1
|
Tan C, Reilly B, Ma G, Murao A, Jha A, Aziz M, Wang P. Neutrophils disrupt B-1a cell homeostasis by targeting Siglec-G to exacerbate sepsis. Cell Mol Immunol 2024; 21:707-722. [PMID: 38789529 PMCID: PMC11214631 DOI: 10.1038/s41423-024-01165-7] [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: 12/12/2023] [Accepted: 04/11/2024] [Indexed: 05/26/2024] Open
Abstract
B-1a cells, an innate-like cell population, are crucial for pathogen defense and the regulation of inflammation through their release of natural IgM and IL-10. In sepsis, B-1a cell numbers are decreased in the peritoneal cavity as they robustly migrate to the spleen. Within the spleen, migrating B-1a cells differentiate into plasma cells, leading to alterations in their original phenotype and functionality. We discovered a key player, sialic acid-binding immunoglobulin-like lectin-G (Siglec-G), which is expressed predominantly on B-1a cells and negatively regulates B-1a cell migration to maintain homeostasis. Siglec-G interacts with CXCR4/CXCL12 to modulate B-1a cell migration. Neutrophils aid B-1a cell migration via neutrophil elastase (NE)-mediated Siglec-G cleavage. Human studies revealed increased NE expression in septic patients. We identified an NE cleavage sequence in silico, leading to the discovery of a decoy peptide that protects Siglec-G, preserves peritoneal B-1a cells, reduces inflammation, and enhances sepsis survival. The role of Siglec-G in inhibiting B-1a cell migration to maintain their inherent phenotype and function is compromised by NE in sepsis, offering valuable insights into B-1a cell homeostasis. Employing a small decoy peptide to prevent NE-mediated Siglec-G cleavage has emerged as a promising strategy to sustain peritoneal B-1a cell homeostasis, alleviate inflammation, and ultimately improve outcomes in sepsis patients.
Collapse
Affiliation(s)
- Chuyi Tan
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bridgette Reilly
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Gaifeng Ma
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Atsushi Murao
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Alok Jha
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA.
| |
Collapse
|
2
|
Liu J, Song K, Lin B, Chen Z, Zuo Z, Fang Y, He Q, Yao X, Liu Z, Huang Q, Guo X. HMGB1 promotes neutrophil PD-L1 expression through TLR2 and mediates T cell apoptosis leading to immunosuppression in sepsis. Int Immunopharmacol 2024; 133:112130. [PMID: 38648712 DOI: 10.1016/j.intimp.2024.112130] [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: 01/16/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
Neutrophils and T lymphocytes are closely related to occurrence of immunosuppression in sepsis. Studies have shown that neutrophil apoptosis decreases and T lymphocyte apoptosis increases in sepsis immunosuppression, but the specific mechanism involved remains unclear. In the present study, we found Toll-like Receptor 2 (TLR2) and programmed death-ligand 1 (PD-L1) were significantly activated in bone marrow neutrophils of wild-type mice after LPS treatment and that they were attenuated by treatment with C29, an inhibitor of TLR2. PD-L1 activation inhibits neutrophil apoptosis, whereas programmed death protein 1 (PD-1)activation promotes apoptosis of T lymphocytes, which leads to immunosuppression. Mechanistically, when sepsis occurs, pro-inflammatory factors and High mobility group box-1 protein (HMGB1) passively released from dead cells cause the up-regulation of PD-L1 through TLR2 on neutrophils. The binding of PD-L1 and PD-1 on T lymphocytes leads to increased apoptosis of T lymphocytes and immune dysfunction, eventually resulting in the occurrence of sepsis immunosuppression. In vivo experiments showed that the HMGB1 inhibitor glycyrrhizic acid (GA) and the TLR2 inhibitor C29 could inhibit the HMGB1/TLR2/PD-L1 pathway, and improving sepsis-induced lung injury. In summary, this study shows that HMGB1 regulates PD-L1 and PD-1 signaling pathways through TLR2, which leads to immunosuppression.
Collapse
Affiliation(s)
- Jinlian Liu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ke Song
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bingqi Lin
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhenfeng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zirui Zuo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yixing Fang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qi He
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaodan Yao
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhifeng Liu
- Department of Medical Critical Care Medicine, General Hospital of Southern Theatre Command of PLA, 2. Guangdong Branch Center, National Clinical Research Center for Geriatric Diseases (Chinese PLA General Hospital), Guangzhou, Guangdong, China.
| | - Qiaobing Huang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Xiaohua Guo
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, National Experimental Education Demonstration Center for Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
3
|
Cho Y, Lee HK, Kim J, Yoo KB, Choi J, Lee Y, Choi M. Prediction of hospital-acquired influenza using machine learning algorithms: a comparative study. BMC Infect Dis 2024; 24:466. [PMID: 38698304 PMCID: PMC11067145 DOI: 10.1186/s12879-024-09358-1] [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: 03/26/2023] [Accepted: 04/26/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Hospital-acquired influenza (HAI) is under-recognized despite its high morbidity and poor health outcomes. The early detection of HAI is crucial for curbing its transmission in hospital settings. AIM This study aimed to investigate factors related to HAI, develop predictive models, and subsequently compare them to identify the best performing machine learning algorithm for predicting the occurrence of HAI. METHODS This retrospective observational study was conducted in 2022 and included 111 HAI and 73,748 non-HAI patients from the 2011-2012 and 2019-2020 influenza seasons. General characteristics, comorbidities, vital signs, laboratory and chest X-ray results, and room information within the electronic medical record were analysed. Logistic Regression (LR), Random Forest (RF), Extreme Gradient Boosting (XGB), and Artificial Neural Network (ANN) techniques were used to construct the predictive models. Employing randomized allocation, 80% of the dataset constituted the training set, and the remaining 20% comprised the test set. The performance of the developed models was assessed using metrics such as the area under the receiver operating characteristic curve (AUC), the count of false negatives (FN), and the determination of feature importance. RESULTS Patients with HAI demonstrated notable differences in general characteristics, comorbidities, vital signs, laboratory findings, chest X-ray result, and room status compared to non-HAI patients. Among the developed models, the RF model demonstrated the best performance taking into account both the AUC (83.3%) and the occurrence of FN (four). The most influential factors for prediction were staying in double rooms, followed by vital signs and laboratory results. CONCLUSION This study revealed the characteristics of patients with HAI and emphasized the role of ventilation in reducing influenza incidence. These findings can aid hospitals in devising infection prevention strategies, and the application of machine learning-based predictive models especially RF can enable early intervention to mitigate the spread of influenza in healthcare settings.
Collapse
Affiliation(s)
- Younghee Cho
- College of Nursing, Yonsei University, Seoul, Republic of Korea
- Department of Digital Health, Samsung SDS, Seoul, Republic of Korea
| | - Hyang Kyu Lee
- College of Nursing, Yonsei University, Seoul, Republic of Korea
| | - Joungyoun Kim
- College of Engineering, University of Seoul, Seoul, Republic of Korea
| | - Ki-Bong Yoo
- Division of Health Administration, Yonsei University, Wonju, Republic of Korea
| | - Jongrim Choi
- College of Nursing, Keimyung University, Daegu, Republic of Korea
| | - Yongseok Lee
- Department of Digital Health, Samsung SDS, Seoul, Republic of Korea
| | - Mona Choi
- College of Nursing, Yonsei University, Seoul, Republic of Korea.
- Mo-Im Kim Nursing Research Institute, College of Nursing, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
4
|
Kim YS, Jeong YS, Bae GH, Kang JH, Lee M, Zabel BA, Bae YS. CD200R high neutrophils with dysfunctional autophagy establish systemic immunosuppression by increasing regulatory T cells. Cell Mol Immunol 2024; 21:349-361. [PMID: 38311677 PMCID: PMC10978921 DOI: 10.1038/s41423-024-01136-y] [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: 04/24/2023] [Revised: 12/21/2023] [Accepted: 01/13/2024] [Indexed: 02/06/2024] Open
Abstract
Distinct neutrophil populations arise during certain pathological conditions. The generation of dysfunctional neutrophils during sepsis and their contribution to septicemia-related systemic immune suppression remain unclear. In this study, using an experimental sepsis model that features immunosuppression, we identified a novel population of pathogenic CD200Rhigh neutrophils that are generated during the initial stages of sepsis and contribute to systemic immune suppression by enhancing regulatory T (Treg) cells. Compared to their CD200Rlow counterparts, sepsis-generated CD200Rhigh neutrophils exhibit impaired autophagy and dysfunction, with reduced chemotactic migration, superoxide anion production, and TNF-α production. Increased soluble CD200 blocks autophagy and neutrophil maturation in the bone marrow during experimental sepsis, and recombinant CD200 treatment in vitro can induce neutrophil dysfunction similar to that observed in CD200Rhigh neutrophils. The administration of an α-CD200R antibody effectively reversed neutrophil dysfunction by enhancing autophagy and protecting against a secondary infection challenge, leading to increased survival. Transcriptome analysis revealed that CD200Rhigh neutrophils expressed high levels of Igf1, which elicits the generation of Treg cells, while the administration of an α-CD200R antibody inhibited Treg cell generation in a secondary infection model. Taken together, our findings revealed a novel CD200Rhigh neutrophil population that mediates the pathogenesis of sepsis-induced systemic immunosuppression by generating Treg cells.
Collapse
Affiliation(s)
- Ye Seon Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yu Sun Jeong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Geon Ho Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Ji Hyeon Kang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Mingyu Lee
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research, Veterans Affairs Hospital, Palo Alto, CA, 94304, USA
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
- Department of Health Science and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
5
|
Miller M, Melis MJ, Miller JRC, Kleyman A, Shankar-Hari M, Singer M. Antibiotics, Sedatives, and Catecholamines Further Compromise Sepsis-Induced Immune Suppression in Peripheral Blood Mononuclear Cells. Crit Care Med 2024; 52:596-606. [PMID: 38483219 DOI: 10.1097/ccm.0000000000006119] [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: 05/24/2024]
Abstract
OBJECTIVES We hypothesized that the immunosuppressive effects associated with antibiotics, sedatives, and catecholamines amplify sepsis-associated immune suppression through mitochondrial dysfunction, and there is a cumulative effect when used in combination. We thus sought to determine the impact of the exemplar drugs ciprofloxacin, propofol, and norepinephrine, used alone and in combination, at clinically relevant concentrations, on the ex vivo functionality of peripheral blood mononuclear cells (PBMCs) drawn from healthy, infected, and septic individuals. DESIGN In vitro/ex vivo investigation. SETTING University laboratory. SUBJECTS Healthy volunteers, infected (nonseptic) patients in the emergency department, and septic ICU patients. INTERVENTIONS PBMCs were isolated from these subjects and treated with ciprofloxacin (100 µg/mL), propofol (50 µg/mL), norepinephrine (10 µg/mL), or all three drugs combined, with and without lipopolysaccharide (100 ng/mL) for 6 or 24 hours. Comparison was made between study groups and against untreated cells. Measurements were made of cell viability, cytokine production, phagocytosis, human leukocyte antigen-DR (HLA-DR) status, mitochondrial membrane potential, mitochondrial reactive oxygen species production, and oxygen consumption. Gene expression in immune and metabolic pathways was investigated in PBMCs sampled from healthy volunteers coincubated with septic serum. MEASUREMENTS AND RESULTS Coincubation with each of the drugs reduced cytokine production and phagocytosis in PBMCs isolated from septic patients, and healthy volunteers coincubated with septic serum. No effect was seen on HLA-DR surface expression. No cumulative effects were seen with the drug combination. Sepsis-induced changes in gene expression and mitochondrial functionality were not further affected by addition of any of the drugs. CONCLUSION Drugs commonly used in critical care lead to significant immune dysfunction ex vivo and enhance sepsis-associated immunosuppression. Further studies are required to identify underlying mechanisms and potential impact on patient outcomes.
Collapse
Affiliation(s)
- Muska Miller
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Miranda J Melis
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - James R C Miller
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Anna Kleyman
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| | - Manu Shankar-Hari
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh, United Kingdom
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, University College London, London, United Kingdom
| |
Collapse
|
6
|
Santacroce E, D’Angerio M, Ciobanu AL, Masini L, Lo Tartaro D, Coloretti I, Busani S, Rubio I, Meschiari M, Franceschini E, Mussini C, Girardis M, Gibellini L, Cossarizza A, De Biasi S. Advances and Challenges in Sepsis Management: Modern Tools and Future Directions. Cells 2024; 13:439. [PMID: 38474403 PMCID: PMC10931424 DOI: 10.3390/cells13050439] [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: 02/01/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Sepsis, a critical condition marked by systemic inflammation, profoundly impacts both innate and adaptive immunity, often resulting in lymphopenia. This immune alteration can spare regulatory T cells (Tregs) but significantly affects other lymphocyte subsets, leading to diminished effector functions, altered cytokine profiles, and metabolic changes. The complexity of sepsis stems not only from its pathophysiology but also from the heterogeneity of patient responses, posing significant challenges in developing universally effective therapies. This review emphasizes the importance of phenotyping in sepsis to enhance patient-specific diagnostic and therapeutic strategies. Phenotyping immune cells, which categorizes patients based on clinical and immunological characteristics, is pivotal for tailoring treatment approaches. Flow cytometry emerges as a crucial tool in this endeavor, offering rapid, low cost and detailed analysis of immune cell populations and their functional states. Indeed, this technology facilitates the understanding of immune dysfunctions in sepsis and contributes to the identification of novel biomarkers. Our review underscores the potential of integrating flow cytometry with omics data, machine learning and clinical observations to refine sepsis management, highlighting the shift towards personalized medicine in critical care. This approach could lead to more precise interventions, improving outcomes in this heterogeneously affected patient population.
Collapse
Affiliation(s)
- Elena Santacroce
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Miriam D’Angerio
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Linda Masini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Irene Coloretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany;
| | - Marianna Meschiari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Erica Franceschini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Cristina Mussini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| |
Collapse
|
7
|
Schofield CJ, Tirouvanziam R, Garratt LW. OMIP-100: A flow cytometry panel to investigate human neutrophil subsets. Cytometry A 2024; 105:81-87. [PMID: 38179854 DOI: 10.1002/cyto.a.24820] [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: 07/07/2023] [Revised: 11/16/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024]
Abstract
This 14-color, 13-antibody optimized multicolor immunofluorescence panel (OMIP) was designed for deep profiling of neutrophil subsets in various types of human samples to contextualize neutrophil plasticity in a range of healthy and diseased states. Markers present in the OMIP allow the profiling of neutrophil subsets associated with ontogeny, migration, phagocytosis capacity, granule release, and immune modulation. For panel design, we ensured that the commonly available fluorophores FITC/AF488, PE, and APC were assigned to the intracellular subset marker Olfactomedin 4, the maturity and activation marker CD10, and whole blood subset marker CD177, respectively. These markers can be easily replaced without affecting the core identification of neutrophils, enabling antibodies to new neutrophil antigens of interest or for fluorescent substrates to assess different neutrophil functions to be easily explored. Panel optimization was performed on whole blood and purified neutrophils. We demonstrate applications on clinical samples (whole blood and saliva) and experimental endpoints (purified neutrophils stimulated through an in vitro transmigration assay). We hope that providing a uniform platform to analyze neutrophil plasticity in various sample types will facilitate the future understanding of neutrophil subsets in health and disease.
Collapse
Affiliation(s)
- Craig J Schofield
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Center for CF & Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Luke W Garratt
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
- Medical School, University of Western Australia, Crawley, Western Australia, Australia
| |
Collapse
|
8
|
Wang L, Zhang G, Sun W, Zhang Y, Tian Y, Yang X, Liu Y. Comprehensive analysis of immune cell landscapes revealed that immune cell ratio eosinophil/B.cell.memory is predictive of survival in sepsis. Eur J Med Res 2023; 28:565. [PMID: 38053180 DOI: 10.1186/s40001-023-01506-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 11/04/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND Immune dysregulation is a feature of sepsis. However, a comprehensive analysis of the immune landscapes in septic patients has not been conducted. OBJECTIVES This study aims to explore the abundance ratios of immune cells in sepsis and investigate their clinical value. METHODS Sepsis transcriptome data sets were downloaded from the NCBI GEO database. The immunedeconv R package was employed to analyze the abundance of immune cells in sepsis patients and calculate the ratios of different immune cell types. Differential analysis of immune cell ratios was performed using the t test. The Spearman rank correlation coefficient was utilized to find the relationships between immune cell abundance and pathways. The prognostic significance of immune cell ratios for patient survival probability was assessed using the log-rank test. In addition, differential gene expression was performed using the limma package, and gene co-expression analysis was executed using the WGCNA package. RESULTS We found significant changes in immune cell ratios between sepsis patients and healthy controls. Some of these ratios were associated with 28-day survival. Certain pathways showed significant correlations with immune cell ratios. Notably, six immune cell ratios demonstrated discriminative ability for patients with systemic inflammatory response syndrome (SIRS), bacterial sepsis, and viral sepsis, with an Area Under the Curve (AUC) larger than 0.84. Patients with a high eosinophil/B.cell.memory ratio exhibited poor survival outcomes. A total of 774 differential genes were identified in sepsis patients with a high eosinophil/B.cell.memory ratio compared to those with a low ratio. These genes were organized into seven co-expression modules associated with relevant pathways, including interferon signaling, T-cell receptor signaling, and specific granule pathways. CONCLUSIONS Immune cell ratios eosinophil/B.cell.memory and NK.cell.activated/NK.cell.resting in sepsis patients can be utilized for disease subtyping, prognosis, and diagnosis. The proposed cell ratios may have higher prognostic values than the neutrophil-to-lymphocyte ratio (NLR).
Collapse
Affiliation(s)
- Lei Wang
- Microbiology and Immunology Department, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Guoan Zhang
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Wenjie Sun
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
- Cangzhou Nanobody Technology Innovation Center, Cangzhou, 061001, Hebei, China
| | - Yan Zhang
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Yi Tian
- Microbiology and Immunology Department, Cangzhou Medical College, Cangzhou, 061001, Hebei, China
| | - Xiaohui Yang
- Science and Technology Experiment Center, Cangzhou Medical College, Cangzhou, 061001, Hebei, China.
- University Nanobody Application Technology Research and Development Center of Hebei Province, Cangzhou, 061001, Hebei, China.
| | - Yingfu Liu
- University Nanobody Application Technology Research and Development Center of Hebei Province, Cangzhou, 061001, Hebei, China.
- Cangzhou Nanobody Technology Innovation Center, Cangzhou, 061001, Hebei, China.
| |
Collapse
|
9
|
Dong Y, Heng G, Zhang J, Shen Y, Lan Z, Wei K, Jin W. Association between corticosteroid use and 28-day mortality in septic shock patients with gram-negative bacterial infection: a retrospective study. Front Med (Lausanne) 2023; 10:1276181. [PMID: 38020171 PMCID: PMC10657847 DOI: 10.3389/fmed.2023.1276181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Although corticosteroids are recommended in the 2021 Surviving Sepsis Campaign (SSC) guidelines, evidence with respect to their effects on short-term mortality remains conflicting. We conducted this study to identify whether corticosteroids alter 28-day mortality in septic shock patients with gram-negative bacterial infection. Materials and methods A total of 621 patients with septic shock and gram-negative bacterial culture results were identified from the Medical Information Mart for Intensive Care IV (MIMIC-IV) database. Propensity score matching (PSM) was performed, and Kaplan-Meier survival curve analyses with log-rank tests were used to determine the relationship between corticosteroid use and the risk of 28-day mortality. Subgroup analyses were conducted to assess whether the conclusions were stable and reliable. Results Corticosteroid administration was associated with increased 28-day mortality in septic shock patients with gram-negative bacterial infection (log-rank test P = 0.028). The incidence of Stage 2 or 3 AKI and the rate of hospital mortality were higher among patients who received corticosteroids. The incidence of Stage 2 or 3 AKI in the early period significantly mediated the relationship between corticosteroid use and 28-day mortality [P =0.046 for the average causal mediation effect (ACME)]. Interaction tests indicated that the effect of corticosteroid use was maintained in patients with a neutrophil-to-lymphocyte ratio (NLR) of <20 (P-value for interaction = 0.027). Conclusion Systemic corticosteroid use could be harmful in septic shock patients with gram-negative bacterial infection, especially in patients with relatively low NLR.
Collapse
Affiliation(s)
- Yi Dong
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Gang Heng
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China
| | - Jianxin Zhang
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China
| | - Yanbing Shen
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China
| | - Zhen Lan
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China
| | - Kunchen Wei
- Department of Respiratory and Critical Care Medicine, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Weidong Jin
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Zhou W, Cao X, Xu Q, Qu J, Sun Y. The double-edged role of neutrophil heterogeneity in inflammatory diseases and cancers. MedComm (Beijing) 2023; 4:e325. [PMID: 37492784 PMCID: PMC10363828 DOI: 10.1002/mco2.325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 07/27/2023] Open
Abstract
Neutrophils are important immune cells act as the body's first line of defense against infection and respond to diverse inflammatory cues. Many studies have demonstrated that neutrophils display plasticity in inflammatory diseases and cancers. Clarifying the role of neutrophil heterogeneity in inflammatory diseases and cancers will contribute to the development of novel treatment strategies. In this review, we have presented a review on the development of the understanding on neutrophil heterogeneity from the traditional perspective and a high-resolution viewpoint. A growing body of evidence has confirmed the double-edged role of neutrophils in inflammatory diseases and tumors. This may be due to a lack of precise understanding of the role of specific neutrophil subsets in the disease. Thus, elucidating specific neutrophil subsets involved in diseases would benefit the development of precision medicine. Thusly, we have summarized the relevance and actions of neutrophil heterogeneity in inflammatory diseases and cancers comprehensively. Meanwhile, we also discussed the potential intervention strategy for neutrophils. This review is intended to deepen our understanding of neutrophil heterogeneity in inflammatory diseases and cancers, while hold promise for precise treatment of neutrophil-related diseases.
Collapse
Affiliation(s)
- Wencheng Zhou
- Department of PharmacyThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouChina
| | - Xinran Cao
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Jiao Qu
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| | - Yang Sun
- State Key Laboratory of Pharmaceutical BiotechnologyDepartment of Biotechnology and Pharmaceutical SciencesSchool of Life ScienceNanjing UniversityNanjingChina
| |
Collapse
|
11
|
de Oliveira Formiga R, Amaral FC, Souza CF, Mendes DAGB, Wanderley CWS, Lorenzini CB, Santos AA, Antônia J, Faria LF, Natale CC, Paula NM, Silva PCS, Fonseca FR, Aires L, Heck N, Starick MR, Queiroz‐Junior CM, Santos FRS, de Souza FRO, Costa VV, Barroso SPC, Morrot A, Van Weyenbergh J, Sordi R, Alisson‐Silva F, Cunha FQ, Rocha EL, Chollet‐Martin S, Hurtado‐Nedelec MM, Martin C, Burgel P, Mansur DS, Maurici R, Macauley MS, Báfica A, Witko‐Sarsat V, Spiller F. Neuraminidase is a host-directed approach to regulate neutrophil responses in sepsis and COVID-19. Br J Pharmacol 2023; 180:1460-1481. [PMID: 36526272 PMCID: PMC9877938 DOI: 10.1111/bph.16013] [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: 03/16/2022] [Revised: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Because pathogen-derived neuraminidase (NEU) stimulates neutrophils, we investigated whether host NEU can be targeted to regulate the neutrophil dysregulation observed in severe infections. EXPERIMENTAL APPROACH The effects of NEU inhibitors on lipopolysaccharide (LPS)-stimulated neutrophils from healthy donors or COVID-19 patients were determined by evaluating the shedding of surface sialic acids, cell activation, and reactive oxygen species (ROS) production. Re-analysis of single-cell RNA sequencing of respiratory tract samples from COVID-19 patients also was carried out. The effects of oseltamivir on sepsis and betacoronavirus-induced acute lung injury were evaluated in murine models. KEY RESULTS Oseltamivir and zanamivir constrained host NEU activity, surface sialic acid release, cell activation, and ROS production by LPS-activated human neutrophils. Mechanistically, LPS increased the interaction of NEU1 with matrix metalloproteinase 9 (MMP-9). Inhibition of MMP-9 prevented LPS-induced NEU activity and neutrophil response. In vivo, treatment with oseltamivir fine-tuned neutrophil migration and improved infection control as well as host survival in peritonitis and pneumonia sepsis. NEU1 also is highly expressed in neutrophils from COVID-19 patients, and treatment of whole-blood samples from these patients with either oseltamivir or zanamivir reduced neutrophil overactivation. Oseltamivir treatment of intranasally infected mice with the mouse hepatitis coronavirus 3 (MHV-3) decreased lung neutrophil infiltration, viral load, and tissue damage. CONCLUSION AND IMPLICATIONS These findings suggest that interplay of NEU1-MMP-9 induces neutrophil overactivation. In vivo, NEU may serve as a host-directed target to dampen neutrophil dysfunction during severe infections.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Flávia C. Amaral
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Camila F. Souza
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Daniel A. G. B. Mendes
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Carlos W. S. Wanderley
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Cristina B. Lorenzini
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Adara A. Santos
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Juliana Antônia
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Lucas F. Faria
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Caio C. Natale
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicholas M. Paula
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Priscila C. S. Silva
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Fernanda R. Fonseca
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Luan Aires
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicoli Heck
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Márick R. Starick
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Celso M. Queiroz‐Junior
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Felipe R. S. Santos
- Department of Biochemistry and Immunology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Filipe R. O. de Souza
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Vivian V. Costa
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Shana P. C. Barroso
- Molecular Biology Laboratory, Institute of Biomedical ResearchMarcilio Dias Naval Hospital, Navy of BrazilRio de JaneiroBrazil
| | - Alexandre Morrot
- Tuberculosis Research Laboratory, Faculty of MedicineFederal University of Rio de JaneiroRio de JaneiroBrazil
- Immunoparasitology LaboratoryOswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological VirologyKU LeuvenLeuvenBelgium
| | - Regina Sordi
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Frederico Alisson‐Silva
- Department of Immunology, Paulo de Goes Institute of MicrobiologyFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - Fernando Q. Cunha
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Edroaldo L. Rocha
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Sylvie Chollet‐Martin
- INSERM UMR 996, ‘Infammation, Microbiome and Immunosurveillance’, Faculty of PharmacyUniversité Paris‐SaclayChâtenay‐MalabryFrance
| | | | - Clémence Martin
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Pierre‐Régis Burgel
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Daniel S. Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Rosemeri Maurici
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonAlbertaCanada
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | | | - Fernando Spiller
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| |
Collapse
|
12
|
Melis MJ, Miller M, Peters VBM, Singer M. The role of hormones in sepsis: an integrated overview with a focus on mitochondrial and immune cell dysfunction. Clin Sci (Lond) 2023; 137:707-725. [PMID: 37144447 PMCID: PMC10167421 DOI: 10.1042/cs20220709] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/09/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Sepsis is a dysregulated host response to infection that results in life-threatening organ dysfunction. Virtually every body system can be affected by this syndrome to greater or lesser extents. Gene transcription and downstream pathways are either up- or downregulated, albeit with considerable fluctuation over the course of the patient's illness. This multi-system complexity contributes to a pathophysiology that remains to be fully elucidated. Consequentially, little progress has been made to date in developing new outcome-improving therapeutics. Endocrine alterations are well characterised in sepsis with variations in circulating blood levels and/or receptor resistance. However, little attention has been paid to an integrated view of how these hormonal changes impact upon the development of organ dysfunction and recovery. Here, we present a narrative review describing the impact of the altered endocrine system on mitochondrial dysfunction and immune suppression, two interlinked and key aspects of sepsis pathophysiology.
Collapse
Affiliation(s)
- Miranda J Melis
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Muska Miller
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Vera B M Peters
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| | - Mervyn Singer
- Bloomsbury Institute of Intensive Care Medicine, Division of Medicine, University College London, London, UK
| |
Collapse
|
13
|
Grudzinska FS, Jasper A, Sapey E, Thickett DR, Mauro C, Scott A, Barlow J. Real-time assessment of neutrophil metabolism and oxidative burst using extracellular flux analysis. Front Immunol 2023; 14:1083072. [PMID: 37180154 PMCID: PMC10166867 DOI: 10.3389/fimmu.2023.1083072] [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: 10/28/2022] [Accepted: 04/05/2023] [Indexed: 05/15/2023] Open
Abstract
Neutrophil responses are critical during inflammatory and infective events, and neutrophil dysregulation has been associated with poor patient outcomes. Immunometabolism is a rapidly growing field that has provided insights into cellular functions in health and disease. Neutrophils are highly glycolytic when activated, with inhibition of glycolysis associated with functional deficits. There is currently very limited data available assessing metabolism in neutrophils. Extracellular flux (XF) analysis assesses real time oxygen consumption and the rate of proton efflux in cells. This technology allows for the automated addition of inhibitors and stimulants to visualise the effect on metabolism. We describe optimised protocols for an XFe96 XF Analyser to (i) probe glycolysis in neutrophils under basal and stimulated conditions, (ii) probe phorbol 12-myristate 13-acetate induced oxidative burst, and (iii) highlight challenges of using XF technology to examine mitochondrial function in neutrophils. We provide an overview of how to analyze XF data and identify pitfalls of probing neutrophil metabolism with XF analysis. In summary we describe robust methods for assessing glycolysis and oxidative burst in human neutrophils and discuss the challenges around using this technique to assess mitochondrial respiration. XF technology is a powerful platform with a user-friendly interface and data analysis templates, however we suggest caution when assessing neutrophil mitochondrial respiration.
Collapse
Affiliation(s)
- Frances S. Grudzinska
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Alice Jasper
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- PIONEER Health Data Research- UK Hub in Acute Care, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - David R. Thickett
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Claudio Mauro
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Aaron Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Jonathan Barlow
- Cellular Health and Metabolism Facility, School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
14
|
Distinct subsets of neutrophils crosstalk with cytokines and metabolites in patients with sepsis. iScience 2023; 26:105948. [PMID: 36756375 PMCID: PMC9900520 DOI: 10.1016/j.isci.2023.105948] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/04/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
Sepsis is a life-threatening condition caused by a dysregulated host response to infection. Despite continued efforts to understand the pathophysiology of sepsis, no effective therapies are currently available. While singular components of the aberrant immune response have been investigated, comprehensive studies linking different data layers are lacking. Using an integrated systems immunology approach, we evaluated neutrophil phenotypes and concomitant changes in cytokines and metabolites in patients with sepsis. Our findings identify differentially expressed mature and immature neutrophil subsets in patients with sepsis. These subsets correlate with various proteins, metabolites, and lipids, including pentraxin-3, angiopoietin-2, and lysophosphatidylcholines, in patients with sepsis. These results enabled the construction of a statistical model based on weighted multi-omics linear regression analysis for sepsis biomarker identification. These findings could help inform early patient stratification and treatment options, and facilitate further mechanistic studies targeting the trifecta of surface marker expression, cytokines, and metabolites.
Collapse
|
15
|
de Oliveira Formiga R, Amaral FC, Souza CF, Mendes DAGB, Wanderley CWS, Lorenzini CB, Santos AA, Antônia J, Faria LF, Natale CC, Paula NM, Silva PCS, Fonseca FR, Aires L, Heck N, Starick MR, Queiroz-Junior CM, Santos FRS, de Souza FRO, Costa VV, Barroso SPC, Morrot A, Van Weyenbergh J, Sordi R, Alisson-Silva F, Cunha FQ, Rocha EL, Chollet-Martin S, Hurtado-Nedelec MM, Martin C, Burgel PR, Mansur DS, Maurici R, Macauley MS, Báfica A, Witko-Sarsat V, Spiller F. Neuraminidase inhibitors rewire neutrophil function in vivo in murine sepsis and ex vivo in COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2020.11.12.379115. [PMID: 33200130 PMCID: PMC7668734 DOI: 10.1101/2020.11.12.379115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Neuraminidase (NEU)-mediated cleavage of surface sialic acid has been demonstrated to regulate leukocyte responses. Here, we report that antiviral NEU inhibitors constrain host NEU activity, surface sialic acid release, ROS production, and NETs released by microbial-activated human neutrophils. In vivo, treatment with Oseltamivir results in infection control and host survival in peritonitis and pneumonia models of sepsis. Single-cell RNA sequencing re-analysis of publicly data sets of respiratory tract samples from critical COVID-19 patients revealed an overexpression of NEU1 in infiltrated neutrophils. Moreover, Oseltamivir or Zanamivir treatment of whole blood cells from severe COVID-19 patients reduces host NEU-mediated shedding of cell surface sialic acid and neutrophil overactivation. These findings suggest that neuraminidase inhibitors can serve as host-directed interventions to dampen neutrophil dysfunction in severe infections.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Université de Paris, Institut Cochin, INSERM U1016, CNRS, Paris, France
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Flávia C. Amaral
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Camila F. Souza
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Daniel A. G. B. Mendes
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Carlos W. S. Wanderley
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Cristina B. Lorenzini
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Adara A. Santos
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Juliana Antônia
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Lucas F. Faria
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Caio C. Natale
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Nicholas M. Paula
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Priscila C. S. Silva
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Fernanda R. Fonseca
- Department of Clinical Medicine, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Luan Aires
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Nicoli Heck
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Márick R. Starick
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Celso M. Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Felipe R. S. Santos
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Filipe R. O. de Souza
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian V. Costa
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Shana P. C. Barroso
- Molecular Biology Laboratory, Institute of Biomedical Research, Marcilio Dias Naval Hospital, Navy of Brazil, RJ, Brazil
| | - Alexandre Morrot
- Tuberculosis Research Laboratory, Faculty of Medicine, Federal University of Rio de Janeiro
- Immunoparasitology Laboratory, Oswaldo Cruz Foundation, FIOCRUZ, Rio de Janeiro, Brazil
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological Virology, KU Leuven, Leuven, Belgium
| | - Regina Sordi
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Frederico Alisson-Silva
- Department of Immunology, Paulo de Goes Institute of Microbiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Edroaldo L. Rocha
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Sylvie Chollet-Martin
- INSERM UMR 996, “Infammation, Microbiome and Immunosurveillance”, Faculty of Pharmacy, Université Paris-Saclay, Châtenay-Malabry, France
| | | | - Clémence Martin
- Université de Paris, Institut Cochin, INSERM U1016, CNRS, Paris, France
- Department of Pneumology, AP-HP, Hôpital Cochin, Paris, France
| | - Pierre-Régis Burgel
- Université de Paris, Institut Cochin, INSERM U1016, CNRS, Paris, France
- Department of Pneumology, AP-HP, Hôpital Cochin, Paris, France
| | - Daniel S. Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Rosemeri Maurici
- Department of Clinical Medicine, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | | | - Fernando Spiller
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| |
Collapse
|
16
|
Lu J, Liu J, Zhu L, Zhang Y, Li A. The effect of age on the clinical characteristics and innate immune cell function in the patients with abdominal sepsis. Front Physiol 2022; 13:952434. [PMID: 36237524 PMCID: PMC9551265 DOI: 10.3389/fphys.2022.952434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
Sepsis is a life-threatening dysregulated host response to infection that compromises organ health, and abdominal sepsis is a commonly presenting critical illness in intensive care units (ICU). In this study, we investigate the effect of age on clinical sepsis characteristics and innate immune cells (neutrophils and monocytes) functionality in abdominal sepsis patients. We recruited 32 patients with abdominal sepsis from the Beijing Ditan Hospital’s ICU from February 2021 to September 2021, and selected 18 healthy volunteers that were age- and sex-matched as controls for a prospective cohort study. Elderly abdominal sepsis patients (age >65 years) had the following altered characteristics compared to nonelderly patient controls: lower mean arterial pressure, monocytes percentage, and red blood cell volume distribution width (p < 0.05); higher neutrophils percentage and neutrophils-to-lymphocytes ratio (p < 0.05); significantly increased monocyte-produced reactive oxygen (p < 0.05); increases neutrophilic secretion of TNF-α, as well as lower monocytic secretion of TNF-α (p < 0.05); higher neutrophil percentage (which was significantly higher in peripheral blood than monocyte percentage). Elderly patients also had significantly increased phagocytic activity in their neutrophils and monocytes (p < 0.05), significantly reduced neutrophils-produced reactive oxygen (p < 0.001), and significantly increased TNF-α secretion by monocytes and neutrophils (p < 0.05). We found that elderly patients have decreased immune cell function and increased release of cytokines compared to younger patients, suggesting individualized treatment plans targeting the elderly septic microenvironment could help prevent organ failure in elderly septic patients and improves patient survival.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Critical Care Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jingyuan Liu
- Department of Critical Care Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Liuluan Zhu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yue Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ang Li
- Department of Critical Care Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Ang Li,
| |
Collapse
|
17
|
Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol 2022; 19:1079-1101. [PMID: 36056148 PMCID: PMC9508259 DOI: 10.1038/s41423-022-00902-0] [Citation(s) in RCA: 151] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractThe immune-inflammatory response is associated with increased nitro-oxidative stress. The aim of this mechanistic review is to examine: (a) the role of redox-sensitive transcription factors and enzymes, ROS/RNS production, and the activity of cellular antioxidants in the activation and performance of macrophages, dendritic cells, neutrophils, T-cells, B-cells, and natural killer cells; (b) the involvement of high-density lipoprotein (HDL), apolipoprotein A1 (ApoA1), paraoxonase-1 (PON1), and oxidized phospholipids in regulating the immune response; and (c) the detrimental effects of hypernitrosylation and chronic nitro-oxidative stress on the immune response. The redox changes during immune-inflammatory responses are orchestrated by the actions of nuclear factor-κB, HIF1α, the mechanistic target of rapamycin, the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, mitogen-activated protein kinases, 5' AMP-activated protein kinase, and peroxisome proliferator-activated receptor. The performance and survival of individual immune cells is under redox control and depends on intracellular and extracellular levels of ROS/RNS. They are heavily influenced by cellular antioxidants including the glutathione and thioredoxin systems, nuclear factor erythroid 2-related factor 2, and the HDL/ApoA1/PON1 complex. Chronic nitro-oxidative stress and hypernitrosylation inhibit the activity of those antioxidant systems, the tricarboxylic acid cycle, mitochondrial functions, and the metabolism of immune cells. In conclusion, redox-associated mechanisms modulate metabolic reprogramming of immune cells, macrophage and T helper cell polarization, phagocytosis, production of pro- versus anti-inflammatory cytokines, immune training and tolerance, chemotaxis, pathogen sensing, antiviral and antibacterial effects, Toll-like receptor activity, and endotoxin tolerance.
Collapse
|
18
|
A 2A adenosine receptor activation prevents neutrophil aging and promotes polarization from N1 towards N2 phenotype. Purinergic Signal 2022; 18:345-358. [PMID: 35838900 PMCID: PMC9391554 DOI: 10.1007/s11302-022-09884-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/02/2022] [Indexed: 01/17/2023] Open
Abstract
Extracellular adenosine is a biologically active signaling molecule that accumulates at sites of metabolic stress in sepsis. Extracellular adenosine has potent immunosuppressive effects by binding to and activating G protein-coupled A2A adenosine receptors (A2AARs) on the surface of neutrophils. A2AAR signaling reproduces many of the phenotypic changes in neutrophils that are characteristic of sepsis, including decreased degranulation, impaired chemotaxis, and diminished ability to ingest and kill bacteria. We hypothesized that A2AARs also suppress neutrophil aging, which precedes cell death, and N1 to N2 polarization. Using human neutrophils isolated from healthy subjects, we demonstrate that A2AAR stimulation slows neutrophil aging, suppresses cell death, and promotes the polarization of neutrophils from an N1 to N2 phenotype. Using genetic knockout and pharmacological blockade, we confirmed that A2AARs decrease neutrophil aging in murine sepsis induced by cecal ligation and puncture. A2AARs expression is increased in neutrophils from septic patients compared to healthy subject but A2AAR expression fails to correlate with aging or N1/N2 polarization. Our data reveals that A2AARs regulate neutrophil aging in healthy but not septic neutrophils.
Collapse
|
19
|
Usefulness of the delta neutrophil index in predicting surgery in patients with foot and ankle infection. PLoS One 2022; 17:e0272574. [PMID: 35926065 PMCID: PMC9352078 DOI: 10.1371/journal.pone.0272574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022] Open
Abstract
Background In foot and ankle infections, cases with apparent soft-tissue necrosis or purulent fluid collections definitely require surgical treatments. However, clinicians often have difficulty in determining whether to perform surgery in ambiguous cases without these findings. This study aimed to investigate the impact of the delta neutrophil index as a predictor of surgical treatment in patients with foot and ankle infections. Methods In total, 66 patients diagnosed with foot and ankle infections who underwent the delta neutrophil index test were retrospectively investigated. Medical records, including data on diabetes mellitus status, delta neutrophil index values, white blood cell count, polymorphonuclear leukocyte count, erythrocyte sedimentation rate, and C-reactive protein level, were retrospectively investigated. Logistic regression models were analyzed for the correlation between biomarkers, such as the delta neutrophil index and surgical treatment. The area under the curve was investigated to evaluate the cut-off value of the logistic model in predicting surgery. Results The relationship between the delta neutrophil index and surgical treatment was analyzed. The delta neutrophil index, adjusted for diabetes mellitus, was the best predictor of future surgical intervention. Based on the Youden index, the cutoff point (the equation’s adjusted by diabetes mellitus) for the prediction of surgical treatment was defined as a probability of 0.3, with sensitivity and specificity of 82.4% and 77.6%, respectively. Conclusions Based on the present study, the delta neutrophil index can help clinicians decide the appropriate surgical treatment for foot and ankle infections at the right time.
Collapse
|
20
|
Wen L, Marki A, Wang Z, Orecchioni M, Makings J, Billitti M, Wang E, Suthahar SSA, Kim K, Kiosses WB, Mikulski Z, Ley K. A humanized β 2 integrin knockin mouse reveals localized intra- and extravascular neutrophil integrin activation in vivo. Cell Rep 2022; 39:110876. [PMID: 35649374 PMCID: PMC10375464 DOI: 10.1016/j.celrep.2022.110876] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 03/28/2022] [Accepted: 05/04/2022] [Indexed: 11/24/2022] Open
Abstract
β2 integrins are leukocyte-specific adhesion molecules that are essential for leukocyte recruitment. The lack of tools for reporting β2 integrin activation in mice hindered the study of β2 integrin-related immune responses in vivo. Here, we generated a humanized β2 integrin knockin mouse strain by targeting the human β2 integrin coding sequence into the mouse Itgb2 locus to enable imaging of β2 integrin activation using the KIM127 (extension) and mAb24 (high-affinity) reporter antibodies. Using a CXCL1-induced acute inflammation model, we show the local dynamics of β2 integrin activation in arresting neutrophils in vivo in venules of the mouse cremaster muscle. Activated integrins are highly concentrated in a small area at the rear of arresting neutrophils in vivo. In a high-dose lipopolysaccharide model, we find that β2 integrins are activated in association with elevated neutrophil adhesion in lung and liver. Thus, these mice enable studies of β2 integrin activation in vivo.
Collapse
Affiliation(s)
- Lai Wen
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Alex Marki
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Zhihao Wang
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Marco Orecchioni
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Jeffrey Makings
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Monica Billitti
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Erpei Wang
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Sujit S A Suthahar
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Kenneth Kim
- Histopathology Core Facility, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - William B Kiosses
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Zbigniew Mikulski
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA; Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
21
|
Changes in immune function and immunomodulatory treatments of septic patients. Clin Immunol 2022; 239:109040. [DOI: 10.1016/j.clim.2022.109040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/23/2022] [Accepted: 05/07/2022] [Indexed: 12/25/2022]
|
22
|
Margotti W, Goldim MPDS, Machado RS, Bagio E, Dacoregio C, Bernades G, Lanzzarin E, Stork S, Cidreira T, Denicol TL, Joaquim L, Danielski LG, Metzker KLL, Bonfante S, Margotti E, Petronilho F. Oxidative stress in multiple organs after sepsis in elderly rats. Exp Gerontol 2022; 160:111705. [DOI: 10.1016/j.exger.2022.111705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/04/2022]
|
23
|
Yu X, Chen J, Tang H, Tu Q, Li Y, Yuan X, Zhang X, Cao J, Molloy DP, Yin Y, Chen D, Song Z, Xu P. Identifying Prokineticin2 as a Novel Immunomodulatory Factor in Diagnosis and Treatment of Sepsis. Crit Care Med 2022; 50:674-684. [PMID: 34582411 PMCID: PMC8923365 DOI: 10.1097/ccm.0000000000005335] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Sepsis remains a highly lethal disease, whereas the precise reasons for death remain poorly understood. Prokineticin2 is a secreted protein that regulates diverse biological processes. Whether prokineticin2 is beneficial or deleterious to sepsis and the underlying mechanisms remain unknown. DESIGN Prospective randomized animal investigation and in vitro studies. SETTING Research laboratory at a medical university hospital. SUBJECTS Prokineticin2 deficiency and wild-type C57BL/6 mice were used for in vivo studies; sepsis patients by Sepsis-3 definitions, patient controls, and healthy controls were used to obtain blood for in vitro studies. INTERVENTIONS Prokineticin2 concentrations were measured and analyzed in human septic patients, patient controls, and healthy individuals. The effects of prokineticin2 on sepsis-related survival, bacterial burden, organ injury, and inflammation were assessed in an animal model of cecal ligation and puncture-induced polymicrobial sepsis. In vitro cell models were also used to study the role of prokineticin2 on antibacterial response of macrophages. MEASUREMENTS AND MAIN RESULTS Prokineticin2 concentration is dramatically decreased in the patients with sepsis and septic shock compared with those of patient controls and healthy controls. Furthermore, the prokineticin2 concentration in these patients died of sepsis or septic shock is significantly lower than those survival patients with sepsis or septic shock, indicating the potential value of prokineticin2 in the diagnosis of sepsis and septic shock, as well as the potential value in predicting mortality in adult patients with sepsis and septic shock. In animal model, recombinant prokineticin2 administration protected against sepsis-related deaths in both heterozygous prokineticin2 deficient mice and wild-type mice and alleviated sepsis-induced multiple organ damage. In in vitro cell models, prokineticin2 enhanced the phagocytic and bactericidal functions of macrophage through signal transducers and activators of transcription 3 pathway which could be abolished by signal transducers and activators of transcription 3 inhibitors S3I-201. Depletion of macrophages reversed prokineticin2-mediated protection against polymicrobial sepsis. CONCLUSIONS This study elucidated a previously unrecognized role of prokineticin2 in clinical diagnosis and treatment of sepsis. The proof-of-concept study determined a central role of prokineticin2 in alleviating sepsis-induced death by regulation of macrophage function, which presents a new strategy for sepsis immunotherapy.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Jingyi Chen
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Hong Tang
- Department of Critical Care Medicine, Department of Surgical Intensive Care Unit, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qianqian Tu
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Yue Li
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xi Yuan
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Xuemei Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Ju Cao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - David Paul Molloy
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, ChongQing Medical University, Chongqing, China
| | - Yibing Yin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine, Chongqing Medical University, Chongqing, China
| | - Dapeng Chen
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Zhixin Song
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
| | - Pingyong Xu
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Chongqing, China
- Key Laboratory of RNA Biology, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
24
|
Peyneau M, Granger V, Wicky PH, Khelifi-Touhami D, Timsit JF, Lescure FX, Yazdanpanah Y, Tran-Dinh A, Montravers P, Monteiro RC, Chollet-Martin S, Hurtado-Nedelec M, de Chaisemartin L. Innate immune deficiencies are associated with severity and poor prognosis in patients with COVID-19. Sci Rep 2022; 12:638. [PMID: 35022495 PMCID: PMC8755788 DOI: 10.1038/s41598-021-04705-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/17/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 can cause acute respiratory distress syndrome, leading to death in many individuals. Evidence of a deleterious role of the innate immune system is accumulating, but the precise mechanisms involved remain unclear. In this study, we investigated the links between circulating innate phagocytes and severity in COVID-19 patients. We performed in-depth phenotyping of neutrophil and monocyte subpopulations and measured soluble activation markers in plasma. Additionally, anti-microbial functions (phagocytosis, oxidative burst, and NETosis) were evaluated on fresh cells from patients. Neutrophils and monocytes had a strikingly disturbed phenotype, and elevated concentrations of activation markers (calprotectin, myeloperoxidase, and neutrophil extracellular traps) were measured in plasma. Critical patients had increased CD13low immature neutrophils, LOX-1 + and CCR5 + immunosuppressive neutrophils, and HLA-DRlow downregulated monocytes. Markers of immature and immunosuppressive neutrophils were strongly associated with severity. Moreover, neutrophils and monocytes of critical patients had impaired antimicrobial functions, which correlated with organ dysfunction, severe infections, and mortality. Together, our results strongly argue in favor of a pivotal role of innate immunity in COVID-19 severe infections and pleads for targeted therapeutic options.
Collapse
Affiliation(s)
- Marine Peyneau
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Bichat Hospital, 46 Rue Henri Huchard, 75018, Paris, France.,INSERM UMR 996, "Inflammation, Microbiome and Immunosurveillance", Faculty of Pharmacy, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Vanessa Granger
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Bichat Hospital, 46 Rue Henri Huchard, 75018, Paris, France.,INSERM UMR 996, "Inflammation, Microbiome and Immunosurveillance", Faculty of Pharmacy, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Paul-Henri Wicky
- AP-HP, Bichat Hospital Medical and Infectious Diseases ICU (MI2), Paris, France.,IAME Université de Paris, Inserm U 1137, Paris, France
| | - Dounia Khelifi-Touhami
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Bichat Hospital, 46 Rue Henri Huchard, 75018, Paris, France
| | - Jean-François Timsit
- AP-HP, Bichat Hospital Medical and Infectious Diseases ICU (MI2), Paris, France.,IAME Université de Paris, Inserm U 1137, Paris, France
| | - François-Xavier Lescure
- IAME Université de Paris, Inserm U 1137, Paris, France.,Infectious Diseases Department, AP-HP, Hôpital Bichat, Paris, France
| | - Yazdan Yazdanpanah
- IAME Université de Paris, Inserm U 1137, Paris, France.,Infectious Diseases Department, AP-HP, Hôpital Bichat, Paris, France
| | - Alexy Tran-Dinh
- Département d'Anesthésie-Réanimation, DMU PARABOL, Université de Paris, AP-HP, Bichat Hospital, Paris, France.,INSERM UMR 1152 - ANR10-LABX-17, Paris, France
| | - Philippe Montravers
- Département d'Anesthésie-Réanimation, DMU PARABOL, Université de Paris, AP-HP, Bichat Hospital, Paris, France.,INSERM UMR 1152 - ANR10-LABX-17, Paris, France
| | - Renato C Monteiro
- Center for Research On Inflammation (CRI), Inflamex Laboratory of Excellence, U1149, CNRS ERL8252, Paris, France.,Immunological Dysfunction Laboratory, APHP, Bichat Hospital, Paris, France
| | - Sylvie Chollet-Martin
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Bichat Hospital, 46 Rue Henri Huchard, 75018, Paris, France.,INSERM UMR 996, "Inflammation, Microbiome and Immunosurveillance", Faculty of Pharmacy, Université Paris-Saclay, 92290, Châtenay-Malabry, France
| | - Margarita Hurtado-Nedelec
- Center for Research On Inflammation (CRI), Inflamex Laboratory of Excellence, U1149, CNRS ERL8252, Paris, France.,Immunological Dysfunction Laboratory, APHP, Bichat Hospital, Paris, France
| | - Luc de Chaisemartin
- Autoimmunity and Hypersensitivity Laboratory, AP-HP, Bichat Hospital, 46 Rue Henri Huchard, 75018, Paris, France. .,INSERM UMR 996, "Inflammation, Microbiome and Immunosurveillance", Faculty of Pharmacy, Université Paris-Saclay, 92290, Châtenay-Malabry, France.
| |
Collapse
|
25
|
Lazzaro A, De Girolamo G, Filippi V, Innocenti GP, Santinelli L, Ceccarelli G, Trecarichi EM, Torti C, Mastroianni CM, d’Ettorre G, Russo A. The Interplay between Host Defense, Infection, and Clinical Status in Septic Patients: A Narrative Review. Int J Mol Sci 2022; 23:ijms23020803. [PMID: 35054993 PMCID: PMC8776148 DOI: 10.3390/ijms23020803] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 01/08/2023] Open
Abstract
Sepsis is a life-threatening condition that arises when the body's response to an infection injures its own tissues and organs. Despite significant morbidity and mortality throughout the world, its pathogenesis and mechanisms are not clearly understood. In this narrative review, we aimed to summarize the recent developments in our understanding of the hallmarks of sepsis pathogenesis (immune and adaptive immune response, the complement system, the endothelial disfunction, and autophagy) and highlight novel laboratory diagnostic approaches. Clinical management is also discussed with pivotal consideration for antimicrobic therapy management in particular settings, such as intensive care unit, altered renal function, obesity, and burn patients.
Collapse
Affiliation(s)
- Alessandro Lazzaro
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Gabriella De Girolamo
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Valeria Filippi
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Giuseppe Pietro Innocenti
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Letizia Santinelli
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Enrico Maria Trecarichi
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (E.M.T.); (C.T.)
| | - Carlo Torti
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (E.M.T.); (C.T.)
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, “Sapienza” University of Rome, 00161 Rome, Italy; (A.L.); (G.D.G.); (V.F.); (G.P.I.); (L.S.); (G.C.); (C.M.M.); (G.d.)
| | - Alessandro Russo
- Infectious and Tropical Disease Unit, Department of Medical and Surgical Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (E.M.T.); (C.T.)
- Correspondence:
| |
Collapse
|
26
|
Stratmann AEP, Wohlgemuth L, Erber ME, Bernhard S, Hug S, Fauler M, Vidoni L, Mohamed AOK, Thomaß BD, Münnich F, Stukan L, Föhr KJ, Mannes M, Huber-Lang MS, Messerer DAC. Simultaneous Measurement of Changes in Neutrophil Granulocyte Membrane Potential, Intracellular pH, and Cell Size by Multiparametric Flow Cytometry. Biomedicines 2021; 9:1504. [PMID: 34829733 PMCID: PMC8614908 DOI: 10.3390/biomedicines9111504] [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: 09/22/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 01/10/2023] Open
Abstract
Neutrophils provide rapid and efficient defense mechanisms against invading pathogens. Upon stimulation with proinflammatory mediators, including complement factors and bacterial peptides, neutrophils respond with changes in their membrane potential, intracellular pH, and cellular size. This study provides an approach to quantify these important changes simultaneously using multiparametric flow cytometry, thereby revealing a typical sequence of neutrophil activation consisting of depolarization, alkalization, and increase in cellular size. Additionally, the time resolution of the flow cytometric measurement is improved in order to allow changes that occur within seconds to be monitored, and thus to enhance the kinetic analysis of the neutrophil response. The method is appropriate for the reliable semiquantitative detection of small variations with respect to an increase, no change, and decrease in those parameters as demonstrated by the screening of various proinflammatory mediators. As a translational outlook, the findings are put into context in inflammatory conditions in vitro as well as in a clinically relevant whole blood model of endotoxemia. Taken together, the multiparametric analysis of neutrophil responsiveness regarding depolarization, alkalization, and changes in cellular size may contribute to a better understanding of neutrophils in health and disease, thus potentially yielding innovative mechanistic insights and possible novel diagnostic and/or prognostic approaches.
Collapse
Affiliation(s)
| | - Lisa Wohlgemuth
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Maike Elisabeth Erber
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Stefan Bernhard
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Stefan Hug
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Michael Fauler
- Institute of General Physiology, Ulm University, 89081 Ulm, Germany
| | - Laura Vidoni
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Adam Omar Khalaf Mohamed
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Bertram Dietrich Thomaß
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Frederik Münnich
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Laura Stukan
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| | - Karl Josef Föhr
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| | - Marco Mannes
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - Markus Stefan Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
| | - David Alexander Christian Messerer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, 89081 Ulm, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
27
|
Kong T, Lee HS, Jeon SY, You JS, Lee JW, Chung HS, Chung SP. Delta neutrophil index and shock index can stratify risk for the requirement for massive transfusion in patients with primary postpartum hemorrhage in the emergency department. PLoS One 2021; 16:e0258619. [PMID: 34653202 PMCID: PMC8519472 DOI: 10.1371/journal.pone.0258619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 10/03/2021] [Indexed: 11/30/2022] Open
Abstract
Background Postpartum hemorrhage (PPH) constitutes a major risk for maternal mortality and morbidity. Unfortunately, the severity of PPH can be underestimated because it is difficult to accurately measure blood loss by visual estimation. The delta neutrophil index (DNI), which reflects circulating immature granulocytes, is automatically calculated in hematological analyzers. We evaluated the significance of the DNI in predicting hemorrhage severity based on the requirement for massive transfusion (MT) in patients with PPH. Methods We retrospectively analyzed data from a prospective registry to evaluate the association between the DNI and MT. Moreover, we assessed the predictive ability of the combination of DNI and shock index (SI) for the requirement for MT. MT was defined as a transfusion of ≥10 units of red blood cells within 24 h of PPH. In total, 278 patients were enrolled in this study and 60 required MT. Results Multivariable logistic regression revealed that the DNI and SI were independent predictors of MT. The optimal cut-off values of ≥3.3% and ≥1.0 for the DNI and SI, respectively, were significantly associated with an increased risk of MT (DNI: positive likelihood ratio [PLR] 3.54, 95% confidence interval [CI] 2.5–5.1 and negative likelihood ratio [NLR] 0.48, 95% CI 0.4–0.7; SI: PLR 3.21, 95% CI 2.4–4.2 and NLR 0.31, 95% CI 0.19–0.49). The optimal cut-off point for predicted probability was calculated for combining the DNI value and SI value with the equation derived from logistic regression analysis. Compared with DNI or SI alone, the combination of DNI and SI significantly improved the specificity, accuracy, and positive likelihood ratio of the MT risk. Conclusion The DNI and SI can be routinely and easily measured in the ED without additional costs or time and can therefore, be considered suitable parameters for the early risk stratification of patients with primary PPH.
Collapse
Affiliation(s)
- Taeyoung Kong
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sun Lee
- Department of Research Affairs, Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Young Jeon
- Department of Research Affairs, Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Sung You
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| | - Jong Wook Lee
- Department of Laboratory Medicine, Konyang University Hospital, Daejon, Republic of Korea
| | - Hyun Soo Chung
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung Phil Chung
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
28
|
Lin R, Li L. Innate Neutrophil Memory Dynamics in Disease Pathogenesis. Handb Exp Pharmacol 2021; 276:43-64. [PMID: 34486096 DOI: 10.1007/164_2021_538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neutrophils, the most abundant leukocytes in circulation and the first responders to infection and inflammation, closely modulate both acute and chronic inflammatory processes. Resting neutrophils constantly patrol vasculature and migrate to tissues when challenges occur. When infection and/or inflammation recede, tissue neutrophils will be subsequently cleaned up by macrophages which collectively contribute to the resolution of inflammation. While most studies focus on the anti-microbial function of neutrophils including phagocytosis, degranulation, and neutrophil extracellular traps (NETs) formation, recent research highlighted additional contributions of neutrophils beyond simply controlling infectious agents. Neutrophils with resolving characteristics may alter the activities of neighboring cells and facilitate inflammation resolution, modulate long-term macrophage and adaptive immune responses, therefore having important impacts on host pathophysiology. The focus of this chapter is to provide an updated assessment of recent progress in the emerging field of neutrophil programming and memory in the context of both acute and chronic diseases.
Collapse
Affiliation(s)
- RuiCi Lin
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, VA, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Liwu Li
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, VA, USA. .,Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
29
|
Khodakhah F, Tahamtan A, Marzban M, Shadab A, Tavakoli-Yaraki M, Hashemi SM, Mokhatri-Azad T, Nakstad B, Salimi V. Hyperglycemia results in decreased immune cell infiltration and increased viral load in the lung in a mouse model of RSV infection. Cytokine 2021; 143:155539. [PMID: 33893002 DOI: 10.1016/j.cyto.2021.155539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Respiratory Syncytial virus (RSV) infection is a feared disease in vulnerable populations with impaired immune responses. There is currently no vaccine against RSV and young children along with elderly people are at increased risk of severe or sometimes life-threatening RSV infection. Hyperglycemia with immunomodulatory patterns can impact on infectious disease outcomes and immune system responses in diabetic patients. Even though research continues to uncover the complex mechanisms underlying RSV immunopathogenesis and diabetes mellitus disease separately, limited information is available about interaction between these two phenomena. Here, we evaluated the influence of hyperglycemia as the hallmark of diabetes mellitus disease on the pathogenesis and immunopathogenesis of RSV in a mouse model. In this experiment, hyperglycemia was induced by intraperitoneal injection of Streptozotocin (STZ), and after diabetes confirmation, mice were infected with RSV-A2, and the immune responses were followed for 5 days until the mice were sacrificed. Analyses on airway immune cell influx, T-Lymphocyte subtypes, cytokines secretion, lung histopathology, and viral load were conducted. Our results showed that hyperglycemia resulted in reduced lung immune cells infiltration totally and it was associated with decreased pathological damage of the lung. Following RSV infection in hyperglycemic mice, the ratio of CD4/CD8 T-Lymphocytes due to CD8+ depletion, increased. Furthermore, the level of IFN-γ and IL-17A cytokines decreased, whereas IL-10 showed an upward trend and the viral load increased in hyperglycemic mice compared with normoglycemic mice. In conclusion, these findings indicate that hyperglycemia can ameliorate and downregulate RSV-induced inflammatory and antiviral responses, and result in increment of viral load.
Collapse
Affiliation(s)
- Farshad Khodakhah
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Tahamtan
- Infectious Diseases Research Centre, Golestan University of Medical Sciences, Gorgan, Iran; Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mona Marzban
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Shadab
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Talat Mokhatri-Azad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Britt Nakstad
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Paediatric and Adolescent Health, University of Botswana, Gaborone, Botswana
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Engelmann C, Clària J, Szabo G, Bosch J, Bernardi M. Pathophysiology of decompensated cirrhosis: Portal hypertension, circulatory dysfunction, inflammation, metabolism and mitochondrial dysfunction. J Hepatol 2021; 75 Suppl 1:S49-S66. [PMID: 34039492 PMCID: PMC9272511 DOI: 10.1016/j.jhep.2021.01.002] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/31/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
Patients with acutely decompensated cirrhosis have a dismal prognosis and frequently progress to acute-on-chronic liver failure, which is characterised by hepatic and extrahepatic organ failure(s). The pathomechanisms involved in decompensation and disease progression are still not well understood, and as specific disease-modifying treatments do not exist, research to identify novel therapeutic targets is of the utmost importance. This review amalgamates the latest knowledge on disease mechanisms that lead to tissue injury and extrahepatic organ failure - such as systemic inflammation, mitochondrial dysfunction, oxidative stress and metabolic changes - and marries these with the classical paradigms of acute decompensation to form a single paradigm. With this detailed breakdown of pathomechanisms, we identify areas for future research. Novel disease-modifying strategies that break the vicious cycle are urgently required to improve patient outcomes.
Collapse
Affiliation(s)
- Cornelius Engelmann
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany; Institute for Liver and Digestive Health, University College London, London, United Kingdom; Section Hepatology, Clinic for Gastroenterology and Rheumatology, University Hospital Leipzig, Leipzig, Germany; Berlin Institute of Health (BIH), Berlin, Germany.
| | - Joan Clària
- European Foundation for the Study of Chronic Liver Failure (EF-Clif) and Grifols Chair, Barcelona, Spain,Biochemistry and Molecular Genetics Service, Hospital ClínicIDIBAPS and CIBERehd, Spain,Department of Biomedical Sciences, University of Barcelona, Barcelona, Spain
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Jaume Bosch
- IDIBAPS and CIBERehd, University of Barcelona, Barcelona, Spain,Department for Biomedical Research (DBMR), Bern University, Bern, Switzerland
| | - Mauro Bernardi
- Department of Medical and Surgical Sciences; Alma Mater Studiorum – University of Bologna; Italy
| |
Collapse
|
31
|
Endothelial Dysfunction and Neutrophil Degranulation as Central Events in Sepsis Physiopathology. Int J Mol Sci 2021; 22:ijms22126272. [PMID: 34200950 PMCID: PMC8230689 DOI: 10.3390/ijms22126272] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a major health problem worldwide. It is a time-dependent disease, with a high rate of morbidity and mortality. In this sense, an early diagnosis is essential to reduce these rates. The progressive increase of both the incidence and prevalence of sepsis has translated into a significant socioeconomic burden for health systems. Currently, it is the leading cause of noncoronary mortality worldwide and represents one of the most prevalent pathologies both in hospital emergency services and in intensive care units. In this article, we review the role of both endothelial dysfunction and neutrophil dysregulation in the physiopathology of this disease. The lack of a key symptom in sepsis makes it difficult to obtain a quick and accurate diagnosis of this condition. Thus, it is essential to have fast and reliable diagnostic tools. In this sense, the use of biomarkers can be a very important alternative when it comes to achieving these goals. Both new biomarkers and treatments related to endothelial dysfunction and neutrophil dysregulation deserve to be further investigated in order to open new venues for the diagnosis, treatment and prognosis of sepsis.
Collapse
|
32
|
Heterogeneity of neutrophils in arterial hypertension. Exp Cell Res 2021; 402:112577. [PMID: 33811902 DOI: 10.1016/j.yexcr.2021.112577] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 01/21/2023]
Abstract
Cellular heterogeneity and diversity are recognized to contribute to the functions of neutrophils under homeostatic and pathological conditions. We previously suggested that the chronic inflammatory responses associated with hypertension (HTN) are related to the participation of different subpopulations of neutrophils. Two populations of neutrophils can be obtained by density gradient centrifugation: normal-density neutrophils (NDN) and low-density neutrophils (LDN). However, the lack of standardized functional protocols has limited phenotypic characterization and functional comparisons of LDN and NDN. Based on their capability to incorporate Na+, maturity and activation stage, we characterized NDN and LDN in blood samples from ten patients with HTN and ten healthy individuals (HI) using flow cytometry. We compared the levels of reactive oxygen species (ROS), generation of neutrophil extracellular traps (NETs) and levels of apoptosis in NDN and LDN. In general, the NDN and LDN subpopulations from patients with HTN exhibited higher levels of sodium influx and ROS, and lower levels of apoptosis than the corresponding NDN and LDN subsets from HI. Transmission electron microscopy revealed NDN and LDN from patients with HTN exhibited alterations to mitochondrial morphology and fewer cytoplasmic granules than the corresponding HI subpopulations. Our results indicate both the NDN and LDN subpopulations enhance the effects of inflammation that contribute to the pathophysiology of HTN. Further detailed studies are required to characterize the events during ontogeny of the myeloid lineage that result in the diverse phenotypic characteristics of each subpopulation of LDN and NDN.
Collapse
|
33
|
Ciupe SM, Boribong BP, Kadelka S, Jones CN. Bistable Mathematical Model of Neutrophil Migratory Patterns After LPS-Induced Epigenetic Reprogramming. Front Genet 2021; 12:633963. [PMID: 33708241 PMCID: PMC7940759 DOI: 10.3389/fgene.2021.633963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/27/2021] [Indexed: 11/17/2022] Open
Abstract
The highly controlled migration of neutrophils toward the site of an infection can be altered when they are trained with lipopolysaccharides (LPS), with high dose LPS enhancing neutrophil migratory pattern toward the bacterial derived source signal and super-low dose LPS inducing either migration toward an intermediary signal or dysregulation and oscillatory movement. Empirical studies that use microfluidic chemotaxis-chip devices with two opposing chemoattractants showed differential neutrophil migration after challenge with different LPS doses. The epigenetic alterations responsible for changes in neutrophil migratory behavior are unknown. We developed two mathematical models that evaluate the mechanistic interactions responsible for neutrophil migratory decision-making when exposed to competing chemoattractants and challenged with LPS. The first model, which considers the interactions between the receptor densities of two competing chemoattractants, their kinases, and LPS, displayed bistability between high and low ratios of primary to intermediary chemoattractant receptor densities. In particular, at equilibrium, we observe equal receptor densities for low LPS (< 15ng/mL); and dominance of receptors for the primary chemoattractant for high LPS (> 15ng/mL). The second model, which included additional interactions with an extracellular signal-regulated kinase in both phosphorylated and non-phosphorylated forms, has an additional dynamic outcome, oscillatory dynamics for both receptors, as seen in the data. In particular, it found equal receptor densities in the absence of oscillation for super-low and high LPS challenge (< 0.4 and 1.1 376 ng/mL). Predicting the mechanisms and the type of external LPS challenge responsible for neutrophils migration toward pro-inflammatory chemoattractants, migration toward pro-tolerant chemoattractants, or oscillatory movement is necessary knowledge in designing interventions against immune diseases, such as sepsis.
Collapse
Affiliation(s)
- Stanca M. Ciupe
- Department of Mathematics, Virginia Tech, Blacksburg, VA, United States
| | - Brittany P. Boribong
- Division of Pediatric Pulmonology, Massachusetts General Hospital, Boston, MA, United States
| | - Sarah Kadelka
- Department of Environmental Systems Science, ETH Zurich, Zurich, Switzerland
| | - Caroline N. Jones
- Department of Bioengineering, University of Texas, Dallas, TX, United States
| |
Collapse
|
34
|
Souto FO, Castanheira FVS, Trevelin SC, Lima BHF, Cebinelli GCM, Turato WM, Auxiliadora-Martins M, Basile-Filho A, Alves-Filho JC, Cunha FQ. Liver X Receptor Activation Impairs Neutrophil Functions and Aggravates Sepsis. J Infect Dis 2021; 221:1542-1553. [PMID: 31783409 DOI: 10.1093/infdis/jiz635] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/27/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Liver X receptors (LXRs) are nuclear receptors activated by oxidized lipids and were previously implicated in several metabolic development and inflammatory disorders. Although neutrophils express both LXR-α and LXR-β, the consequences of their activation, particularly during sepsis, remain unknown. METHODS We used the model of cecal ligation and puncture (CLP) to investigate the role of LXR activation during sepsis. RESULTS In this study, we verified that LXR activation reduces neutrophil chemotactic and killing abilities in vitro. Mice treated with LXR agonists showed higher sepsis-induced mortality, which could be associated with reduced neutrophil infiltration at the infectious foci, increased bacteremia, systemic inflammatory response, and multiorgan failure. In contrast, septic mice treated with LXR antagonist showed increased number of neutrophils in the peritoneal cavity, reduced bacterial load, and multiorgan dysfunction. More important, neutrophils from septic patients showed increased ABCA1 messenger ribonucleic acid levels (a marker of LXR activation) and impaired chemotactic response toward CXCL8 compared with cells from healthy individuals. CONCLUSIONS Therefore, our findings suggest that LXR activation impairs neutrophil functions, which might contribute to poor sepsis outcome.
Collapse
Affiliation(s)
- Fabrício O Souto
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife, Brazil
| | - Fernanda V S Castanheira
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center of Research of Inflammatory Diseases, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Silvia C Trevelin
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,King's College London, British Heart Foundation Centre, School of Cardiovascular Medicine and Sciences, London, United Kingdom
| | - Braulio H F Lima
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Walter M Turato
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Maria Auxiliadora-Martins
- Department of Pharmacology, Surgery and Anatomy, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Anibal Basile-Filho
- Department of Pharmacology, Surgery and Anatomy, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Jose Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center of Research of Inflammatory Diseases, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Q Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Center of Research of Inflammatory Diseases, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
35
|
Cui J, Oehrl S, Ahmad F, Brenner T, Uhle F, Nusshag C, Rupp C, Funck F, Meisel S, Weigand MA, Morath C, Schäkel K. Detection of In Vivo Inflammasome Activation for Predicting Sepsis Mortality. Front Immunol 2021; 11:613745. [PMID: 33613537 PMCID: PMC7889521 DOI: 10.3389/fimmu.2020.613745] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 12/11/2020] [Indexed: 12/17/2022] Open
Abstract
Sepsis is a severe life-threatening syndrome caused by dysregulated host responses to infection. Biomarkers that allow for monitoring the patient's immune status are needed. Recently, a flow cytometry-based detection of in vivo inflammasome activation by formation of cytoplasmic aggregates of ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain) has been proposed. Here we report on the frequency of ASC-speck+ leukocytes correlating with the survival of sepsis. 25 patients with sepsis were sampled consecutively for 7 days. Blood, serum samples and patient data were collected according to the guidelines of the PredARRT-Sep-Trial. Flow cytometric analysis was performed on fresh whole blood samples to investigate the formation of ASC-specks in leukocyte subsets. Serum samples were analyzed for production of IL-1ß, IL-18 and additional inflammatory markers. ASC-speck formation was found to be increased in leukocytes from sepsis patients compared to healthy donor controls. The absolute number of ASC-speck+ neutrophils peaked on day 1. For monocytes, the highest percentage and maximum absolute number of ASC-speck+ cells were detected on day 6 and day 7. Inflammatory cytokines were elevated on day 1 and declined thereafter, with exception of IL-18. Survival analysis showed that patients with lower absolute numbers of ASC-speck+ monocytes (<1,650 cells/ml) on day 6 had a lower probability to survive, with a hazard ratio (HR) of 10.178. Thus, the frequency of ASC-speck+ monocytes on day 6 after onset of sepsis may serve to identify patients at risk of death from sepsis.
Collapse
Affiliation(s)
- Jing Cui
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany.,Department of Dermatology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Stephanie Oehrl
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Fareed Ahmad
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care, University Hospital Essen, Essen, Germany.,Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Florian Uhle
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christoph Rupp
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Funck
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan Meisel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christian Morath
- Department of Nephrology, Heidelberg University Hospital, Heidelberg, Germany
| | - Knut Schäkel
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
36
|
Inhibition of Aerobic Glycolysis Promotes Neutrophil to Influx to the Infectious Site Via CXCR2 in Sepsis. Shock 2021; 53:114-123. [PMID: 30829852 DOI: 10.1097/shk.0000000000001334] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent evidences suggest that metabolic reprogramming plays an important role in the regulation of innate inflammatory response; however, the specific mechanism is unclear. In this study, we found that glycolytic inhibitor 2-deoxyglucose (2-DG) significantly improved the survival rate in cecal ligation and puncture (CLP)-induced septic mice. 2-DG-treated mice developed increased neutrophil migration to the infectious site and more efficient bacterial clearance than untreated mice. 2-DG reversed the down-regulation of chemokine receptor 2 (CXCR2) and the impaired chemotaxis induced by CLP in mice or lipopolysaccharides (LPS) in human neutrophils. Furthermore, 2-DG reversed the down-regulation of CXCR2 in neutrophils by decreasing the expression of G protein-coupled receptor kinase-2 (GRK2), a serin-threonine protein kinase that mediated the internalization of chemokine receptors, which was induced via the inhibition of extracellular regulated protein kinases (ERK) phosphorylation and the promotion of P38 phosphorylation. Finally, SB225002, a CXCR2 antagonist, partially blocked the protective effects of 2-DG in sepsis. Together, we found a novel mechanism for the migration of neutrophils regulated by metabolism and suggested that aerobic glycolysis might be a potential target of intervention in sepsis.
Collapse
|
37
|
Mikhalchik EV, Borodina IV, Vlasova IV, Vakhrusheva TV, Gorbunov NP, Panasenko OM, Titkova SM, Anurov MV, Ivakhov GB, Ermakov IV, Teplyshev AV, Klinov DV. [Biomarkers of system inflammation in local and diffuse peritonitis]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 66:411-418. [PMID: 33140736 DOI: 10.18097/pbmc20206605411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In cases of any acute surgical abdominal disease the progression of purulent inflammation can lead to local or diffuse peritonitis. The indicators of the degree and specificity of the inflammatory response in blood such as cytokine concentration, neutrophil activity, plasma antioxidant capacity (thiols concentration) could be considered as potential predictors of complications. The luminol-dependent chemiluminescence (CL) response of blood activated by the phorbol ester (PMA), and the concentration of cytokines IL-6, IL-8, IL-10, myeloperoxidase (MPO) and thiols in plasma were measured in patients with uncomplicated condition (group 1, n=8), local peritonitis (group 2, n=9) or diffuse peritonitis (group 3, n=9) at admission to surgery (before surgical operation, b/o), immediately after surgical operation (a/o) and a day after surgery (1 day) as well as in healthy volunteers (norm, n=12). In all time-points the cytokines and MPO concentrations measured by ELISA, in group 3 were higher than in healthy volunteers and in patients in groups 1 and 2. Blood CL demonstrated a more than 5-fold increase above the normal values in all patients, and was also higher in group 2 as compared to group 1 (b/o and a/o). Patients in group 3 had shown both maximum and minimum of CL values, which could be a consequence of neutrophil priming or exhaustion ("immune paralysis"), respectively. The same patients' plasma exhibited low thiol concentration (≤30% vs normal values). In patients with fatal outcomes (group 3, n=2) within a day after surgery, either a decrease of the CL to zero values concurrently with elevated IL-8 and IL-6 concentrations and low thiol levels was observed, or CL exceeded normal values more than 20 times with concurrent complete exhaustion of the plasma thiol pool. No clear dependency between the plasma parameters and neutrophil activity was found. Hence a parameter set for prognosis and/or early diagnosis of infectious complications in acute abdominal pathology should include different biomarkers of the inflammatory response: cytokine profile (IL-6, IL-8, IL-10), MPO and neutrophil activity, antioxidant plasma capacity (e.g., total thiols concentration).
Collapse
Affiliation(s)
- E V Mikhalchik
- Federal Research Clinical Center of Physical Chemical Medicine, FMBA of Russia, Moscow, Russia
| | - I V Borodina
- Federal Research Clinical Center of Physical Chemical Medicine, FMBA of Russia, Moscow, Russia
| | - I V Vlasova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Institute for Regenerative Medicine, Moscow, Russia
| | - T V Vakhrusheva
- Federal Research Clinical Center of Physical Chemical Medicine, FMBA of Russia, Moscow, Russia
| | - N P Gorbunov
- Federal Research Clinical Center of Physical Chemical Medicine, FMBA of Russia, Moscow, Russia
| | - O M Panasenko
- Federal Research Clinical Center of Physical Chemical Medicine, FMBA of Russia, Moscow, Russia
| | - S M Titkova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - M V Anurov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - G B Ivakhov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - I V Ermakov
- Pirogov Russian National Research Medical University, Moscow, Russia
| | | | - D V Klinov
- Federal Research Clinical Center of Physical Chemical Medicine, FMBA of Russia, Moscow, Russia
| |
Collapse
|
38
|
Crosstalk Between Lung and Extrapulmonary Organs in Infection and Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:333-350. [PMID: 33788201 DOI: 10.1007/978-3-030-63046-1_18] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Acute and chronic lung inflammation is a risk factor for various diseases involving lungs and extrapulmonary organs. Intercellular and interorgan networks, including crosstalk between lung and brain, intestine, heart, liver, and kidney, coordinate host immunity against infection, protect tissue, and maintain homeostasis. However, this interaction may be counterproductive and cause acute or chronic comorbidities due to dysregulated inflammation in the lung. In this chapter, we review the relationship of the lung with other key organs during normal cell processes and disease development. We focus on how pneumonia may lead to a systemic pathophysiological response to acute lung injury and chronic lung disease through organ interactions, which can facilitate the development of undesirable and even deleterious extrapulmonary sequelae.
Collapse
|
39
|
Beunders R, Schütz MJ, van Groenendael R, Leijte GP, Kox M, van Eijk LT, Pickkers P. Endotoxemia-Induced Release of Pro-inflammatory Mediators Are Associated With Increased Glomerular Filtration Rate in Humans in vivo. Front Med (Lausanne) 2020; 7:559671. [PMID: 33251227 PMCID: PMC7674961 DOI: 10.3389/fmed.2020.559671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/08/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction: Sepsis is the most prevalent cause of Acute Kidney Injury (AKI). Conversely, in some septic patients the glomerular filtration rate (GFR) is augmented. The role of the inflammatory response and blood pressure to induce this increased GFR is unknown. Herein, we relate inflammatory mediators and blood pressure to the iohexol clearance-derived “true” GFR and kidney injury markers during systemic inflammation in healthy volunteers. Methods: Twelve healthy subjects underwent experimental endotoxemia (i.v. administration of 2 ng/kg Escherichia coli-derived lipopolysaccharide, LPS). As a gold-standard to determine the GFR, iohexol plasma clearance (GFRiohexol) was calculated during a 6-h period on the day before (baseline) as well as 2 and 24 h after LPS administration. Intra-arterial blood pressure was recorded continuously using a radial artery catheter. Circulating inflammatory mediators and urinary excretion of kidney injury markers were serially measured. Results: Experimental endotoxemia profoundly increased plasma concentrations of inflammatory mediators, including [mean ± SD or median [IQR] peak values (pg/mL) of tumor necrosis factor (TNF)-α: 92 ± 40, interleukin (IL)-6: 1,246 ± 605, IL-8: 374 ± 120, IL-10: 222 ± 119, IL-1 receptor antagonist (RA): 8,955 ± 2,429, macrophage chemoattractant protein (MCP)-1: 2,885 [2,706 – 3,765], vascular adhesion molecule (VCAM)-1: 296,105 ± 34,822, intercellular adhesion molecule (ICAM)-1: 25,0170 ± 41,764]. Mean arterial pressure decreased with 13 ± 11 mmHg (p < 0.0001). No significant increase in the urinary excretion of tubular injury markers was observed following LPS administration. GFRiohexol increased from 97 ± 6 at baseline to 118 ± 10 mL/min/1.73m2 (p < 0.0001) post-LPS administration and returned to baseline levels at 24 h post-LPS (99 ± 9 mL/min/1.73m2). Peak plasma concentrations of IL-6 (R2 = 0.66, p = 0.001) and IL-8 (R2 = 0.51, p = 0.009), MCP-1 (R2 = 0.38, p = 0.03) and VCAM-1 levels (R2 = 0.37, p = 0.04) correlated with the increase in GFRiohexol, whereas a trend was observed for TNF-α (R2 = 0.33, p = 0.0509) and IL-1RA (R2 = 0.28, p = 0.08). None of the kidney injury markers or changes in blood pressure were associated with GFRiohexol. In multiple linear regression analysis, both peak IL-6 (p = 0.002) and IL-8 (p = 0.01) concentrations remained significantly correlated with GFRiohexol, without collinearity. Discussion: Concentrations of pro-inflammatory cytokines, but not blood pressure, are correlated with the endotoxemia-induced increase in GFR in healthy volunteers. These findings may indicate that inflammatory mediators orchestrate the augmented GFR observed in a subgroup of sepsis patients.
Collapse
Affiliation(s)
- Remi Beunders
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Maren J Schütz
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Roger van Groenendael
- Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Guus P Leijte
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Lucas T van Eijk
- Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Anesthesiology, Pain and Palliative Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands.,Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
40
|
Resende CB, Borges I, Gonçalves WA, Carneiro R, Rezende BM, Pinho V, Nobre V, Teixeira MM. Neutrophil activity in sepsis: a systematic review. ACTA ACUST UNITED AC 2020; 53:e7851. [PMID: 33111742 PMCID: PMC7584151 DOI: 10.1590/1414-431x20207851] [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: 07/02/2020] [Accepted: 08/04/2020] [Indexed: 12/04/2022]
Abstract
The neutrophil is an important cell in host defense against infections, acting as the first line of microorganism control. However, this cell exhibits dysregulated activity in sepsis and may contribute to the pathogenesis of the disease. This systematic review aimed to highlight the major scientific findings regarding neutrophil activity in sepsis reported in clinical and experimental research published in the last 10 years. The search was conducted in the Virtual Health Library of PAHO-WHO (BVS) and PubMed databases, and articles published between January 2007 and May 2017 in Portuguese, English, and Spanish were eligible. Article selection was carried out independently by two reviewers (CB and IB). A total of 233 articles were found, of which 87 were identified on PubMed and 146 on BVS. Eighty-two articles were duplicates. Of the remaining 151 articles, 19 met the inclusion criteria after title, abstract, and full-text analysis. Overall, research in clinical samples and animal models of sepsis showed reduced capacity of neutrophils to migrate and delayed apoptosis, but there was no consensus on the phagocytic activity of neutrophils in sepsis. Molecules, such as pentraxin 3 (PTX3), have been analyzed as potential diagnostic markers in sepsis but the diversity of soluble molecules detected in blood samples of sepsis patients did not enable further understanding of the correlation of these circulating molecules with neutrophil activity during sepsis. Optimal understanding of the function of neutrophils in sepsis remains a challenge that, if overcome, would eventually allow targeted therapeutic interventions in patients affected by this severe syndrome.
Collapse
Affiliation(s)
- C B Resende
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.,Núcleo Interdisciplinar de Investigação em Medicina Intensiva, Belo Horizonte, MG, Brasil
| | - I Borges
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.,Núcleo Interdisciplinar de Investigação em Medicina Intensiva, Belo Horizonte, MG, Brasil.,Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - W A Gonçalves
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - R Carneiro
- Hospital das Clínicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.,Núcleo Interdisciplinar de Investigação em Medicina Intensiva, Belo Horizonte, MG, Brasil
| | - B M Rezende
- Departamento de Enfermagem Básica, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - V Pinho
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| | - V Nobre
- Núcleo Interdisciplinar de Investigação em Medicina Intensiva, Belo Horizonte, MG, Brasil.,Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - M M Teixeira
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil
| |
Collapse
|
41
|
Fine N, Tasevski N, McCulloch CA, Tenenbaum HC, Glogauer M. The Neutrophil: Constant Defender and First Responder. Front Immunol 2020; 11:571085. [PMID: 33072112 PMCID: PMC7541934 DOI: 10.3389/fimmu.2020.571085] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
The role of polymorphonuclear neutrophils (PMNs) in biology is often recognized during pathogenesis associated with PMN hyper- or hypo-functionality in various disease states. However, in the vast majority of cases, PMNs contribute to resilience and tissue homeostasis, with continuous PMN-mediated actions required for the maintenance of health, particularly in mucosal tissues. PMNs are extraordinarily well-adapted to respond to and diminish the damaging effects of a vast repertoire of infectious agents and injurious processes that are encountered throughout life. The commensal biofilm, a symbiotic polymicrobial ecosystem that lines the mucosal surfaces, is the first line of defense against pathogenic strains that might otherwise dominate, and is therefore of critical importance for health. PMNs regularly interact with the commensal flora at the mucosal tissues in health and limit their growth without developing an overt inflammatory reaction to them. These PMNs exhibit what is called a para-inflammatory phenotype, and have reduced inflammatory output. When biofilm growth and makeup are disrupted (i.e., dysbiosis), clinical symptoms associated with acute and chronic inflammatory responses to these changes may include pain, erythema and swelling. However, in most cases, these responses indicate that the immune system is functioning properly to re-establish homeostasis and protect the status quo. Defects in this healthy everyday function occur as a result of PMN subversion by pathological microbial strains, genetic defects or crosstalk with other chronic inflammatory conditions, including cancer and rheumatic disease, and this can provide some avenues for therapeutic targeting of PMN function. In other cases, targeting PMN functions could worsen the disease state. Certain PMN-mediated responses to pathogens, for example Neutrophil Extracellular Traps (NETs), might lead to undesirable symptoms such as pain or swelling and tissue damage/fibrosis. Despite collateral damage, these PMN responses limit pathogen dissemination and more severe damage that would otherwise occur. New data suggests the existence of unique PMN subsets, commonly associated with functional diversification in response to particular inflammatory challenges. PMN-directed therapeutic approaches depend on a greater understanding of this diversity. Here we outline the current understanding of PMNs in health and disease, with an emphasis on the positive manifestations of tissue and organ-protective PMN-mediated inflammation.
Collapse
Affiliation(s)
- Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Nikola Tasevski
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | | | - Howard C Tenenbaum
- Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Dental Oncology, Maxillofacial and Ocular Prosthetics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
42
|
Abstract
ABSTRACT Neutrophils play a critical role in the eradication of pathogenic organisms, particularly bacteria. However, in the septic patient the prolonged activation and accumulation of neutrophils may augment tissue and organ injury. This review discusses the different activation states and chemotaxis of neutrophils in septic patients. Neutrophil killing of bacteria and the formation of neutrophil extracellular traps represent important components of the innate immune response and they become dysregulated during sepsis, possibly through changes in their metabolism. Delayed neutrophil apoptosis may contribute to organ injury, or allow better clearance of pathogens. Neutrophils provide a friendly immune response to clear infections, but excessive activation and recruitment has the potential to turn them into potent foes.
Collapse
|
43
|
Abstract
Cirrhosis is a multisystemic disease wherein inflammatory responses originating from advanced liver disease and its sequelae affect distant compartments. Patients with cirrhosis are susceptible to bacterial infections, which may precipitate acute decompensation and acute-on-chronic liver failure, both of which are associated with high short-term mortality. Innate immune cells are an essential first line of defence against pathogens. Activation of liver macrophages (Kupffer cells) and resident mastocytes generate proinflammatory and vaso-permeating mediators that induce accumulation of neutrophils, lymphocytes, eosinophils and monocytes in the liver, and promote tissue damage. During cirrhosis progression, damage- and pathogen-associated molecular patterns activate immune cells and promote development of systemic inflammatory responses which may involve different tissues and compartments. The antibacterial function of circulating neutrophils and monocytes is gradually and severely impaired as cirrhosis worsens, contributing to disease progression. The mechanisms underlying impaired antimicrobial responses are complex and incompletely understood. This review focuses on the continuous and distinct perturbations arising in innate immune cells during cirrhosis, including their impact on disease progression, as well as reviewing potential therapeutic targets.
Collapse
|
44
|
Santos I, Colaço HG, Neves-Costa A, Seixas E, Velho TR, Pedroso D, Barros A, Martins R, Carvalho N, Payen D, Weis S, Yi HS, Shong M, Moita LF. CXCL5-mediated recruitment of neutrophils into the peritoneal cavity of Gdf15-deficient mice protects against abdominal sepsis. Proc Natl Acad Sci U S A 2020; 117:12281-12287. [PMID: 32424099 PMCID: PMC7275717 DOI: 10.1073/pnas.1918508117] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Sepsis is a life-threatening organ dysfunction condition caused by a dysregulated host response to an infection. Here we report that the circulating levels of growth and differentiation factor-15 (GDF15) are strongly increased in septic shock patients and correlate with mortality. In mice, we find that peptidoglycan is a potent ligand that signals through the TLR2-Myd88 axis for the secretion of GDF15, and that Gdf15-deficient mice are protected against abdominal sepsis due to increased chemokine CXC ligand 5 (CXCL5)-mediated recruitment of neutrophils into the peritoneum, leading to better local bacterial control. Our results identify GDF15 as a potential target to improve sepsis treatment. Its inhibition should increase neutrophil recruitment to the site of infection and consequently lead to better pathogen control and clearance.
Collapse
Affiliation(s)
- Isa Santos
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
- Serviço de Cirurgia Geral, Hospital de São Bernardo-Centro Hospitalar de Setúbal EPE, 2910-446 Setúbal, Portugal
| | - Henrique G Colaço
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Ana Neves-Costa
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Elsa Seixas
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Tiago R Velho
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Dora Pedroso
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - André Barros
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Rui Martins
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Nuno Carvalho
- Serviço de Cirurgia Geral, Hospital Garcia de Orta, 2801-951 Almada, Portugal
- Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisboa, Portugal
| | - Didier Payen
- INSERM, UMR 1160, Universite Paris 7 Denis Diderot, Universite-Sorbonne Cité, 75013 Paris, France
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, Jena University Hospital, 07747 Jena, Germany
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07747 Jena, Germany
- Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany
| | - Hyon-Seung Yi
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, 35015 Daejeon, Korea
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University School of Medicine, 35015 Daejeon, Korea
| | - Luís F Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal;
- Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisboa, Portugal
| |
Collapse
|
45
|
Jeong HM, Bang CS, Lee JJ, Baik GH. Delta Neutrophil Index for the Prediction of Prognosis in Acute Gastrointestinal Diseases; Diagnostic Test Accuracy Meta-Analysis. J Clin Med 2020; 9:jcm9041133. [PMID: 32326479 PMCID: PMC7230994 DOI: 10.3390/jcm9041133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 11/23/2022] Open
Abstract
Delta neutrophil index (DNI) is a novel diagnostic and prognostic biomarker of various infectious or inflammatory conditions. However, data on optimal measurement time are scarce, and no studies have evaluated the potential role of the DNI as a prognostic biomarker of gastrointestinal diseases with diagnostic test accuracy meta-analysis. Core databases were searched. The inclusion criteria were as follows: patients who have gastrointestinal diseases and DNI measurements presenting diagnostic indices for predicting the prognosis, including severity, surgical outcomes, and mortality from gastrointestinal diseases. We identified twelve studies for the systematic review and ten studies for the quantitative analysis. Pooled area under the curve, sensitivity, specificity, and diagnostic odds ratio of DNI at the initial admission date were 0.82 (95% confidence interval: 0.78–0.85), 0.75 (0.52–0.89), 0.76 (0.63–0.86), and 10 (3–35), respectively. Meta-regression showed no reasons for heterogeneity and publication bias was not detected. Fagan’s nomogram indicated that the posterior probability of ‘poor prognosis’ was 76% if the test was positive, and ‘no poor prognosis’ was 25% if the test was negative. The DNI can be considered as a reliable initial measurement biomarker for predicting prognosis in patients with gastrointestinal diseases,
Collapse
Affiliation(s)
- Hae Min Jeong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon 24253, Korea; (H.M.J.); (G.H.B.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Korea
| | - Chang Seok Bang
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon 24253, Korea; (H.M.J.); (G.H.B.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Korea
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24253, Korea;
- Correspondence: ; Tel.: +82-33-240-5821; Fax: +82-33-241-8064
| | - Jae Jun Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24253, Korea;
- Department of Anesthesiology and Pain Medicine, Hallym University College of Medicine, Chuncheon 24253, Korea
| | - Gwang Ho Baik
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon 24253, Korea; (H.M.J.); (G.H.B.)
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24253, Korea
| |
Collapse
|
46
|
Prognostic value of the delta neutrophil index in pediatric cardiac arrest. Sci Rep 2020; 10:3497. [PMID: 32103031 PMCID: PMC7044231 DOI: 10.1038/s41598-020-60126-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/07/2020] [Indexed: 12/17/2022] Open
Abstract
The delta neutrophil index (DNI), which reflects the ratio of circulating immature neutrophils, has been reported to be highly predictive of mortality in systemic inflammation. We investigated the prognostic significance of DNI value for early mortality and neurologic outcomes after pediatric cardiac arrest (CA). We retrospectively analyzed the data of eligible patients (<19 years in age). Among 85 patients, 55 subjects (64.7%) survived and 36 (42.4%) showed good outcomes at 30 days after CA. Cox regression analysis revealed that the DNI values immediately after the return of spontaneous circulation, at 24 hours and 48 hours after CA, were related to an increased risk for death within 30 days after CA (P < 0.001). A DNI value of higher than 3.3% at 24 hours could significantly predict both 30-day mortality (hazard ratio: 11.8; P < 0.001) and neurologic outcomes (odds ratio: 8.04; P = 0.003). The C statistic for multivariable prediction models for 30-day mortality (incorporating DNI at 24 hours, compression time, and serum sodium level) was 0.799, and the area under the receiver operating characteristic curve of DNI at 24 hours for poor neurologic outcome was 0.871. Higher DNI was independently associated with 30-day mortality and poor neurologic outcomes after pediatric CA.
Collapse
|
47
|
Cho KB, Park CH, Kim J, Tin TD, Kwak SH. Protective role of curcumin against lipopolysaccharide-induced inflammation and apoptosis in human neutrophil. Anesth Pain Med (Seoul) 2020; 15:41-48. [PMID: 33329788 PMCID: PMC7713868 DOI: 10.17085/apm.2020.15.1.41] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/23/2019] [Accepted: 10/04/2019] [Indexed: 11/17/2022] Open
Abstract
Background Sepsis, an uncontrolled host response to infection, may be life-threatening organ injury. Neutrophils play a critical role in regulation of host immune response to infection. Curcumin, known as a spice and food coloring agent, possesses anti-inflammatory properties. In this study, we investigated the effects of curcumin on lipopolysaccharide (LPS)-induced neutrophil activation with its signaling pathways. Methods Isolated human neutrophils were incubated without or with LPS and curcumin, and the expression of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, and IL-8 were assessed by enzyme-linked immunosorbent assays. The expression of mitogen-activated protein kinases such as p38, extracellularsignal-regulated kinase (ERK)1/2, and c-Jun N-terminal kinase (JNK) were evaluated by Western blot analysis. Neutrophil apoptosis was also measured by fluorescence-activated cell sorting (annexin V/propidium iodide) in LPS-stimulated neutrophils under treatment with curcumin. Results Curcumin attenuated expression of TNF-α, IL-6, and IL-8 and the phosphorylation levels of p38 and JNK, but not ERK1/2, in LPS-stimulated neutrophils. Additionally, curcumin restored the delayed neutrophil apoptosis by LPS-stimulated neutrophils(19.7 ± 3.2 to 38.2 ± 0.5%, P < 0.05). Conclusions Our results reveal the underlying mechanism of how curcumin attenuate neutrophil activation and suggest potential clinic applications of curcumin supplementation for patients with severe sepsis and septic shock. Additional clinical studies are required to confirm these in vitro findings.
Collapse
Affiliation(s)
- Kyu Bum Cho
- Department of Anesthesiology and Pain Medicine, Kwangju Christian Hospital, Gwangju, Korea
| | - Cheon Hee Park
- Department of Anesthesiology and Pain Medicine, Kwangju Christian Hospital, Gwangju, Korea
| | - Joungmin Kim
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea
| | - Tran Duc Tin
- Brain Korea 21 Project, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| | - Sang-Hyun Kwak
- Department of Anesthesiology and Pain Medicine, Chonnam National University Medical School and Hospital, Gwangju, Korea.,Brain Korea 21 Project, Center for Creative Biomedical Scientists at Chonnam National University, Gwangju, Korea
| |
Collapse
|
48
|
Liang W, Chen K, Gong W, Yoshimura T, Le Y, Wang Y, Wang JM. The Contribution of Chemoattractant GPCRs, Formylpeptide Receptors, to Inflammation and Cancer. Front Endocrinol (Lausanne) 2020; 11:17. [PMID: 32038501 PMCID: PMC6993212 DOI: 10.3389/fendo.2020.00017] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/09/2020] [Indexed: 12/14/2022] Open
Abstract
A hallmark of inflammatory responses is leukocyte mobilization, which is mediated by pathogen and host released chemotactic factors that activate Gi-protein-coupled seven-transmembrane receptors (GPCRs) on host cell surface. Formylpeptide receptors (FPRs, Fprs in mice) are members of the chemoattractant GPCR family, shown to be critical in myeloid cell trafficking during infection, inflammation, immune responses, and cancer progression. Accumulating evidence demonstrates that both human FPRs and murine Fprs are involved in a number of patho-physiological processes because of their expression on a wide variety of cell types in addition to myeloid cells. The unique capacity of FPRs (Fprs) to interact with numerous structurally unrelated chemotactic ligands enables these receptors to participate in orchestrated disease initiation, progression, and resolution. One murine Fpr member, Fpr2, and its endogenous agonist peptide, Cathelicidin-related antimicrobial peptide (CRAMP), have been demonstrated as key mediators of colon mucosal homeostasis and protection from inflammation and associated tumorigenesis. Recent availability of genetically engineered mouse models greatly expanded the understanding of the role of FPRs (Fprs) in pathophysiology that places these molecules in the list of potential targets for therapeutic intervention of diseases.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Keqiang Chen
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Teizo Yoshimura
- Department of Pathology and Experimental Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yingying Le
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Ji Ming Wang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| |
Collapse
|
49
|
Infection-induced innate antimicrobial response disorders: from signaling pathways and their modulation to selected biomarkers. Cent Eur J Immunol 2020; 45:104-116. [PMID: 32425688 PMCID: PMC7226557 DOI: 10.5114/ceji.2020.94712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Severe infections are a major public health problem responsible for about 40-65% of hospitalizations in intensive care units (ICU). The high mortality (30-50%) of persons diagnosed with severe infection is caused by largely unknown mechanisms of sepsis-induced immune system response. Severe infections with dynamic progress are accompanied with SIRS (systemic inflammatory reaction syndrome) and CARS (compensatory anti-inflammatory response syndrome), and require a biological treatment appropriate to the phase of immune response. The mechanisms responsible for severe infection related to immune system response particularly attract extensive interest of non-specific defense mechanisms, including signaling pathways of Toll-like receptors (mainly TLR4 and TLR2) that recognize distinct pathogen-associated molecular patterns (PAMP) and play a critical role in innate immune response. There are attempts of treatment, followed by blocking ligand binding with TLR or modulation of intracellular signaling pathways, to inhibit signal transduction. Moreover, researches regarding new and more efficient diagnostics biomarkers were mostly focused on indicators related to innate response to infection as well as connections of pro-inflammatory response with anti-inflammatory response.According to these studies, in case of ICU septic patients with high-risk of mortality, the solution for the problem will require mainly early immune and genetic diagnostics (e.g. cytokines, microRNA, cluster of differentiation-64 [CD64], triggering receptor expressed on myeloid cells-1 [TREM-1], and high mobility group box 1 protein [HMGB1]).
Collapse
|
50
|
Yao RQ, Ren C, Wang JN, Wu GS, Zhu XM, Xia ZF, Yao YM. Publication Trends of Research on Sepsis and Host Immune Response during 1999-2019: A 20-year Bibliometric Analysis. Int J Biol Sci 2020; 16:27-37. [PMID: 31892843 PMCID: PMC6930382 DOI: 10.7150/ijbs.37496] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/28/2019] [Indexed: 11/05/2022] Open
Abstract
Introduction: Sepsis is an intractable disorder, which is associated with high risk of organ dysfunction and even death, while its pathogenesis remains largely unclear. Our study aims to study the research trend on sepsis and host immune response, and compare the contribution of publications from different countries, institutions, journals and authors. Materials and Methods: We extracted all relevant publications with regard to sepsis and immune response during 1999-2019 from Web of Science. GraphPad Prism 6, and VOSviewer software were used to collect and analyze the publication trend in related field. Results: We identified a total of 1225 publications with citation frequency of 40511 times up to March 30, 2019. The United States accounted for the largest number of publications (36.3%), 51.9% of total citations as well as the highest H-index (72). The sum of publications from China ranked the second, while the overall citations (1935) and H-index (22) ranked the eighth and the seventh, respectively. Journal of Shock had published most papers related to the topic on sepsis and immune response. Ayala A SA, has published the most papers in this field (31), while Hotchkiss RS presented with the most citation frequency (3532). The keyword “regulatory T cell” appeared most recently with an average appearing years of 2014.0. The “immunosuppression related research” seemed to be the hotspot in relevant scope. Conclusions: The United States made the most outstanding contribution within this important field. There is a mismatch between the quantity and quality of publications from China. Latest progress can be tracked in journal of Shock. Immunosuppression related researches may be hotspots in the near future.
Collapse
Affiliation(s)
- Ren-Qi Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China.,Department of Burn Surgery, Changhai Hospital, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Chao Ren
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Jun-Nan Wang
- Basic Medical College, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Guo-Sheng Wu
- Department of Burn Surgery, Changhai Hospital, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Xiao-Mei Zhu
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| | - Zhao-Fan Xia
- Department of Burn Surgery, Changhai Hospital, the Naval Medical University, Shanghai 200433, People's Republic of China
| | - Yong-Ming Yao
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100048, People's Republic of China
| |
Collapse
|