed to develop a novel in vitro model to generate tolDCs under chronic inflammatory conditions. We have successfully generated tolDCs by exposing them to sublethal lipopolysaccharide (LPS) for 72 h while preserving cell viability. Considering that IL-10-induced tolDCs (IL-10-tolDCs) are well-established models, we compared the immunological tolerance between LPS-tolDCs and IL-10-tolDCs. Our findings indicated that both LPS-tolDCs and IL-10-tolDCs exhibited reduced expression of maturation markers, whereas their levels of inhibitory markers were elevated. Furthermore, the immunoregulatory activities of LPS-tolDCs and IL-10-tolDCs were found to be comparable. These dysfunctions include impaired antigen presenting capacity and suppression of T cell activation, proliferation, and differentiation. Notably, compared with IL-10-tolDCs, LPS-tolDCs showed a reduced response in maturation and cytokine production upon stimulation, indicating their potential as a better model for research. Overall, in comparison with IL-10-tolDCs, our data suggest that the immunological dysfunctions shown in LPS-tolDCs could more effectively elucidate the increased susceptibility to secondary infections during sepsis. Consequently, LPS-tolDCs have emerged as promising therapeutic targets for ameliorating the immunosuppressed state in septic patients.
Collapse
|
9
|
Li T, Xu B, Li W, Cheng X, Tantai W, Zheng H, Zhao L, Li N, Han C. Allosteric inhibitor of SHP2 enhances macrophage endocytosis and bacteria elimination by increasing caveolae activation and protects against bacterial sepsis. Pharmacol Res 2024; 201:107096. [PMID: 38320736 DOI: 10.1016/j.phrs.2024.107096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
The uncontrolled bacterial infection-induced cytokine storm and sequential immunosuppression are commonly observed in septic patients, which indicates that the activation of phagocytic cells and the efficient and timely elimination of bacteria are crucial for combating bacterial infections. However, the role of dysregulated immune cells and their disrupted function in sepsis remains unclear. Here, we found that macrophages exhibited the impaired endocytosis capabilities in sepsis by Single-cell RNA sequencing and bulk RNA sequencing. Caveolae protein Caveolin-1 (Cav-1) of macrophages was inactivated by SHP2 rapidly during Escherichia coli (E.coli) infection. Allosteric inhibitor of SHP2 effectively maintains Cav-1 phosphorylation to enhance macrophage to endocytose and eliminate bacteria. Additionally, TLR4 endocytosis of macrophage was also enhanced upon E.coli infection by SHP099, inducing an increased and rapidly resolved inflammatory response. In vivo, pretreatment or posttreatment with inhibitor of SHP2 significantly reduced the bacterial burden in organs and mortality of mice subjected E.coli infection or CLP-induced sepsis. The cotreatment of inhibitor of SHP2 with an antibiotic conferred complete protection against mortality in mice. Our findings suggest that Cav-1-mediated endocytosis and bacterial elimination may play a critical role in the pathogenesis of sepsis, highlighting inhibitor of SHP2 as a potential therapeutic agent for sepsis.
Collapse
Affiliation(s)
- Tianliang Li
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China
| | - Bing Xu
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Wenqian Li
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaotao Cheng
- Suzhou Jizhi Medical Corporation, Jiangsu 215400, China
| | - Wenjing Tantai
- Department of Histology and Embryology and Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai 200433, China
| | - Haiyan Zheng
- Department of Emergency, Dongfang Hospital, Tongji University, Shanghai 200210, China
| | - Liming Zhao
- Department of Emergency, Dongfang Hospital, Tongji University, Shanghai 200210, China.
| | - Nan Li
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China.
| | - Chaofeng Han
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Naval Medical University, Shanghai 200433, China; Department of Histology and Embryology and Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
10
|
Yanar KE, Gür C, Değirmençay Ş, Aydın Ö, Aktaş MS, Baysal S. Insulin-like growth factor-1 expression levels in pro-inflammatory response in calves with neonatal systemic inflammatory response syndrome. Vet Immunol Immunopathol 2024; 268:110706. [PMID: 38159440 DOI: 10.1016/j.vetimm.2023.110706] [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: 11/17/2023] [Revised: 12/13/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
The objective of this study was to investigate the mRNA expression of insulin-like growth factor-1 (IGF-1), pro-inflammatory cytokines (IL-1β, IL-6, IL-18, and TNF-α), serum immunoglobulin profiles (IgG and IgM), and lipid peroxidation status (MDA) in relation to pro-inflammatory cytokines. A case-controlled, prospective, and observational investigation was completed on 85 calves. Total RNA was isolated from whole blood samples of both the SIRS and healthy calves, followed by reverse transcription into cDNA. The resulting cDNAs were mixed with iTaq Universal SYBR Green Supermix and primers specific to the relevant genes using the Rotor-Gene Q instrument. After the reaction was completed, gene expressions were normalised against β-actin using the 2-ΔΔCT method. The mRNA levels of pro-inflammatory cytokines namely (IL-1β [SIRS: 2.15 ± 0.55, Control: 1.13 ± 0.62; P = 0.001], IL-6 [SIRS: 2.82 ± 0.52, Control: 0.91 ± 0.11; P < 0.001], IL-18 [SIRS: 1.92 ± 0.41, Control: 0.99 ± 0.13; P < 0.001], and TNF-α [SIRS: 2.59 ± 0.28, Control: 0.93 ± 0.09; P < 0.001]) and IGF-1 (SIRS: 3.55 ± 0.55, Control: 0.91 ± 0.15; P < 0.001) were up-regulated in calves with SIRS, while serum IgG (SIRS: 4.16 ± 0.26, Control: 1.73 ± 0.17; P < 0.001), IgM (SIRS: 1.55 ± 0.11, Control: 1.09 ± 0.13; P < 0.001), and MDA levels (SIRS: 41.12 ± 3.48, Control: 3.76 ± 0.81; P < 0.001) increased significantly in these calves. Furthermore, significant (P < 0.01) positive correlations were found in calves with SIRS in relation to the expression levels of IL-1β, IL-6, IL-18, TNF-α, IGF-1, serum immunoglobulins, and MDA levels. These results suggest that IGF-1 could be a valuable pro-inflammatory marker, considering its high positive correlation with the expression levels of pro-inflammatory cytokines (IL-1β, IL-6, IL-18, and TNF-α) and markers (MDA, IgG, and IgM) in calves with SIRS.
Collapse
Affiliation(s)
- Kerim Emre Yanar
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Cihan Gür
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Şükrü Değirmençay
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Ömer Aydın
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Mustafa Sinan Aktaş
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sümeyye Baysal
- Department of Internal Medicine, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
11
|
Marcello M, Virzì GM, Marturano D, de Cal M, Marchionna N, Sgarabotto L, De Rosa S, Ronco C, Zanella M. The Cytotoxic Effect of Septic Plasma on Healthy RBCs: Is Eryptosis a New Mechanism for Sepsis? Int J Mol Sci 2023; 24:14176. [PMID: 37762478 PMCID: PMC10531772 DOI: 10.3390/ijms241814176] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Sepsis is a life-threatening multiple-organ dysfunction induced by infection and is one of the leading causes of mortality and critical illness worldwide. The pathogenesis of sepsis involves the alteration of several biochemical pathways such as immune response, coagulation, dysfunction of endothelium and tissue damage through cellular death and/or apoptosis. Recently, in vitro and in vivo studies reported changes in the morphology and in the shape of human red blood cells (RBCs) causing erythrocyte death (eryptosis) during sepsis. Characteristics of eryptosis include cell shrinkage, membrane blebbing, and surface exposure to phosphatidylserine (PS), which attract macrophages. The aim of this study was to evaluate the in vitro induction of eryptosis on healthy RBCs exposed to septic plasma at different time points. Furthermore, we preliminary investigated the in vivo levels of eryptosis in septic patients and its relationship with Endotoxin Activity Assay (EAA), mortality and other biological markers of inflammation and oxidative stress. We enrolled 16 septic patients and 16 healthy subjects (no systemic inflammation in the last 3 months) as a control group. At diagnosis, we measured Interleukin-6 (IL-6) and Myeloperoxidase (MPO). For in vitro study, healthy RBCs were exposed to the plasma of septic patients and CTR for 15 min, 1, 2, 4 and 24 h. Morphological markers of death and eryptosis were evaluated by flow cytometric analyses. The cytotoxic effect of septic plasma on RBCs was studied in vitro at 15 min, 1, 2, 4 and 24 h. Healthy RBCs incubated with plasma from septic patients went through significant morphological changes and eryptosis compared to those exposed to plasma from the control group at all time points (all, p < 0.001). IL-6 and MPO levels were significantly higher in septic patients than in controls (both, p < 0.001). The percentage of AnnexinV-binding RBCs was significantly higher in septic patients with EAA level ≥0.60 (positive EAA: 32.4%, IQR 27.6-36.2) compared to septic patients with EAA level <0.60 (negative EAA: 14.7%, IQR 5.7-30.7) (p = 0.04). Significant correlations were observed between eryptosis and EAA levels (Spearman rho2 = 0.50, p < 0.05), IL-6 (Spearman rho2 = 0.61, p < 0.05) and MPO (Spearman rho2 = 0.70, p < 0.05). In conclusion, we observed a quick and great cytotoxic effect of septic plasma on healthy RBCs and a strong correlation with other biomarkers of severity of sepsis. Based on these results, we confirmed the pathological role of eryptosis in sepsis and we hypothesized its use as a biomarker of sepsis, potentially helping physicians to face important treatment decisions.
Collapse
Affiliation(s)
- Matteo Marcello
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| | - Grazia Maria Virzì
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| | - Davide Marturano
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
- Nephrology, Dialysis and Transplantation Unit, Department of Medicine, University of Padova, 35100 Padova, Italy
| | - Massimo de Cal
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| | - Nicola Marchionna
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| | - Luca Sgarabotto
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| | - Silvia De Rosa
- Centre for Medical Sciences-CISMed, University of Trento, Via S. Maria Maddalena 1, 38122 Trento, Italy
- Anesthesia and Intensive Care, Santa Chiara Regional Hospital, APSS, 38122 Trento, Italy
| | - Claudio Ronco
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| | - Monica Zanella
- Department of Nephrology, Dialysis and Transplant, St Bortolo Hospital, 36100 Vicenza, Italy (M.Z.)
- IRRIV-International Renal Research Institute, 36100 Vicenza, Italy
| |
Collapse
|
12
|
Grey EL, McClendon J, Suresh J, Alper S, Janssen WJ, Bryant SJ. Thiol-Michael Addition Microparticles: Their Synthesis, Characterization, and Uptake by Macrophages. ACS Biomater Sci Eng 2023; 9:4223-4240. [PMID: 37379254 PMCID: PMC10619202 DOI: 10.1021/acsbiomaterials.3c00441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Polymeric microparticles are promising biomaterial platforms for targeting macrophages in the treatment of disease. This study investigates microparticles formed by a thiol-Michael addition step-growth polymerization reaction with tunable physiochemical properties and their uptake by macrophages. The hexafunctional thiol monomer dipentaerythritol hexa-3-mercaptopropionate (DPHMP) and tetrafunctional acrylate monomer di(trimethylolpropane) tetraacrylate (DTPTA) were reacted in a stepwise dispersion polymerization, achieving tunable monodisperse particles over a size range (1-10 μm) relevant for targeting macrophages. An off-stoichiometry thiol-acrylate reaction afforded facile secondary chemical functionalization to create particles with different chemical moieties. Uptake of the microparticles by RAW 264.7 macrophages was highly dependent on treatment time, particle size, and particle chemistry with amide, carboxyl, and thiol terminal chemistries. The amide-terminated particles were non-inflammatory, while the carboxyl- and thiol-terminated particles induced pro-inflammatory cytokine production in conjunction with particle phagocytosis. Finally, a lung-specific application was explored through time-dependent uptake of amide-terminated particles by human alveolar macrophages in vitro and mouse lungs in vivo without inducing inflammation. The findings demonstrate a promising microparticulate delivery vehicle that is cyto-compatible, is non-inflammatory, and exhibits high rates of uptake by macrophages.
Collapse
Affiliation(s)
- Emerson L. Grey
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Jazalle McClendon
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Joshita Suresh
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| | - Scott Alper
- Department of Immunology and Genomic Medicine, Center for Genes, Environment and Health, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - William J. Janssen
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12631 East 17th Avenue, Aurora, CO 80045, USA
| | - Stephanie J. Bryant
- Department of Chemical and Biological Engineering, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
- Materials Science & Engineering Program, University of Colorado, 4001 Discovery Dr, Boulder, CO 80309-0613, USA
- BioFrontiers Institute, University of Colorado, 3415 Colorado Ave, Boulder, CO 80309-0596, USA
| |
Collapse
|
13
|
Nagaraju N, Varma A, Taksande A, Meshram RJ. Bone Marrow Changes in Septic Shock: A Comprehensive Review. Cureus 2023; 15:e42517. [PMID: 37637609 PMCID: PMC10457471 DOI: 10.7759/cureus.42517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
Septic shock is a life-threatening condition characterized by systemic inflammation resulting from a severe infection. Although the primary focus of sepsis research has traditionally been on the dysfunctional immune response, recent studies have highlighted the important role of bone marrow in the pathophysiology of septic shock. The bone marrow, traditionally regarded as the hematopoietic organ responsible for blood cell production, undergoes significant changes during sepsis, contributing to the overall immune dysregulation observed in this condition. This comprehensive review aims to provide a detailed overview of the bone marrow changes associated with septic shock. It explores the alterations in the bone marrow microenvironment, hematopoietic progenitor cells, and the subsequent effects on leukocyte production and function. Key cellular and molecular mechanisms involved in bone marrow dysfunction during septic shock are discussed, including the dysregulation of cytokines, chemokines, growth factors, and signaling pathways. Furthermore, this review highlights the clinical implications of bone marrow changes in septic shock. It emphasizes the impact of altered hematopoiesis on immune cell populations, such as neutrophils, monocytes, and lymphocytes, and their role in the progression and outcome of sepsis. The potential prognostic value of bone marrow parameters and the therapeutic implications of targeting bone marrow dysfunction are also addressed. The review summarizes relevant preclinical and clinical studies to comprehensively understand the current knowledge of bone marrow changes in septic shock. The limitations and challenges of studying bone marrow in the context of sepsis are acknowledged, and future directions for research are proposed.
Collapse
Affiliation(s)
- Nimmanagoti Nagaraju
- Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Ashish Varma
- Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Amar Taksande
- Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Revat J Meshram
- Paediatrics, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
14
|
Han L, Wu X, Wang O, Luan X, Velander WH, Aynardi M, Halstead ES, Bonavia AS, Jin R, Li G, Li Y, Wang Y, Dong C, Lei Y. Mesenchymal stromal cells and alpha-1 antitrypsin have a strong synergy in modulating inflammation and its resolution. Theranostics 2023; 13:2843-2862. [PMID: 37284443 PMCID: PMC10240832 DOI: 10.7150/thno.83942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/25/2023] [Indexed: 06/08/2023] Open
Abstract
Rationale: Trauma, surgery, and infection can cause severe inflammation. Both dysregulated inflammation intensity and duration can lead to significant tissue injuries, organ dysfunction, mortality, and morbidity. Anti-inflammatory drugs such as steroids and immunosuppressants can dampen inflammation intensity, but they derail inflammation resolution, compromise normal immunity, and have significant adverse effects. The natural inflammation regulator mesenchymal stromal cells (MSCs) have high therapeutic potential because of their unique capabilities to mitigate inflammation intensity, enhance normal immunity, and accelerate inflammation resolution and tissue healing. Furthermore, clinical studies have shown that MSCs are safe and effective. However, they are not potent enough, alone, to completely resolve severe inflammation and injuries. One approach to boost the potency of MSCs is to combine them with synergistic agents. We hypothesized that alpha-1 antitrypsin (A1AT), a plasma protein used clinically and has an excellent safety profile, was a promising candidate for synergism. Methods: This investigation examined the efficacy and synergy of MSCs and A1AT to mitigate inflammation and promote resolution, using in vitro inflammatory assay and in vivo mouse acute lung injury model. The in vitro assay measured cytokine releases, inflammatory pathways, reactive oxygen species (ROS), and neutrophil extracellular traps (NETs) production by neutrophils and phagocytosis in different immune cell lines. The in vivo model monitored inflammation resolution, tissue healing, and animal survival. Results: We found that the combination of MSCs and A1AT was much more effective than each component alone in i) modulating cytokine releases and inflammatory pathways, ii) inhibiting ROS and NETs production by neutrophils, iii) enhancing phagocytosis and, iv) promoting inflammation resolution, tissue healing, and animal survival. Conclusion: These results support the combined use of MSCs, and A1AT is a promising approach for managing severe, acute inflammation.
Collapse
Affiliation(s)
- Li Han
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University; University Park, PA, 16802, USA
| | - Xinran Wu
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
| | - Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln; Lincoln, NE, 68588, USA
| | - Xiao Luan
- Biomedical Center of Qingdao University; Qingdao, Shandong, 266000, China
| | - William H. Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska-Lincoln; Lincoln, NE, 68588, USA
| | - Michael Aynardi
- Department of Orthopedics Surgery, Pennsylvania State University College of Medicine; Hershey, PA, 17033, USA
| | - E. Scott Halstead
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Anthony S. Bonavia
- Division of Critical Care Medicine, Department of Anesthesiology and Perioperative Medicine, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Rong Jin
- Department of Neurosurgery, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Guohong Li
- Department of Neurosurgery, Pennsylvania State Milton S Hershey Medical Center; Hershey, PA, 17033, USA
| | - Yulong Li
- Department of Emergency Medicine, University of Nebraska Medical Center; Omaha, NE, 68105, USA
| | - Yong Wang
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
| | - Cheng Dong
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
| | - Yuguo Lei
- Department of Biomedical Engineering, Pennsylvania State University; University Park, PA, 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University; University Park, PA, 16802, USA
| |
Collapse
|
15
|
Ma Y, Kemp SS, Yang X, Wu MH, Yuan SY. Cellular mechanisms underlying the impairment of macrophage efferocytosis. Immunol Lett 2023; 254:41-53. [PMID: 36740099 PMCID: PMC9992097 DOI: 10.1016/j.imlet.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
The phagocytosis and clearance of dying cells by macrophages, a process termed efferocytosis, is essential for both maintaining homeostasis and promoting tissue repair after infection or sterile injury. If not removed in a timely manner, uncleared cells can undergo secondary necrosis, and necrotic cells lose membrane integrity, release toxic intracellular components, and potentially induce inflammation or autoimmune diseases. Efferocytosis also initiates the repair process by producing a wide range of pro-reparative factors. Accumulating evidence has revealed that macrophage efferocytosis defects are involved in the development and progression of a variety of inflammatory and autoimmune diseases. The underlying mechanisms of efferocytosis impairment are complex, disease-dependent, and incompletely understood. In this review, we will first summarize the current knowledge about the normal signaling and metabolic processes of macrophage efferocytosis and its importance in maintaining tissue homeostasis and repair. We then will focus on analyzing the molecular and cellular mechanisms underlying efferocytotic abnormality (impairment) in disease or injury conditions. Next, we will discuss the potential molecular targets for enhanced efferocytosis in animal models of disease. To provide a balanced view, we will also discuss some deleterious effects of efferocytosis.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Scott S Kemp
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Mack H Wu
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| | - Sarah Y Yuan
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA.
| |
Collapse
|
16
|
Extracellular CIRP dysregulates macrophage bacterial phagocytosis in sepsis. Cell Mol Immunol 2023; 20:80-93. [PMID: 36471113 PMCID: PMC9794804 DOI: 10.1038/s41423-022-00961-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/12/2022] [Indexed: 12/12/2022] Open
Abstract
In sepsis, macrophage bacterial phagocytosis is impaired, but the mechanism is not well elucidated. Extracellular cold-inducible RNA-binding protein (eCIRP) is a damage-associated molecular pattern that causes inflammation. However, whether eCIRP regulates macrophage bacterial phagocytosis is unknown. Here, we reported that the bacterial loads in the blood and peritoneal fluid were decreased in CIRP-/- mice and anti-eCIRP Ab-treated mice after sepsis. Increased eCIRP levels were correlated with decreased bacterial clearance in septic mice. CIRP-/- mice showed a marked increase in survival after sepsis. Recombinant murine CIRP (rmCIRP) significantly decreased the phagocytosis of bacteria by macrophages in vivo and in vitro. rmCIRP decreased the protein expression of actin-binding proteins, ARP2, and p-cofilin in macrophages. rmCIRP significantly downregulated the protein expression of βPIX, a Rac1 activator. We further demonstrated that STAT3 and βPIX formed a complex following rmCIRP treatment, preventing βPIX from activating Rac1. We also found that eCIRP-induced STAT3 phosphorylation was required for eCIRP's action in actin remodeling. Inhibition of STAT3 phosphorylation prevented the formation of the STAT3-βPIX complex, restoring ARP2 and p-cofilin expression and membrane protrusion in rmCIRP-treated macrophages. The STAT3 inhibitor stattic rescued the macrophage phagocytic dysfunction induced by rmCIRP. Thus, we identified a novel mechanism of macrophage phagocytic dysfunction caused by eCIRP, which provides a new therapeutic target to ameliorate sepsis.
Collapse
|
17
|
Matveeva O, Nechipurenko Y, Lagutkin D, Yegorov YE, Kzhyshkowska J. SARS-CoV-2 infection of phagocytic immune cells and COVID-19 pathology: Antibody-dependent as well as independent cell entry. Front Immunol 2022; 13:1050478. [PMID: 36532011 PMCID: PMC9751203 DOI: 10.3389/fimmu.2022.1050478] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Our review summarizes the evidence that COVID-19 can be complicated by SARS-CoV-2 infection of immune cells. This evidence is widespread and accumulating at an increasing rate. Research teams from around the world, studying primary and established cell cultures, animal models, and analyzing autopsy material from COVID-19 deceased patients, are seeing the same thing, namely that some immune cells are infected or capable of being infected with the virus. Human cells most vulnerable to infection include both professional phagocytes, such as monocytes, macrophages, and dendritic cells, as well as nonprofessional phagocytes, such as B-cells. Convincing evidence has accumulated to suggest that the virus can infect monocytes and macrophages, while data on infection of dendritic cells and B-cells are still scarce. Viral infection of immune cells can occur directly through cell receptors, but it can also be mediated or enhanced by antibodies through the Fc gamma receptors of phagocytic cells. Antibody-dependent enhancement (ADE) most likely occurs during the primary encounter with the pathogen through the first COVID-19 infection rather than during the second encounter, which is characteristic of ADE caused by other viruses. Highly fucosylated antibodies of vaccinees seems to be incapable of causing ADE, whereas afucosylated antibodies of persons with acute primary infection or convalescents are capable. SARS-CoV-2 entry into immune cells can lead to an abortive infection followed by host cell pyroptosis, and a massive inflammatory cascade. This scenario has the most experimental evidence. Other scenarios are also possible, for which the evidence base is not yet as extensive, namely productive infection of immune cells or trans-infection of other non-immune permissive cells. The chance of a latent infection cannot be ruled out either.
Collapse
Affiliation(s)
- Olga Matveeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | - Denis Lagutkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Center of Phthisiopulmonology and Infectious Diseases under the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Yegor E. Yegorov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- German Red Cross Blood Service Baden-Württemberg – Hessen, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, Tomsk State University, Tomsk, Russia
| |
Collapse
|
18
|
Tomášková V, Mýtniková A, Hortová Kohoutková M, Mrkva O, Skotáková M, Šitina M, Helánová K, Frič J, Pařenica J, Šrámek V, Helán M. Prognostic value of soluble endoglin in patients with septic shock and severe COVID-19. Front Med (Lausanne) 2022; 9:972040. [PMID: 36117974 PMCID: PMC9470754 DOI: 10.3389/fmed.2022.972040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/15/2022] [Indexed: 12/01/2022] Open
Abstract
Sepsis is a clinical syndrome characterized by a dysregulated response to infection. It represents a leading cause of mortality in ICU patients worldwide. Although sepsis is in the point of interest of research for several decades, its clinical management and patient survival are improving slowly. Monitoring of the biomarkers and their combinations could help in early diagnosis, estimation of prognosis and patient's stratification and response to the treatment. Circulating soluble endoglin (sEng) is the cleaved extracellular part of transmembrane glycoprotein endoglin. As a biomarker, sEng has been tested in several pathologic conditions where its elevation was associated with endothelial dysfunction. In this study we have tested the ability of sEng to predict mortality and its correlation with other clinical characteristics in the cohort of septic shock patients (n = 37) and patients with severe COVID-19 (n = 40). In patients with COVID-19 sEng did not predict mortality or correlate with markers of organ dysfunction. In contrast, in septic shock the level of sEng was significantly higher in patients with early mortality (p = 0.019; AUC = 0.801). Moreover, sEng levels correlated with signs of circulatory failure (required dose of noradrenalin and lactate levels; p = 0.002 and 0.016, respectively). The predominant clinical problem in patients with COVID-19 was ARDS, and although they often showed signs of other organ dysfunction, circulatory failure was exceptional. This potentially explains the difference between sEng levels in COVID-19 and septic shock. In conclusion, we have confirmed that sEng may reflect the extent of the circulatory failure in septic shock patients and thus could be potentially used for the early identification of patients with the highest degree of endothelial dysfunction who would benefit from endothelium-targeted individualized therapy.
Collapse
Affiliation(s)
- Veronika Tomášková
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Alexandra Mýtniková
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | | | - Ondřej Mrkva
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Monika Skotáková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Michal Šitina
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Kateřina Helánová
- Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Cardiology, University Hospital Brno, Brno, Czechia
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
- Department of Modern Immunotherapy Research, Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Jiří Pařenica
- Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Cardiology, University Hospital Brno, Brno, Czechia
| | - Vladimír Šrámek
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Helán
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
- *Correspondence: Martin Helán
| |
Collapse
|
19
|
Bosáková V, De Zuani M, Sládková L, Garlíková Z, Jose SS, Zelante T, Hortová Kohoutková M, Frič J. Lung Organoids—The Ultimate Tool to Dissect Pulmonary Diseases? Front Cell Dev Biol 2022; 10:899368. [PMID: 35912110 PMCID: PMC9326165 DOI: 10.3389/fcell.2022.899368] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/24/2022] [Indexed: 11/15/2022] Open
Abstract
Organoids are complex multicellular three-dimensional (3D) in vitro models that are designed to allow accurate studies of the molecular processes and pathologies of human organs. Organoids can be derived from a variety of cell types, such as human primary progenitor cells, pluripotent stem cells, or tumor-derived cells and can be co-cultured with immune or microbial cells to further mimic the tissue niche. Here, we focus on the development of 3D lung organoids and their use as disease models and drug screening tools. We introduce the various experimental approaches used to model complex human diseases and analyze their advantages and disadvantages. We also discuss validation of the organoids and their physiological relevance to the study of lung diseases. Furthermore, we summarize the current use of lung organoids as models of host-pathogen interactions and human lung diseases such as cystic fibrosis, chronic obstructive pulmonary disease, or SARS-CoV-2 infection. Moreover, we discuss the use of lung organoids derived from tumor cells as lung cancer models and their application in personalized cancer medicine research. Finally, we outline the future of research in the field of human induced pluripotent stem cell-derived organoids.
Collapse
Affiliation(s)
- Veronika Bosáková
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Marco De Zuani
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Lucie Sládková
- Institute of Hematology and Blood Transfusion, Prague, Czechia
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czechia
| | - Zuzana Garlíková
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Shyam Sushama Jose
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
| | - Teresa Zelante
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Jan Frič
- International Clinical Research Center, St. Anne’s University Hospital Brno, Brno, Czechia
- Institute of Hematology and Blood Transfusion, Prague, Czechia
- *Correspondence: Jan Frič,
| |
Collapse
|
20
|
Rathinavel S, Indrakumar J, Korrapati PS, Dharmalingam S. Synthesis and fabrication of amine functionalized SBA-15 incorporated PVA/Curcumin nanofiber for skin wound healing application. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2021.128185] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
21
|
Kingren MS, Starr ME, Saito H. Divergent Sepsis Pathophysiology in Older Adults. Antioxid Redox Signal 2021; 35:1358-1375. [PMID: 34210173 PMCID: PMC8905233 DOI: 10.1089/ars.2021.0056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022]
Abstract
Significance: Both incidence and mortality rates of sepsis significantly increase with advanced age, and the majority of sepsis patients are late middle-aged or older. With the proportion of older adults rapidly increasing in developed countries, age-dependent sepsis vulnerability is an urgent medical issue. Due to an increasing life expectancy, postsepsis complications and health care costs are expected to increase as well. Recent Advances: Older patients suffer from higher sepsis incidence and mortality rates, likely resulting from frequent comorbidities, increased coagulation, dysgylcemia, and altered immune responses. Critical Issues: Despite a large number of ongoing clinical and basic research studies, there is currently no effective therapeutic strategy targeting older patients with severe sepsis. The disparity between clinical and basic studies is a problem, and this is largely due to the use of animal models lacking clinical relevance. Although the majority of sepsis cases occur in older adults, most laboratory animals used for sepsis research are very young. Further, despite the wide use of combination fluid and antibiotic treatment in intensive care unit (ICU) patients, most animal research does not include such treatment. Future Directions: Because sepsis is a systemic disease with multiple organ dysfunction, combined therapy approaches, not those targeting single pathways or single organs, are essential. As for preclinical research, it is critical to confirm new findings using aged animal models with clinically relevant ICU-like medical treatments. Antioxid. Redox Signal. 35, 1358-1375.
Collapse
Affiliation(s)
- Meagan S. Kingren
- Aging and Critical Care Research Laboratory, Departments of University of Kentucky, Lexington, Kentucky, USA
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Marlene E. Starr
- Aging and Critical Care Research Laboratory, Departments of University of Kentucky, Lexington, Kentucky, USA
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
- Surgery, University of Kentucky, Lexington, Kentucky, USA
| | - Hiroshi Saito
- Aging and Critical Care Research Laboratory, Departments of University of Kentucky, Lexington, Kentucky, USA
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, USA
- Surgery, University of Kentucky, Lexington, Kentucky, USA
- Physiology, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
22
|
Li H, Tang D, Chen J, Hu Y, Cai X, Zhang P. The Clinical Value of GDF15 and Its Prospective Mechanism in Sepsis. Front Immunol 2021; 12:710977. [PMID: 34566964 PMCID: PMC8456026 DOI: 10.3389/fimmu.2021.710977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/17/2021] [Indexed: 12/23/2022] Open
Abstract
Growth differentiation factor 15 (GDF15) is involved in the occurrence and development of many diseases, and there are few studies on its relationship with sepsis. This article aims to explore the clinical value of GDF15 in sepsis and to preliminarily explore its prospective regulatory effect on macrophage inflammation and its functions. We recruited 320 subjects (132 cases in sepsis group, 93 cases in nonsepsis group, and 95 cases in control group), then detected the serum GDF15 levels and laboratory indicators, and further investigated the correlation between GDF15 and laboratory indicators, and also analyzed the clinical value of GDF15 in sepsis diagnosis, severity assessment, and prognosis. In vitro, we used LPS to stimulate THP-1 and RAW264.7 cells to establish the inflammatory model, and detected the expression of GDF15 in the culture medium and cells under the inflammatory state. After that, we added GDF15 recombinant protein (rGDF15) pretreatment to explore its prospective regulatory effect on macrophage inflammation and its functions. The results showed that the serum GDF15 levels were significantly increased in the sepsis group, which was correlated with laboratory indexes of organ damage, coagulation indexes, inflammatory factors, and SOFA score. GDF15 also has a high AUC in the diagnosis of sepsis, which can be further improved by combining with other indicators. The dynamic monitoring of GDF15 levels can play an important role in the judgment and prognosis of sepsis. In the inflammatory state, the expression of intracellular and extracellular GDF15 increased. GDF15 can reduce the levels of cytokines, inhibit M1 polarization induced by LPS, and promote M2 polarization. Moreover, GDF15 also enhances the phagocytosis and bactericidal function of macrophages. Finally, we observed a decreased level of the phosphorylation of JAK1/STAT3 signaling pathway and the nuclear translocation of NF-κB p65 with the pretreatment of rGDF15. In summary, our study found that GDF15 has good clinical application value in sepsis and plays a protective role in the development of sepsis by regulating the functions of macrophages and inhibiting the activation of JAK1/STAT3 pathway and nuclear translocation of NF-κB p65.
Collapse
Affiliation(s)
- Huan Li
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Dongling Tang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juanjuan Chen
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanhui Hu
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Cai
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pingan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Hortová-Kohoutková M, Lázničková P, Frič J. How immune-cell fate and function are determined by metabolic pathway choice: The bioenergetics underlying the immune response. Bioessays 2020; 43:e2000067. [PMID: 33191545 DOI: 10.1002/bies.202000067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
Immune cells are highly dynamic in their response to the tissue environment. Most immune cells rapidly change their metabolic profile to obtain sufficient energy to engage in defensive or homeostatic processes. Such "immunometabolism" is governed through intermediate metabolites, and has a vital role in regulating immune-cell function. The underlying metabolic reactions are shaped by the abundance and accessibility of specific nutrients, as well as the overall metabolic status of the host. Here, we discuss how different immune-cell types gain a sufficient energy supply. We then explain how immune cells perform various functions under challenged conditions and expend energy to sustain homeostasis. Finally, we speculate on how the immune-cell metabolic profile might be modulated in health and disease, by manipulating nutrient availability. By such intervention, the recovery of patient with dysregulated immune system responses might be sped up and the fitness of an individual efficiently restored.
Collapse
Affiliation(s)
| | - Petra Lázničková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic.,Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
24
|
What's New in Shock, November 2020? Shock 2020; 54:583-585. [PMID: 33048866 DOI: 10.1097/shk.0000000000001674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
25
|
Hortová-Kohoutková M, Lázničková P, Bendíčková K, De Zuani M, Andrejčinová I, Tomášková V, Suk P, Šrámek V, Helán M, Frič J. Differences in monocyte subsets are associated with short-term survival in patients with septic shock. J Cell Mol Med 2020; 24:12504-12512. [PMID: 32949213 PMCID: PMC7686971 DOI: 10.1111/jcmm.15791] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 12/25/2022] Open
Abstract
Sepsis is characterized by dynamic changes of the immune system resulting in deregulated inflammation and failure of homoeostasis and can escalate to septic shock. Circulating monocytes and other innate immune cells are among the first ones to recognize and clear pathogens. Monocytes have an important role in sepsis and septic shock and have been studied as potential diagnostic markers. In total, forty‐two patients with septic shock were recruited and blood samples obtained within first 12 hours of ICU admission. We showed that frequency of classical and intermediate monocytes assessed at the time of admission to the intensive care unit are significantly distinct in patients with septic shock who survived longer that five days from those who died. These parameters correlate significantly with differences in serum levels of inflammatory cytokines MCP‐1, IL‐6, IL‐8, IL‐10, and IL‐18, and with the proportion of helper and cytotoxic T cells. The described changes in frequency of monocyte subsets and their activation status may predict short‐term septic shock survival and help with fast identification of the group of vulnerable patients, who may profit from tailored therapy.
Collapse
Affiliation(s)
| | - Petra Lázničková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Marco De Zuani
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Veronika Tomášková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Suk
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vladimír Šrámek
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martin Helán
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Anesthesiology and Intensive Care, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|