1
|
Mon-Wei Yu S, King E, Fribourg M, Hartzell S, Tsou L, Gee L, D'Agati VD, Thurman JM, He JC, Cravedi P. A Newly Identified Protective Role of C5a Receptor 1 in Kidney Tubules against Toxin-Induced Acute Kidney Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2024:S0002-9440(24)00367-5. [PMID: 39427763 DOI: 10.1016/j.ajpath.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/03/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Acute kidney injury (AKI) remains a major reason for hospitalization with limited therapeutic options. Although complement activation is implicated in AKI, the role of C5a receptor 1 (C5aR1) in kidney tubular cells is unclear. We used aristolochic acid nephropathy (AAN) and folic acid nephropathy models to establish the role of C5aR1 in kidney tubules during AKI in germline C5ar1-/- mice, myeloid cell-specific mice, and kidney tubule-specific C5ar1 knockout mice. After aristolochic acid and folic acid injection, C5ar1-/- mice had increased AKI severity and a higher degree of tubular injury. Macrophage depletion in C5ar1-/- mice or myeloid cell-specific C5ar1 deletion did not affect the outcomes of aristolochic acid-induced AKI. RNA-sequencing data from renal tubular epithelial cells (RTECs) showed that C5ar1 deletion was associated with the down-regulation of mitochondrial metabolism and ATP production transcriptional pathways. Metabolic studies confirmed reduced mitochondrial membrane potential at baseline and increased mitochondrial oxidative stress after injury in C5ar1-/- RTECs. Moreover, C5ar1-/- RTECs had enhanced glycolysis, glucose uptake, and lactate production on injury, corroborated by metabolomics analysis of kidneys from AAN mice. Kidney tubule-specific C5ar1 knockout mice recapitulated exacerbated AKI observed in C5ar1-/- mice in AAN and folic acid nephropathy. Our data indicate that C5aR1 signaling in kidney tubules exerts renoprotective effects against toxin-induced AKI by limiting overt glycolysis and maintaining mitochondrial function, revealing a novel link between the complement system and tubular cell metabolism.
Collapse
Affiliation(s)
- Samuel Mon-Wei Yu
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York.
| | - Emily King
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York
| | - Miguel Fribourg
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York
| | - Susan Hartzell
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York
| | - Liam Tsou
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York
| | - Logan Gee
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York
| | - Vivette D D'Agati
- Department of Pathology and Cell Biology, Columbia University, New York, New York
| | - Joshua M Thurman
- Medicine-Renal Med Diseases/Hypertension, Colorado University, Aurora, Colorado
| | - John Cijiang He
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York; James J. Peters Veteran Administration Medical Center, New York, New York
| | - Paolo Cravedi
- Nephrology Division, Department of Medicine, Mount Sinai Hospital, New York, New York.
| |
Collapse
|
2
|
Guo G, Liu Z, Yu J, You Y, Li M, Wang B, Tang J, Han P, Wu J, Shen H. Neutrophil Function Conversion Driven by Immune Switchpoint Regulator against Diabetes-Related Biofilm Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310320. [PMID: 38035713 DOI: 10.1002/adma.202310320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/28/2023] [Indexed: 12/02/2023]
Abstract
Reinforced biofilm structures and dysfunctional neutrophils induced by excessive oxidative stress contribute to the refractoriness of diabetes-related biofilm infections (DRBIs). Herein, in contrast to traditional antibacterial therapies, an immune switchpoint-driven neutrophil immune function conversion strategy based on a deoxyribonuclease I loaded vanadium carbide MXene (DNase-I@V2 C) nanoregulator is proposed to treat DRBIs via biofilm lysis and redirecting neutrophil functions from NETosis to phagocytosis in diabetes. Owing to its intrinsic superoxide dismutase/catalase-like activities, DNase-I@V2 C effectively scavenges reactive oxygen species (ROS) in a high oxidative stress microenvironment to maintain the biological activity of DNase-I. By increasing the depth of biofilm penetration of DNase-I, DNase-I@V2 C thoroughly degrades extracellular DNA and neutrophil extracellular traps (NETs) in extracellular polymeric substances, thus breaking the physical barrier of biofilms. More importantly, as an immune switchpoint regulator, DNase-I@V2 C can skew neutrophil functions from NETosis toward phagocytosis by intercepting ROS-NE/MPO-PAD4 and activating ROS-PI3K-AKT-mTOR pathways in diabetic microenvironment, thereby eliminating biofilm infections. Biofilm lysis and synergistic neutrophil function conversion exert favorable therapeutic effects on biofilm infections in vitro and in vivo. This study serves as a proof-of-principle demonstration of effectively achieving DRBIs with high therapeutic efficacy by regulating immune switchpoint to reverse neutrophil functions.
Collapse
Affiliation(s)
- Geyong Guo
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Zihao Liu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Jinlong Yu
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Yanan You
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Fudan University, Shanghai, 200090, P. R. China
| | - Mingzhang Li
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Boyong Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Jin Tang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Pei Han
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Jianrong Wu
- Department of Ultrasound in Medicine, Shanghai Institute of Ultrasound in Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| | - Hao Shen
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, P. R. China
| |
Collapse
|
3
|
Ishii M, Rohrer B. Anaphylatoxin C5a receptor signaling induces mitochondrial fusion and sensitizes retinal pigment epithelial cells to oxidative stress. Biochim Biophys Acta Gen Subj 2023; 1867:130374. [PMID: 37187450 PMCID: PMC10330548 DOI: 10.1016/j.bbagen.2023.130374] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023]
Abstract
Mitochondrial dynamics is a morphological balance between fragmented and elongated shapes, reflecting mitochondrial metabolic status, cellular damage, and mitochondrial dysfunction. The anaphylatoxin C5a derived from complement component 5 cleavage, enhances cellular responses involved in pathological stimulation, innate immune responses, and host defense. However, the specific response of C5a and its receptor, C5a receptor (C5aR), in mitochondria is unclear. Here, we tested whether the C5a/C5aR signaling axis affects mitochondrial morphology in human-derived retinal pigment epithelial cell monolayers (ARPE-19). C5aR activation with the C5a polypeptide induced mitochondrial elongation. In contrast, oxidatively stressed cells (H2O2) responded to C5a with an enhancement of mitochondrial fragmentation and an increase in the number of pyknotic nuclei. C5a/C5aR signaling increased the expression of mitochondrial fusion-related protein, mitofusin-1 (MFN1) and - 2 (MFN2), as well as enhanced optic atrophy-1 (Opa1) cleavage, which are required for mitochondrial fusion events, whereas the mitochondrial fission protein, dynamin-related protein-1 (Drp1), and mitogen-activated protein kinase (MAPK)-dependent extracellular signal-regulated protein kinase (Erk1/2) phosphorylation were not affected. Moreover, C5aR activation increased the frequency of endoplasmic reticulum (ER)-mitochondria contacts. Finally, oxidative stress induced in a single cell within an RPE monolayer (488 nm blue laser spot stimulation) induced a bystander effect of mitochondrial fragmentation in adjacent surrounding cells only in C5a-treated monolayers. These results suggest that C5a/C5aR signaling produced an intermediate state, characterized by increased mitochondrial fusion and ER-mitochondrial contacts, that sensitizes cells to oxidative stress, leading to mitochondrial fragmentation and cell death.
Collapse
Affiliation(s)
- Masaaki Ishii
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425, USA.
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, SC 29425, USA; Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA; Department of Neurosciences, Medical University of South Carolina, Charleston, SC, 29425, USA.
| |
Collapse
|
4
|
Fu XZ, Wang Y. Interferon-γ regulates immunosuppression in septic mice by promoting the Warburg effect through the PI3K/AKT/mTOR pathway. Mol Med 2023; 29:95. [PMID: 37434129 PMCID: PMC10337057 DOI: 10.1186/s10020-023-00690-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 06/20/2023] [Indexed: 07/13/2023] Open
Abstract
BACKGROUND The main cause of high mortality from sepsis is that immunosuppression leads to life-threatening organ dysfunction, and reversing immunosuppression is key to sepsis treatment. Interferon γ (IFNγ) is a potential therapy for immunosuppression of sepsis, promoting glycolysis to restore metabolic defects in monocytes, but the mechanism of treatment is unclear. METHODS To explore the immunotherapeutic mechanism of IFNγ, this study linked the Warburg effect (aerobic glycolysis) to immunotherapy for sepsis and used cecal ligation perforation (CLP) and lipopolysaccharide (LPS) to stimulate dendritic cells (DC) to establish in vivo and in vitro sepsis models, Warburg effect inhibitors (2-DG) and PI3K pathway inhibitors (LY294002) were used to explore the mechanism by which IFNγ regulates immunosuppression in mice with sepsis through the Warburg effect. RESULTS IFNγ markedly inhibited the reduction in cytokine secretion from lipopolysaccharide (LPS)-stimulated splenocytes. IFNγ-treated mice had significantly increased the percentages of positive costimulatory receptor CD86 on Dendritic cells expressing and expression of splenic HLA-DR. IFNγ markedly reduced DC-cell apoptosis by upregulating the expression of Bcl-2 and downregulating the expression of Bax. CLP-induced formation of regulatory T cells in the spleen was abolished in IFNγ -treated mice. IFNγ treatment reduced the expression of autophagosomes in DC cells. IFNγ significant reduce the expression of Warburg effector-related proteins PDH, LDH, Glut1, and Glut4, and promote glucose consumption, lactic acid, and intracellular ATP production. After the use of 2-DG to suppress the Warburg effect, the therapeutic effect of IFNγ was suppressed, demonstrating that IFNγ reverses immunosuppression by promoting the Warburg effect. Moreover, IFNγ increased the expression of phosphoinositide 3-kinases (PI3K), protein kinase B (Akt), rapamycin target protein (mTOR), hypoxia-inducible factor-1 (HIF-1α), pyruvate dehydrogenase kinase (PDK1) protein, the use of 2-DG and LY294002 can inhibit the expression of the above proteins, LY294002 also inhibits the therapeutic effect of IFNγ. CONCLUSIONS It was finally proved that IFNγ promoted the Warburg effect through the PI3K/Akt/mTOR pathway to reverse the immunosuppression caused by sepsis. This study elucidates the potential mechanism of the immunotherapeutic effect of IFNγ in sepsis, providing a new target for the treatment of sepsis.
Collapse
Affiliation(s)
- Xu-Zhe Fu
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
5
|
Wang Y, Zhu CL, Li P, Liu Q, Li HR, Yu CM, Deng XM, Wang JF. The role of G protein-coupled receptor in neutrophil dysfunction during sepsis-induced acute respiratory distress syndrome. Front Immunol 2023; 14:1112196. [PMID: 36891309 PMCID: PMC9986442 DOI: 10.3389/fimmu.2023.1112196] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Sepsis is defined as a life-threatening dysfunction due to a dysregulated host response to infection. It is a common and complex syndrome and is the leading cause of death in intensive care units. The lungs are most vulnerable to the challenge of sepsis, and the incidence of respiratory dysfunction has been reported to be up to 70%, in which neutrophils play a major role. Neutrophils are the first line of defense against infection, and they are regarded as the most responsive cells in sepsis. Normally, neutrophils recognize chemokines including the bacterial product N-formyl-methionyl-leucyl-phenylalanine (fMLP), complement 5a (C5a), and lipid molecules Leukotriene B4 (LTB4) and C-X-C motif chemokine ligand 8 (CXCL8), and enter the site of infection through mobilization, rolling, adhesion, migration, and chemotaxis. However, numerous studies have confirmed that despite the high levels of chemokines in septic patients and mice at the site of infection, the neutrophils cannot migrate to the proper target location, but instead they accumulate in the lungs, releasing histones, DNA, and proteases that mediate tissue damage and induce acute respiratory distress syndrome (ARDS). This is closely related to impaired neutrophil migration in sepsis, but the mechanism involved is still unclear. Many studies have shown that chemokine receptor dysregulation is an important cause of impaired neutrophil migration, and the vast majority of these chemokine receptors belong to the G protein-coupled receptors (GPCRs). In this review, we summarize the signaling pathways by which neutrophil GPCR regulates chemotaxis and the mechanisms by which abnormal GPCR function in sepsis leads to impaired neutrophil chemotaxis, which can further cause ARDS. Several potential targets for intervention are proposed to improve neutrophil chemotaxis, and we hope that this review may provide insights for clinical practitioners.
Collapse
Affiliation(s)
- Yi Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Cheng-long Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Peng Li
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Qiang Liu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hui-ru Li
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Faculty of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Chang-meng Yu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiao-ming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Faculty of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Jia-feng Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
6
|
Damascena HL, Silveira WAA, Castro MS, Fontes W. Neutrophil Activated by the Famous and Potent PMA (Phorbol Myristate Acetate). Cells 2022; 11:2889. [PMID: 36139464 PMCID: PMC9496763 DOI: 10.3390/cells11182889] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
This review will briefly outline the major signaling pathways in PMA-activated neutrophils. PMA is widely used to understand neutrophil pathways and formation of NETs. PMA activates PKC; however, we highlight some isoforms that contribute to specific functions. PKC α, β and δ contribute to ROS production while PKC βII and PKC ζ are involved in cytoskeleton remodeling. Actin polymerization is important for the chemotaxis of neutrophils and its remodeling is connected to ROS balance. We suggest that, although ROS and production of NETs are usually observed together in PMA-activated neutrophils, there might be a regulatory mechanism balancing both. Interestingly, we suggest that serine proteases might determine the PAD4 action. PAD4 could be responsible for the activation of the NF-κB pathway that leads to IL-1β release, triggering the cleavage of gasdermin D by serine proteases such as elastase, leading to pore formation contributing to release of NETs. On the other hand, when serine proteases are inhibited, NETs are formed by citrullination through the PAD4 pathway. This review puts together results from the last 31 years of research on the effects of PMA on the neutrophil and proposes new insights on their interpretation.
Collapse
Affiliation(s)
| | | | | | - Wagner Fontes
- Laboratory of Protein Chemistry and Biochemistry, Department of Cell Biology, Institute of Biological Sciences, University of Brasília, Federal District, Brasilia 70910-900, Brazil
| |
Collapse
|
7
|
Pan T, Sun S, Chen Y, Tian R, Chen E, Tan R, Wang X, Liu Z, Liu J, Qu H. Immune effects of PI3K/Akt/HIF-1α-regulated glycolysis in polymorphonuclear neutrophils during sepsis. Crit Care 2022; 26:29. [PMID: 35090526 PMCID: PMC8796568 DOI: 10.1186/s13054-022-03893-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/10/2022] [Indexed: 12/29/2022] Open
Abstract
Background Effective removal of pathogenic bacteria is key to improving the prognosis of sepsis. Polymorphonuclear neutrophils (PMNs) are the most important components of innate cellular immunity and play vital roles in clearing pathogenic bacteria. However, the metabolic characteristics and immunomodulatory pathways of PMNs during sepsis have not been investigated. In the present study, we explored the immune metabolism characteristics of PMNs and the mechanism by which neutrophilic glycolysis is regulated during sepsis. Methods Metabolomics analysis was performed on PMNs isolated from 14 septic patients, 26 patients with acute appendicitis, and 19 healthy volunteers. Transcriptome analysis was performed on the PMNs isolated from the healthy volunteers and the patients with sepsis to assess glycolysis and investigate its mechanism. Lipopolysaccharide (LPS) was used to stimulate the neutrophils isolated from the healthy volunteers at different time intervals to build an LPS-tolerant model. Chemotaxis, phagocytosis, lactate production, oxygen consumption rate (OCR), and extracellular acidification rate (ECAR) were evaluated. Results Transcriptomics showed significant changes in glycolysis and the mTOR/HIF-1α signaling pathway during sepsis. Metabolomics revealed that the Warburg effect was significantly altered in the patients with sepsis. We discovered that glycolysis regulated PMNs’ chemotaxis and phagocytosis functions during sepsis. Lactate dehydrogenase A (LDHA) downregulation was a key factor in the inhibition of glycolysis in PMNs. This study confirmed that the PI3K/Akt-HIF-1α pathway was involved in the LDHA expression level and also influenced PMNs’ chemotaxis and phagocytosis functions. Conclusions The inhibition of glycolysis contributed to neutrophil immunosuppression during sepsis and might be controlled by PI3K/Akt-HIF-1α pathway-mediated LDHA downregulation. Our study provides a scientific theoretical basis for the management and treatment of patients with sepsis and promotes to identify therapeutic target for the improvement of immune function in sepsis. Supplementary Information The online version contains supplementary material available at 10.1186/s13054-022-03893-6.
Collapse
|
8
|
Cui CS, Kumar V, Gorman DM, Clark RJ, Lee JD, Woodruff TM. In Vivo Pharmacodynamic Method to Assess Complement C5a Receptor Antagonist Efficacy. ACS Pharmacol Transl Sci 2022; 5:41-51. [PMID: 35059568 DOI: 10.1021/acsptsci.1c00227] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Indexed: 12/17/2022]
Abstract
The complement C5a receptor 1 (C5aR1) has been studied as a potential therapeutic target for autoimmune and inflammatory diseases, with several drug candidates identified. Understanding the pharmacokinetics and pharmacodynamics of a drug candidate is a crucial preclinical step that allows for a greater understanding of a compound's in vivo biodistribution and target engagement to assist in clinical dose selection and dosing frequency. However, few in vivo pharmacodynamic methods have been described for C5a inhibitors. In this study, we, therefore, developed a complete in vivo pharmacodynamic assay in mice and applied this method to the peptide-based C5aR1 antagonists PMX53 and JPE-1375. Intravenous administration of recombinant mouse C5a induced rapid neutrophil mobilization and plasma TNF elevation over a 60 min period. By using C5a receptor-deficient mice, we demonstrated that this response was driven primarily through C5aR1. We next identified using this model that both PMX53 and JPE-1375 have similar in vivo working doses that can inhibit C5aR1-mediated neutrophilia and cytokine production in a dose as low as 1 mg/kg following intravenous injection. However, the in vivo active duration for PMX53 lasted for up to 6 h, significantly longer than that for JPE-1375 (<2 h). Pharmacokinetic analysis demonstrated rapid plasma distribution and elimination of both compounds, although PMX53 had a longer half-life, which allowed for the development of an accurate pharmacokinetic/pharmacodynamic model. Overall, our study developed a robust in vivo pharmacodynamic model for C5aR1 inhibitors in mice that may assist in preclinical translational studies of therapeutic drug candidates targeting C5a and its receptors.
Collapse
Affiliation(s)
- Cedric S Cui
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Vinod Kumar
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Declan M Gorman
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Richard J Clark
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia.,Queensland Brain Institute, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| |
Collapse
|
9
|
Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther 2021; 6:407. [PMID: 34824200 PMCID: PMC8613465 DOI: 10.1038/s41392-021-00816-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection. Over decades, advanced understanding of host-microorganism interaction has gradually unmasked the genuine nature of sepsis, guiding toward new definition and novel therapeutic approaches. Diverse clinical manifestations and outcomes among infectious patients have suggested the heterogeneity of immunopathology, while systemic inflammatory responses and deteriorating organ function observed in critically ill patients imply the extensively hyperactivated cascades by the host defense system. From focusing on microorganism pathogenicity, research interests have turned toward the molecular basis of host responses. Though progress has been made regarding recognition and management of clinical sepsis, incidence and mortality rate remain high. Furthermore, clinical trials of therapeutics have failed to obtain promising results. As far as we know, there was no systematic review addressing sepsis-related molecular signaling pathways and intervention therapy in literature. Increasing studies have succeeded to confirm novel functions of involved signaling pathways and comment on efficacy of intervention therapies amid sepsis. However, few of these studies attempt to elucidate the underlining mechanism in progression of sepsis, while other failed to integrate preliminary findings and describe in a broader view. This review focuses on the important signaling pathways, potential molecular mechanism, and pathway-associated therapy in sepsis. Host-derived molecules interacting with activated cells possess pivotal role for sepsis pathogenesis by dynamic regulation of signaling pathways. Cross-talk and functions of these molecules are also discussed in detail. Lastly, potential novel therapeutic strategies precisely targeting on signaling pathways and molecules are mentioned.
Collapse
Affiliation(s)
- Yun-Yu Zhang
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Bo-Tao Ning
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| |
Collapse
|
10
|
Ehrnthaller C, Braumüller S, Kellermann S, Gebhard F, Perl M, Huber-Lang M. Complement Factor C5a Inhibits Apoptosis of Neutrophils-A Mechanism in Polytrauma? J Clin Med 2021; 10:jcm10143157. [PMID: 34300323 PMCID: PMC8303460 DOI: 10.3390/jcm10143157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/15/2021] [Indexed: 01/03/2023] Open
Abstract
Life-threatening polytrauma results in early activation of the complement and apoptotic system, as well as leukocytes, ultimately leading to the clearance of damaged cells. However, little is known about interactions between the complement and apoptotic systems in PMN (polymorphonuclear neutrophils) after multiple injuries. PMN from polytrauma patients and healthy volunteers were obtained and assessed for apoptotic events along the post-traumatic time course. In vitro studies simulated complement activation by the exposure of PMN to C3a or C5a and addressed both the intrinsic and extrinsic apoptotic pathway. Specific blockade of the C5a-receptor 1 (C5aR1) on PMN was evaluated for efficacy to reverse complement-driven alterations. PMN from polytrauma patients exhibited significantly reduced apoptotic rates up to 10 days post trauma compared to healthy controls. Polytrauma-induced resistance was associated with significantly reduced Fas-ligand (FasL) and Fas-receptor (FasR) on PMN and in contrast, significantly enhanced FasL and FasR in serum. Simulation of systemic complement activation revealed for C5a, but not for C3a, a dose-dependent abrogation of PMN apoptosis in both intrinsic and extrinsic pathways. Furthermore, specific blockade of the C5aR1 reversed C5a-induced PMN resistance to apoptosis. The data suggest an important regulatory and putative mechanistic and therapeutic role of the C5a/C5aR1 interaction on PMN apoptosis after polytrauma.
Collapse
Affiliation(s)
- Christian Ehrnthaller
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
- Department of Orthopedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), University Hospital, LMU Munich, 81377 Munich, Germany
- Correspondence: (C.E.); (M.H.-L.)
| | - Sonja Braumüller
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
| | - Stephanie Kellermann
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
| | - Florian Gebhard
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 89081 Ulm, Germany; (F.G.); (M.P.)
| | - Mario Perl
- Department of Traumatology, Hand-, Plastic-, and Reconstructive Surgery, Center of Surgery, University of Ulm, 89081 Ulm, Germany; (F.G.); (M.P.)
- Department of Traumatology and Orthopaedic Surgery, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology (ITI), University of Ulm, 89081 Ulm, Germany; (S.B.); (S.K.)
- Correspondence: (C.E.); (M.H.-L.)
| |
Collapse
|
11
|
Nording H, Baron L, Haberthür D, Emschermann F, Mezger M, Sauter M, Sauter R, Patzelt J, Knoepp K, Nording A, Meusel M, Meyer-Saraei R, Hlushchuk R, Sedding D, Borst O, Eitel I, Karsten CM, Feil R, Pichler B, Erdmann J, Verschoor A, Chavakis E, Chavakis T, von Hundelshausen P, Köhl J, Gawaz M, Langer HF. The C5a/C5a receptor 1 axis controls tissue neovascularization through CXCL4 release from platelets. Nat Commun 2021; 12:3352. [PMID: 34099640 PMCID: PMC8185003 DOI: 10.1038/s41467-021-23499-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 03/28/2021] [Indexed: 02/05/2023] Open
Abstract
Platelets contribute to the regulation of tissue neovascularization, although the specific factors underlying this function are unknown. Here, we identified the complement anaphylatoxin C5a-mediated activation of C5a receptor 1 (C5aR1) on platelets as a negative regulatory mechanism of vessel formation. We showed that platelets expressing C5aR1 exert an inhibitory effect on endothelial cell functions such as migration and 2D and 3D tube formation. Growth factor- and hypoxia-driven vascularization was markedly increased in C5ar1-/- mice. Platelet-specific deletion of C5aR1 resulted in a proangiogenic phenotype with increased collateralization, capillarization and improved pericyte coverage. Mechanistically, we found that C5a induced preferential release of CXC chemokine ligand 4 (CXCL4, PF4) from platelets as an important antiangiogenic paracrine effector molecule. Interfering with the C5aR1-CXCL4 axis reversed the antiangiogenic effect of platelets both in vitro and in vivo.In conclusion, we identified a mechanism for the control of tissue neovascularization through C5a/C5aR1 axis activation in platelets and subsequent induction of the antiangiogenic factor CXCL4.
Collapse
Affiliation(s)
- Henry Nording
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany ,grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany
| | - Lasse Baron
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - David Haberthür
- grid.5734.50000 0001 0726 5157Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Frederic Emschermann
- grid.10392.390000 0001 2190 1447University Hospital, Department of Cardiovascular Medicine, Eberhard Karls University, Tübingen, Germany
| | - Matthias Mezger
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Manuela Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Reinhard Sauter
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Johannes Patzelt
- grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Kai Knoepp
- grid.9018.00000 0001 0679 2801Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Halle (Saale), Germany
| | - Anne Nording
- grid.10392.390000 0001 2190 1447Institute of Medical Genetics and Applied Genomics, Eberhard Karls University, Tübingen, Germany
| | - Moritz Meusel
- grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Roza Meyer-Saraei
- grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Ruslan Hlushchuk
- grid.5734.50000 0001 0726 5157Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Daniel Sedding
- grid.9018.00000 0001 0679 2801Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, Martin-Luther-University Halle (Saale), Halle (Saale), Germany
| | - Oliver Borst
- grid.10392.390000 0001 2190 1447University Hospital, Department of Cardiovascular Medicine, Eberhard Karls University, Tübingen, Germany
| | - Ingo Eitel
- grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| | - Christian M. Karsten
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Robert Feil
- grid.10392.390000 0001 2190 1447Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Bernd Pichler
- grid.10392.390000 0001 2190 1447Institute for Preclinical Imaging, Eberhard Karls University, Tübingen, Germany
| | - Jeanette Erdmann
- grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.4562.50000 0001 0057 2672Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Admar Verschoor
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Emmanouil Chavakis
- grid.411088.40000 0004 0578 8220Department for Internal Medicine III/Cardiology, University Hospital of the Johann-Wolfgang Goethe University, Frankfurt am Main, Germany
| | - Triantafyllos Chavakis
- grid.4488.00000 0001 2111 7257Department of Clinical Pathobiochemistry, Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, Technische Universität Dresden, Dresden, Germany
| | - Philipp von Hundelshausen
- grid.5252.00000 0004 1936 973XInstitute for Cardiovascular Prevention, Ludwig Maximilians University Munich, Munich, Germany
| | - Jörg Köhl
- grid.4562.50000 0001 0057 2672Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany ,grid.239573.90000 0000 9025 8099Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Meinrad Gawaz
- grid.10392.390000 0001 2190 1447University Hospital, Department of Cardiovascular Medicine, Eberhard Karls University, Tübingen, Germany
| | - Harald F. Langer
- Cardioimmunology Group, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany ,grid.452396.f0000 0004 5937 5237DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck, Germany ,grid.412468.d0000 0004 0646 2097University Hospital, Medical Clinic II, University Heart Center Lübeck, Lübeck, Germany
| |
Collapse
|
12
|
Sommerfeld O, Medyukhina A, Neugebauer S, Ghait M, Ulferts S, Lupp A, König R, Wetzker R, Schulz S, Figge MT, Bauer M, Press AT. Targeting Complement C5a Receptor 1 for the Treatment of Immunosuppression in Sepsis. Mol Ther 2021; 29:338-346. [PMID: 32966769 PMCID: PMC7791006 DOI: 10.1016/j.ymthe.2020.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/25/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
Complement factor C5a was originally identified as a powerful promoter of inflammation through activation of the C5a receptor 1 (C5ar1). Recent evidence suggests involvement of C5a not only in pro- but also in anti-inflammatory signaling. The present study aims to unveil the role of C5ar1 as potential therapeutic target in a murine sepsis model. Our study discloses a significantly increased survival in models of mild to moderate but not severe sepsis of C5ar1-deficient mice. The decreased mortality of C5ar1-deficient mice is accompanied by improved pathogen clearance and largely preserved liver function. C5ar1-deficient mice exhibited a significantly increased production of the pro-inflammatory mediator interferon-γ (IFN-γ) and a decreased production of the anti-inflammatory cytokine interleukin-10 (IL-10). Together, these data uncover C5a signaling as a mediator of immunosuppressive processes during sepsis and describe the C5ar1 and related changes of the IFN-γ to IL-10 ratio as markers for the immunological (dys)function accompanying sepsis.
Collapse
Affiliation(s)
- Oliver Sommerfeld
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Anna Medyukhina
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany
| | - Sophie Neugebauer
- Institute of Clinical Chemistry and Laboratory Diagnostics, Jena University Hospital, Jena, Germany
| | - Mohamed Ghait
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Svenja Ulferts
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Rainer König
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany; Network Modeling, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute Jena, Jena, Germany
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Stefan Schulz
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany; Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Marc Thilo Figge
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany; Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany; Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Adrian T Press
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.
| |
Collapse
|
13
|
Blood Leukocyte Signaling Pathways as Predictors of Severity of Acute Pancreatitis. Pancreas 2021; 50:710-718. [PMID: 34016897 PMCID: PMC8195735 DOI: 10.1097/mpa.0000000000001832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Clinical practice lacks biomarkers to predict the severity of acute pancreatitis (AP). We studied if intracellular signaling of circulating leukocytes could predict persistent organ dysfunction (OD) and secondary infections in AP. METHODS A venous blood sample was taken from 174 patients with AP 72 hours or less from onset of symptoms and 31 healthy controls. Phosphorylation levels (p) of appropriately stimulated signal transducer and activator of transcription 1 (STAT1), STAT6, nuclear factor-κB (NF-κB), Akt, and nonstimulated STAT3 in monocytes, neutrophils, and lymphocytes was measured using phosphospecific flow cytometry. RESULTS The patients showed higher pSTAT3 and lower pSTAT1, pSTAT6, pNF-κB, and pAkt than healthy controls. pSTAT3 in all leukocyte subtypes studied increased, and pSTAT1 in monocytes and T cells decreased in an AP severity-wise manner. In patients without OD at sampling, high pSTAT3 in monocytes and T lymphocytes were associated with development of persistent OD. In patients with OD, low interleukin-4-stimulated pSTAT6 in monocytes and neutrophils and Escherichia coli-stimulated pNF-κB in neutrophils predicted OD persistence. High pSTAT3 in monocytes, CD8+ T cells, and neutrophils; low pSTAT1 in monocytes and T cells; and low pNF-κB in lymphocytes predicted secondary infections. CONCLUSIONS Leukocyte STAT3, STAT1, STAT6, and NF-κΒ phosphorylations are potential predictors of AP severity.
Collapse
|
14
|
Activated peripheral blood mononuclear cell mediators trigger astrocyte reactivity. Brain Behav Immun 2019; 80:879-888. [PMID: 31176000 DOI: 10.1016/j.bbi.2019.05.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/28/2019] [Accepted: 05/30/2019] [Indexed: 12/23/2022] Open
Abstract
Sepsis is characterized by a severe and disseminated inflammation. In the central nervous system, sepsis promotes synaptic dysfunction and permanent cognitive impairment. Besides sepsis-induced neuronal dysfunction, glial cell response has been gaining considerable attention with microglial activation as a key player. By contrast, astrocytes' role during acute sepsis is still underexplored. Astrocytes are specialized immunocompetent cells involved in brain surveillance. In this context, the potential communication between the peripheral immune system and astrocytes during acute sepsis still remains unclear. We hypothesized that peripheral blood mononuclear cell (PBMC) mediators are able to affect the brain during an episode of acute sepsis. With this in mind, we first performed a data-driven transcriptome analysis of blood from septic patients to identify common features among independent clinical studies. Our findings evidenced pronounced impairment in energy-related signaling pathways in the blood of septic patients. Since astrocytes are key for brain energy homeostasis, we decided to investigate the communication between PBMC mediators and astrocytes in a rat model of acute sepsis, induced by cecal ligation and perforation (CLP). In the CLP animals, we identified widespread in vivo brain glucose hypometabolism. Ex vivo analyses demonstrated astrocyte reactivity along with reduced glutamate uptake capacity during sepsis. Also, by exposing cultured astrocytes to mediators released by PBMCs from CLP animals, we reproduced the energetic failure observed in vivo. Finally, by pharmacologically inhibiting phosphoinositide 3-kinase (PI3K), a central metabolic pathway downregulated in the blood of septic patients and reduced in the CLP rat brain, we mimicked the PBMC mediators effect on glutamate uptake but not on glucose metabolism. These results suggest that PBMC mediators are capable of directly mediating astrocyte reactivity and contribute to the brain energetic failure observed in acute sepsis. Moreover, the evidence of PI3K participation in this process indicates a potential target for therapeutic modulation.
Collapse
|
15
|
Lin Y, Zhang Y, Yu H, Tian R, Wang G, Li F. Identification of unique key genes and miRNAs in latent tuberculosis infection by network analysis. Mol Immunol 2019; 112:103-114. [PMID: 31082644 DOI: 10.1016/j.molimm.2019.04.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/17/2019] [Accepted: 04/30/2019] [Indexed: 02/04/2023]
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (M.tb). New cases are now mainly caused by the progression of latent tuberculosis infection (LTBI). Thus, methods to diagnose and treat LTBI are urgently needed to prevent the development of active TB in infected individuals and the subsequent spread of the disease. In this study, a systems biology approach was utilized to obtain numerous microarray data sets for mRNAs and microRNAs (miRNAs) expressed in the peripheral blood mononuclear cells (PBMCs) of TB patients and individuals with LTBI. Within these data sets, we identified the differentially expressed mRNAs and miRNAs and further investigated which differentially expressed genes and miRNAs were uniquely expressed during LTBI. The Database for Annotation, Visualization and Integrated Discovery (DAVID) was employed to analyze the functional annotations and pathway classifications of the identified genes. To further understand the unique miRNA-gene regulatory network of LTBI, we constructed a protein-protein interaction (PPI) network for the targeted genes. The PPI network included 39 genes that were differentially and uniquely expressed in PBMCs of individuals with LTBI, and KEGG pathway enrichment analysis showed that these genes were predominantly involved in the PI3K-Akt signaling pathway, which plays an important role in chronic inflammation. DIANA TOOLs-mirPath analysis revealed that the identified miRNAs in the miRNA-gene regulatory network for LTBI were mainly associated with the Hippo signaling pathway, which functions in the development of inflammation. Quantitative real-time PCR verified the up expression of hsa-miR-212-3p and its predicted target gene -MAPK1 which had low expression and was a major component of the PPI network, and MAPK1 expression was correlated with the clinicopathological characteristics of LTBI by receiver operating characteristic (ROC) curve analysis. Therefore, MAPK1 has potential to be a new investigable marker during LTBI, which merits our further study and solution. The unique aberrant miRNA-gene regulatory network and the related PPI network identified in this study provide insight into the molecular mechanisms of the immune response to LTBI, and thus, may aid in the development of a novel treatment strategy.
Collapse
Affiliation(s)
- Yan Lin
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, 130021, China
| | - Yuwei Zhang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, 130021, China
| | - Huiyuan Yu
- School of Public Health, Jilin University, Changchun, Jilin, 130021, China
| | - Ruonan Tian
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, 130021, China
| | - Guoqing Wang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, 130021, China; The Key Laboratory for Bionics Engineering, Ministry of Education, China, Jilin University, Changchun, Jilin, 130021, China
| | - Fan Li
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medicine, Jilin University, Changchun, Jilin, 130021, China; The Key Laboratory for Bionics Engineering, Ministry of Education, China, Jilin University, Changchun, Jilin, 130021, China; Engineering Research Center for Medical Biomaterials of Jilin Province, Jilin University, Changchun, Jilin, 130021, China; Key Laboratory for Biomedical Materials of Jilin Province, Jilin University, Changchun, Jilin, 130021, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang, China.
| |
Collapse
|
16
|
Bauer M, Coldewey SM, Leitner M, Löffler B, Weis S, Wetzker R. Deterioration of Organ Function As a Hallmark in Sepsis: The Cellular Perspective. Front Immunol 2018; 9:1460. [PMID: 29997622 PMCID: PMC6028602 DOI: 10.3389/fimmu.2018.01460] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/12/2018] [Indexed: 01/12/2023] Open
Abstract
Development of organ dysfunction discriminates sepsis from uncomplicated infection. The paradigm shift implicated by the new sepsis-3 definition holds that initial impairment of any organ can pave the way for multiple organ dysfunction and death. Moreover, the role of the systemic inflammatory response, central element in previous sepsis definitions, has been questioned. Most strikingly, a so far largely underestimated defense mechanism of the host, i.e., "disease tolerance," which aims at maintaining host vitality without reducing pathogen load, has gained increasing attention. Here, we summarize evidence that a dysregulation of critical cellular signaling events, also in non-immune cells, might provide a conceptual framework for sepsis-induced dysfunction of parenchymal organs in the absence of significant cell death. We suggest that key signaling mediators, such as phosphoinositide 3-kinase, mechanistic target of rapamycin, and AMP-activated protein kinase, control the balance of damage and repair processes and thus determine the fate of affected organs and ultimately the host. Therapeutic targeting of these multifunctional signaling mediators requires cell-, tissue-, or organ-specific approaches. These novel strategies might allow stopping the domino-like damage to further organ systems and offer alternatives beyond the currently available strictly supportive therapeutic options.
Collapse
Affiliation(s)
- Michael Bauer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Sina M Coldewey
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Margit Leitner
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Bettina Löffler
- Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Institute of Medical Microbiology, Jena University Hospital, Jena, Germany
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany.,Center for Infectious Disease and Infection Control, Jena University Hospital, Jena, Germany
| | - Reinhard Wetzker
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| |
Collapse
|
17
|
Halbgebauer R, Schmidt CQ, Karsten CM, Ignatius A, Huber-Lang M. Janus face of complement-driven neutrophil activation during sepsis. Semin Immunol 2018; 37:12-20. [PMID: 29454576 DOI: 10.1016/j.smim.2018.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 12/28/2022]
Abstract
During local and systemic inflammation, the complement system and neutrophil granulocytes are activated not only by pathogens, but also by released endogenous danger signals. It is recognized increasingly that complement-mediated neutrophil activation plays an ambivalent role in sepsis pathophysiology. According to the current definition, the onset of organ dysfunction is a hallmark of sepsis. The preceding organ damage can be caused by excessive complement activation and neutrophil actions against the host, resulting in bystander injury of healthy tissue. However, in contrast, persistent and overwhelming inflammation also leads to a reduction in neutrophil responsiveness as well as complement components and thus may render patients at enhanced risk of spreading infection. This review provides an overview on the molecular and cellular processes that link complement with the two-faced functional alterations of neutrophils in sepsis. Finally, we describe novel tools to modulate this interplay beneficially in order to improve outcome.
Collapse
Affiliation(s)
- R Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Hospital, Helmholtzstr. 8/1, 89081 Ulm, Germany.
| | - C Q Schmidt
- Institute of Pharmacology of Natural Products and Clinical Pharmacology, Ulm University, Helmholtzstr. 20, 89081 Ulm, Germany.
| | - C M Karsten
- Institute for Systemic Inflammation Research, University of Luebeck, Ratzeburger Allee 160, 23562 Luebeck, Germany.
| | - A Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Helmholtzstr. 14, 89081 Ulm, Germany.
| | - M Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, Ulm University Hospital, Helmholtzstr. 8/1, 89081 Ulm, Germany.
| |
Collapse
|
18
|
Chiang CK, Tworak A, Kevany BM, Xu B, Mayne J, Ning Z, Figeys D, Palczewski K. Quantitative phosphoproteomics reveals involvement of multiple signaling pathways in early phagocytosis by the retinal pigmented epithelium. J Biol Chem 2017; 292:19826-19839. [PMID: 28978645 PMCID: PMC5712622 DOI: 10.1074/jbc.m117.812677] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/22/2017] [Indexed: 12/12/2022] Open
Abstract
One of the major biological functions of the retinal pigmented epithelium (RPE) is the clearance of shed photoreceptor outer segments (POS) through a multistep process resembling phagocytosis. RPE phagocytosis helps maintain the viability of photoreceptors that otherwise could succumb to the high metabolic flux and photo-oxidative stress associated with visual processing. The regulatory mechanisms underlying phagocytosis in the RPE are not fully understood, although dysfunction of this process contributes to the pathogenesis of multiple human retinal degenerative disorders, including age-related macular degeneration. Here, we present an integrated transcriptomic, proteomic, and phosphoproteomic analysis of phagocytosing RPE cells, utilizing three different experimental models: the human-derived RPE-like cell line ARPE-19, cultured murine primary RPE cells, and RPE samples from live mice. Our combined results indicated that early stages of phagocytosis in the RPE are mainly characterized by pronounced changes in the protein phosphorylation level. Global phosphoprotein enrichment analysis revealed involvement of PI3K/Akt, mechanistic target of rapamycin (mTOR), and MEK/ERK pathways in the regulation of RPE phagocytosis, confirmed by immunoblot analyses and in vitro phagocytosis assays. Most strikingly, phagocytosis of POS by cultured RPE cells was almost completely blocked by pharmacological inhibition of phosphorylation of Akt. Our findings, along with those of previous studies, indicate that these phosphorylation events allow the RPE to integrate multiple signals instigated by shed POS at different stages of the phagocytic process.
Collapse
Affiliation(s)
- Cheng-Kang Chiang
- From the Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- the Department of Chemistry, National Dong Hwa University, No. 1 Sec. 2 Da Hsueh Road, Shoufeng, Hualien 97401, Taiwan
| | | | | | - Bo Xu
- From the Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Janice Mayne
- From the Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Zhibin Ning
- From the Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Daniel Figeys
- From the Ottawa Institute of Systems Biology and Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada,
- the Canadian Institute for Advanced Research, Toronto, Ontario M5G 1Z8, Canada
| | - Krzysztof Palczewski
- the Department of Pharmacology and
- the Cleveland Center for Membrane and Structural Biology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, and
| |
Collapse
|
19
|
Fattahi F, Kalbitz M, Malan EA, Abe E, Jajou L, Huber-Lang MS, Bosmann M, Russell MW, Zetoune FS, Ward PA. Complement-induced activation of MAPKs and Akt during sepsis: role in cardiac dysfunction. FASEB J 2017; 31:4129-4139. [PMID: 28572445 DOI: 10.1096/fj.201700140r] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/15/2017] [Indexed: 01/27/2023]
Abstract
Polymicrobial sepsis in mice causes myocardial dysfunction after generation of the complement anaphylatoxin, complement component 5a (C5a). C5a interacts with its receptors on cardiomyocytes (CMs), resulting in redox imbalance and cardiac dysfunction that can be functionally measured and quantitated using Doppler echocardiography. In this report we have evaluated activation of MAPKs and Akt in CMs exposed to C5a in vitro and after cecal ligation and puncture (CLP) in vivo In both cases, C5a in vitro caused activation (phosphorylation) of MAPKs and Akt in CMs, which required availability of both C5a receptors. Using immunofluorescence technology, activation of MAPKs and Akt occurred in left ventricular (LV) CMs, requiring both C5a receptors, C5aR1 and -2. Use of a water-soluble p38 inhibitor curtailed activation in vivo of MAPKs and Akt in LV CMs as well as the appearance of cytokines and histones in plasma from CLP mice. When mouse macrophages were exposed in vitro to LPS, activation of MAPKs and Akt also occurred. The copresence of the p38 inhibitor blocked these activation responses. Finally, the presence of the p38 inhibitor in CLP mice reduced the development of cardiac dysfunction. These data suggest that polymicrobial sepsis causes cardiac dysfunction that appears to be linked to activation of MAPKs and Akt in heart.-Fattahi, F., Kalbitz, M., Malan, E. A., Abe, E., Jajou, L., Huber-Lang, M. S., Bosmann, M., Russell, M. W., Zetoune, F. S., Ward, P. A. Complement-induced activation of MAPKs and Akt during sepsis: role in cardiac dysfunction.
Collapse
Affiliation(s)
- Fatemeh Fattahi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Miriam Kalbitz
- Department of Orthopaedic Trauma, Hand, Plastic, and Reconstructive Surgery, University Hospital of Ulm, Ulm, Germany
| | - Elizabeth A Malan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Elizabeth Abe
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lawrence Jajou
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Markus S Huber-Lang
- Department of Orthopaedic Trauma, Hand, Plastic, and Reconstructive Surgery, University Hospital of Ulm, Ulm, Germany
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis, University Medical Center, Mainz, Germany
| | - Mark W Russell
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Firas S Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA;
| |
Collapse
|
20
|
Xuan NT, Hoang NH, Nhung VP, Duong NT, Ha NH, Hai NV. Regulation of dendritic cell function by insulin/IGF-1/PI3K/Akt signaling through klotho expression. J Recept Signal Transduct Res 2016; 37:297-303. [PMID: 27808000 DOI: 10.1080/10799893.2016.1247862] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Insulin or insulin-like growth factor 1 (IGF-1) promotes the activation of phosphoinositide 3 kinase (PI3K)/Akt signaling in immune cells including dendritic cells (DCs), the most potent professional antigen-presenting cells for naive T cells. Klotho, an anti-aging protein, participates in the regulation of the PI3K/Akt signaling, thus the Ca2+-dependent migration is reduced in klotho-deficient DCs. The present study explored the effects of insulin/IGF-1 on DC function through klotho expression. To this end, the mouse bone marrow cells were isolated and cultured with GM-CSF to attain bone marrow-derived DCs (BMDCs). Cells were treated with insulin or IGF-1 and followed by stimulating with lipopolysaccharides (LPS). Tumor necrosis factor (TNF)-α formation was examined by enzyme-linked immunosorbent assay (ELISA). Phagocytosis was analyzed by FITC-dextran uptake assay. The expression of klotho was determined by quantitative PCR, immunoprecipitation and western blotting. As a result, treatment of the cells with insulin/IGF-1 resulted in reducing the klotho expression as well as LPS-stimulated TNF-α release and increasing the FITC-dextran uptake but unaltering reactive oxygen species (ROS) production in BMDCs. The effects were abolished by using pharmacological inhibition of PI3K/Akt with LY294002 and paralleled by transfecting DCs with klotho siRNA. In conclusion, the regulation of klotho sensitive DC function by IGF-1 or insulin is mediated through PI3K/Akt signaling pathway in BMDCs.
Collapse
Affiliation(s)
- Nguyen Thi Xuan
- a Institute of Genome Research, Vietnam Academy of Science and Technology , Cau Giay , Ha Noi , Vietnam
| | - Nguyen Huy Hoang
- a Institute of Genome Research, Vietnam Academy of Science and Technology , Cau Giay , Ha Noi , Vietnam
| | - Vu Phuong Nhung
- a Institute of Genome Research, Vietnam Academy of Science and Technology , Cau Giay , Ha Noi , Vietnam
| | - Nguyen Thuy Duong
- a Institute of Genome Research, Vietnam Academy of Science and Technology , Cau Giay , Ha Noi , Vietnam
| | - Nguyen Hai Ha
- a Institute of Genome Research, Vietnam Academy of Science and Technology , Cau Giay , Ha Noi , Vietnam
| | - Nong Van Hai
- a Institute of Genome Research, Vietnam Academy of Science and Technology , Cau Giay , Ha Noi , Vietnam
| |
Collapse
|
21
|
Morioka S, Nigorikawa K, Sasaki J, Hazeki K, Kasuu Y, Sasaki T, Hazeki O. Myeloid cell-specific inositol polyphosphate-4-phosphatase type I knockout mice impair bacteria clearance in a murine peritonitis model. Innate Immun 2016; 22:444-51. [DOI: 10.1177/1753425916652714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 05/10/2016] [Indexed: 11/17/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K)/Akt signaling has been implicated in the anti-inflammatory response in a mouse model of endotoxemia and sepsis. The present study focused on the role of inositol polyphosphate-4-phosphatase type I (Inpp4a), which dephosphorylates PtdIns(3,4)P2 to PtdIns(3)P, in bacterial infections. We prepared myeloid cell-specific Inpp4a-conditional knockout mice. Macrophages from these mice showed increased Akt phosphorylation and reduced production of inflammatory cytokines in response to LPS or Escherichia coli in vitro. The Inpp4a knockout mice survived for a shorter time than wild type mice after i.p. infection with E. coli, with less production of inflammatory cytokines. Additionally, E. coli clearance from blood and lung was significantly impaired in the knockout mice. A likely mechanism is that the Inpp4a-catalyzed dephosphorylation of PtdIns(3,4)P2 down-regulates Akt pathways, which, in turn, increases the production of inflammatory mediators. This mechanism at least fits the decreased E. coli clearance and short survival in the Inpp4a knockout mice.
Collapse
Affiliation(s)
- Shin Morioka
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Kiyomi Nigorikawa
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Junko Sasaki
- Department of Pathology and Immunology, Akita University School of Medicine, Akita 010-8543, Japan
| | - Kaoru Hazeki
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Yoshihiro Kasuu
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| | - Takehiko Sasaki
- Department of Pathology and Immunology, Akita University School of Medicine, Akita 010-8543, Japan
| | - Osamu Hazeki
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
22
|
Huang X, Dai Z, Cai L, Sun K, Cho J, Albertine KH, Malik AB, Schraufnagel DE, Zhao YY. Endothelial p110γPI3K Mediates Endothelial Regeneration and Vascular Repair After Inflammatory Vascular Injury. Circulation 2016; 133:1093-103. [PMID: 26839042 DOI: 10.1161/circulationaha.115.020918] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 01/29/2016] [Indexed: 01/11/2023]
Abstract
BACKGROUND The integrity of endothelial monolayer is a sine qua non for vascular homeostasis and maintenance of tissue-fluid balance. However, little is known about the signaling pathways regulating regeneration of the endothelial barrier after inflammatory vascular injury. METHODS AND RESULTS Using genetic and pharmacological approaches, we demonstrated that endothelial regeneration selectively requires activation of p110γPI3K signaling, which thereby mediates the expression of the endothelial reparative transcription factor Forkhead box M1 (FoxM1). We observed that FoxM1 induction in the pulmonary vasculature was inhibited in mice treated with a p110γ-selective inhibitor and in Pik3cg(-/-) mice after lipopolysaccharide challenge. Pik3cg(-/-) mice exhibited persistent lung inflammation induced by sepsis and sustained increase in vascular permeability. Restoration of expression of either p110γ or FoxM1 in pulmonary endothelial cells of Pik3cg(-/-) mice restored endothelial regeneration and normalized the defective vascular repair program. We also observed diminished expression of p110γ in pulmonary vascular endothelial cells of patients with acute respiratory distress syndrome, suggesting that impaired p110γ-FoxM1 vascular repair signaling pathway is a critical factor in persistent leaky lung microvessels and edema formation in the disease. CONCLUSIONS We identify p110γ as the critical mediator of endothelial regeneration and vascular repair after sepsis-induced inflammatory injury. Thus, activation of p110γ-FoxM1 endothelial regeneration may represent a novel strategy for the treatment of inflammatory vascular diseases.
Collapse
Affiliation(s)
- Xiaojia Huang
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Zhiyu Dai
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Lei Cai
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Kai Sun
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Jaehyung Cho
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Kurt H Albertine
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Asrar B Malik
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - Dean E Schraufnagel
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.)
| | - You-Yang Zhao
- From Department of Pharmacology (X.H., Z.D., L.C., K.S., J.C., A.B.M., Y.-Y.Z.), Center for Lung and Vascular Biology (X.H., Z.D., L.C., K.S., A.B.M., Y.-Y.Z.), Department of Medicine (D.E.S.), University of Illinois College of Medicine, Chicago; and Departments of Pediatrics and Medicine, University of Utah School of Medicine, Salt Lake City (K.H.A.).
| |
Collapse
|
23
|
Okeke EB, Uzonna JE. In Search of a Cure for Sepsis: Taming the Monster in Critical Care Medicine. J Innate Immun 2016; 8:156-70. [PMID: 26771196 DOI: 10.1159/000442469] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022] Open
Abstract
In spite of over half a century of research, sepsis still constitutes a major problem in health care delivery. Although advances in research have significantly increased our knowledge of the pathogenesis of sepsis and resulted in better prognosis and improved survival outcome, sepsis still remains a major challenge in modern medicine with an increase in occurrence predicted and a huge socioeconomic burden. It is generally accepted that sepsis is due to an initial hyperinflammatory response. However, numerous efforts aimed at targeting the proinflammatory cytokine network have been largely unsuccessful and the search for novel potential therapeutic targets continues. Recent studies provide compelling evidence that dysregulated anti-inflammatory responses may also contribute to sepsis mortality. Our previous studies on the role of regulatory T cells and phosphoinositide 3-kinases in sepsis highlight immunological approaches that could be explored for sepsis therapy. In this article, we review the current and emerging concepts in sepsis, highlight novel potential therapeutic targets and immunological approaches for sepsis treatment and propose a biphasic treatment approach for management of the condition.
Collapse
Affiliation(s)
- Emeka B Okeke
- Department of Immunology, Faculty of Medicine, University of Manitoba, Winnipeg, Man., Canada
| | | |
Collapse
|
24
|
Zhou H, Gao S, Duan X, Liang S, Scott DA, Lamont RJ, Wang H. Inhibition of serum- and glucocorticoid-inducible kinase 1 enhances TLR-mediated inflammation and promotes endotoxin-driven organ failure. FASEB J 2015; 29:3737-49. [PMID: 25993992 DOI: 10.1096/fj.15-270462] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 05/11/2015] [Indexed: 12/12/2022]
Abstract
Serum- and glucocorticoid-regulated kinase (SGK)1 is associated with several important pathologic conditions and plays a modulatory role in adaptive immune responses. However, the involvement and functional role of SGK1 in innate immune responses remain entirely unknown. In this study, we establish that SGK1 is a novel and potent negative regulator of TLR-induced inflammation. Pharmacologic inhibition of SGK1 or suppression by small interfering RNA enhances proinflammatory cytokine (TNF, IL-12, and IL-6) production in TLR-engaged monocytes, a result confirmed in Cre-loxP-mediated SGK1-deficient cells. SGK1 inhibition or gene deficiency results in increased phosphorylation of IKK, IκBα, and NF-κB p65 in LPS-stimulated cells. Enhanced NF-κB p65 DNA binding also occurs upon SGK1 inhibition. The subsequent enhancement of proinflammatory cytokines is dependent on the phosphorylation of TGF-β-activated kinase 1 (TAK1), as confirmed by TAK1 gene silencing. In vivo relevance was established in a murine endotoxin model, in which we found that SGK1 inhibition aggravates the severity of multiple organ damage and enhances the inflammatory response by heightening both proinflammatory cytokine levels and neutrophil infiltration. These findings have identified an anti-inflammatory function of SGK1, elucidated the underlying intracellular mechanisms, and establish, for the first time, that SGK1 holds potential as a novel target for intervention in the control of inflammatory diseases.
Collapse
Affiliation(s)
- Huaxin Zhou
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shegan Gao
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Xiaoxian Duan
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Shuang Liang
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - David A Scott
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Richard J Lamont
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Huizhi Wang
- *Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA; Department of Oncology, Cancer Institute, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China; and Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
25
|
Liu J, Tan Y, Zhang J, Zou L, Deng G, Xu X, Wang F, Ma Z, Zhang J, Zhao T, Liu Y, Li Y, Zhu B, Guo B. C5aR, TNF-α, and FGL2 contribute to coagulation and complement activation in virus-induced fulminant hepatitis. J Hepatol 2015; 62:354-62. [PMID: 25200905 DOI: 10.1016/j.jhep.2014.08.050] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Revised: 08/29/2014] [Accepted: 08/31/2014] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Viral fulminant hepatitis (FH) is a disease with a high mortality rate. Activation of the complement system correlates with the development of FH. However, the key factors mediating complement activation in FH remain elusive. METHODS Liver tissues were isolated from FH patients infected by hepatitis B virus (HBV) and from mice infected with murine hepatitis virus strain 3 (MHV-3). Wild type mice were treated with or without antagonists of C5aR or TNF-α, and mice deficient for C5aR (C5aR(-/-)), Fgl2 (Fgl2(-/-)), and Tnfα (Tnfα(-/-)) mice were not treated with the antagonists. C5b-9, C5aR, FGL2, CD31, CD11b, fibrin, TNF-α, and complement C3 cleavage products were detected by immunohistochemistry, immunofluorescence, or ELISA. Sorted liver sinusoidal endothelial cells (LSECs) or myeloid-derived (CD11b(+)) cells were stimulated with C5a, TNF-α or MHV-3 in vitro. The mRNA expressions levels of Fgl2 and Tnfα were determined by qRT-PCR analyses. RESULTS We observed that complement activation, coagulation and pro-inflammatory cytokine production were upregulated in the HBV(+) patients with FH. Similar observations were made in the murine FH models. Complement activation and coagulation were significantly reduced in MHV-3 infected mice in the absence of C5aR, Tnfα or Fgl2. The MHV-3 infected C5aR(-/-) mice exhibited reduced numbers of infiltrated inflammatory CD11b(+) cells and a reduced expression of TNF-α and FGL2. Moreover, C5a administration stimulated TNF-α production by CD11b(+) cells, which in turn promoted the expression of FGL2 in CD31(+) LSEC-like cells in vitro. Administration of antagonists against C5aR or TNF-α ameliorated MHV-3-induced FH. CONCLUSIONS Our results demonstrate that C5aR, TNF-α, and FGL2 form an integral network that contributes to coagulation and complement activation, and suggest that those are potential therapeutic targets in viral FH intervention.
Collapse
Affiliation(s)
- Jianjun Liu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Histology & Embryology, Third Military Medical University, Chongqing, China
| | - Yulong Tan
- Department of Immunology, Third Military Medical University, Chongqing, China
| | - Jinyu Zhang
- Department of Immunology, Third Military Medical University, Chongqing, China
| | - Liyun Zou
- Department of Immunology, Third Military Medical University, Chongqing, China
| | - Guohong Deng
- Institute of Infectious Diseases, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xueqing Xu
- Department of Medical Genetics, Third Military Medical University, Chongqing, China
| | - Feng Wang
- Department of Laboratory Medicine, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhengwei Ma
- Institute of Hepatobiliary Surgery & Southwest Hospital, Third Military Medical University, District Shapingba, Chongqing, China
| | - Jue Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Tingting Zhao
- Department of Immunology, Third Military Medical University, Chongqing, China
| | - Yunlai Liu
- Department of Histology & Embryology, Third Military Medical University, Chongqing, China
| | - Yongsheng Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| | - Bo Guo
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Immunology, Third Military Medical University, Chongqing, China.
| |
Collapse
|
26
|
Complement mediates a primed inflammatory response after traumatic lung injury. J Trauma Acute Care Surg 2014; 76:601-8; discussion 608-9. [PMID: 24553525 DOI: 10.1097/ta.0000000000000129] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Pulmonary contusion (PC) is a common, potentially lethal injury that results in the priming for exaggerated responses to subsequent immune challenge such as an infection (second hit). We hypothesize a PC-induced complement (C) activation participates in the priming effect for a second hit. METHODS Male, 8 weeks to 9 weeks, C57BL/6 mice (wild-type, C5) underwent blunt chest trauma resulting in PC. At 3 hours/24 hours after injury, the inflammatory response was measured in tissue, serum, and bronchoalveolar lavage (BAL). The thrombin inhibitor, hirudin, was used to determine if injury-induced thrombin participated in the activation of C. Injury-primed responses were tested by challenging injured mice with bacterial endotoxin (lipopolysaccharide, LPS) as a second hit. Inflammatory responses were assessed at 4 hours after LPS challenge. Data were analyzed using one-way analysis of variance with Bonferroni multiple comparison posttest (significance, p ≤ 0.05). Protocols were approved by the Institutional Animal Care and Use Committee. RESULTS We found significantly increased levels of C5a in the BAL of injured animals as early as 24 hours, persisting for up to 72 hours after injury. Hirudin-treated injured mice had significantly decreased levels of thrombin in the BAL that correlated with reduced C5a levels. Injured mice challenged with intratracheal (IT) LPS had increased C5a and inflammatory response. Conversely, inhibition of C5a or its receptor, C5aR, before LPS challenge correlated with decreased inflammatory responses; C5a-deficient mice showed a similar loss of primed response to LPS challenge. CONCLUSION Complement C5a levels in the BAL are increased over several days after PC. Premorbid inhibition of thrombin markedly decreases C5a levels after PC, suggesting that thrombin-induced C activation is the major pathway of activation after PC. Similarly, inhibition of C5a after PC will decrease injury-primed responses to LPS stimulation. Our findings suggest cross-talk between the coagulation and complement systems that induce immune priming after PC.
Collapse
|
27
|
Liu G, Bi Y, Wang R, Shen B, Zhang Y, Yang H, Wang X, Liu H, Lu Y, Han F. Kinase AKT1 negatively controls neutrophil recruitment and function in mice. THE JOURNAL OF IMMUNOLOGY 2013; 191:2680-90. [PMID: 23904165 DOI: 10.4049/jimmunol.1300736] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Neutrophils are critically involved in host defense and inflammatory injury. However, intrinsic signaling mechanisms controlling neutrophil recruitment and activities are poorly defined. In this article, we showed that protein kinase AKT1 (also known as PKBα) is the dominant isoform expressed in neutrophils and is downregulated upon bacterial infection and neutrophil activation. AKT1 deficiency resulted in severe disease progression accompanied by recruitment of neutrophils and enhanced bactericidal activity in the acute inflammatory lung injury (ALI) and the Staphylococcus aureus infection mouse models. Moreover, the depletion of neutrophils efficiently reversed the aggravated inflammatory response, but adoptive transfer of AKT1(-/-) neutrophils could potentiate the inflammatory immunity, indicating an intrinsic effect of the neutrophil in modulating inflammation in AKT1(-/-) mice. In the ALI model, the infiltration of neutrophils into the inflammatory site was associated with enhanced migration capacity, whereas inflammatory stimuli could promote neutrophil apoptosis. In accordance with these findings, neutralization of CXCR2 attenuated neutrophil infiltration and delayed the occurrence of inflammation. Finally, the enhanced bactericidal activity and inflammatory immunity of AKT-deficient neutrophils were mediated by a STAT1-dependent, but not a mammalian target of rapamycin-dependent, pathway. Thus, our findings indicated that the AKT1-STAT1 signaling axis negatively regulates neutrophil recruitment and activation in ALI and S. aureus infection in mice.
Collapse
Affiliation(s)
- Guangwei Liu
- Department of Immunology, Shanghai Medical College, Fudan University, Shanghai 200023, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Adiponectin inhibits neutrophil phagocytosis of Escherichia coli by inhibition of PKB and ERK 1/2 MAPK signalling and Mac-1 activation. PLoS One 2013; 8:e69108. [PMID: 23935932 PMCID: PMC3723777 DOI: 10.1371/journal.pone.0069108] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 06/06/2013] [Indexed: 11/24/2022] Open
Abstract
Full length adiponectin is a potent immune modulatory adipokine, impacting upon the actions of several immune cells. Neutrophil oxidative burst has been shown to decrease in response to adiponectin, and we speculated that it could have other effects on neutrophil function. Here we report that adiponectin reduces the phagocytic ability of human neutrophils, decreasing significantly the ingestion of opsonised E. coli by these cells in whole blood (p<0.05) and as isolated neutrophils (p<0.05). We then determined the mechanisms involved. We observed that the activation of Mac-1, the receptor engaged in complement-mediated phagocytosis, was decreased by adiponectin in response to E. coli stimulation. Moreover, treatment of neutrophils with adiponectin prior to incubation with E. coli significantly inhibited signalling through the PI3K/PKB and ERK 1/2 pathways, with a parallel reduction of F-actin content. Studies with pharmacological inhibitors showed that inhibition of PI3K/PKB, but not ERK 1/2 signalling was able to prevent the activation of Mac-1. In conclusion, we propose that adiponectin negatively affects neutrophil phagocytosis, reducing the uptake of E. coli and inhibiting Mac-1 activation, the latter by blockade of the PI3K/PKB signal pathway.
Collapse
|
29
|
Pio R, Ajona D, Lambris JD. Complement inhibition in cancer therapy. Semin Immunol 2013; 25:54-64. [PMID: 23706991 DOI: 10.1016/j.smim.2013.04.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 04/13/2013] [Indexed: 02/08/2023]
Abstract
For decades, complement has been recognized as an effector arm of the immune system that contributes to the destruction of tumor cells. In fact, many therapeutic strategies have been proposed that are based on the intensification of complement-mediated responses against tumors. However, recent studies have challenged this paradigm by demonstrating a tumor-promoting role for complement. Cancer cells seem to be able to establish a convenient balance between complement activation and inhibition, taking advantage of complement initiation without suffering its deleterious effects. Complement activation may support chronic inflammation, promote an immunosuppressive microenvironment, induce angiogenesis, and activate cancer-related signaling pathways. In this context, inhibition of complement activation would be a therapeutic option for treating cancer. This concept is relatively new and deserves closer attention. In this article, we summarize the mechanisms of complement activation on cancer cells, the cancer-promoting effect of complement initiation, and the rationale behind the use of complement inhibition as a therapeutic strategy against cancer.
Collapse
Affiliation(s)
- Ruben Pio
- Oncology Division, Center for Applied Medical Research-CIMA, Pamplona, Spain. rpio.@unav.es
| | | | | |
Collapse
|
30
|
Prince A. Innate Immune Responses in Ventilator-Associated Pneumonia. MUCOSAL IMMUNOLOGY OF ACUTE BACTERIAL PNEUMONIA 2013. [PMCID: PMC7121904 DOI: 10.1007/978-1-4614-5326-0_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Ventilator-associated pneumonia (VAP) is a common complication of mechanical ventilation, resulting in substantial morbidity, mortality, and health care cost. Early upper airway colonization by pathogenic bacteria and microaspiration are the primary pathogenic events leading to VAP. Patients at risk for VAP have defects in structural/mechanical defenses of the respiratory tract. In addition, critical illness, including sepsis, trauma, and postoperative states, is associated with profound defects in both innate and acquired antibacterial immunity, influencing antimicrobial effector functions of both leukocytes and structural/parenchymal cells. Factors present within the lung microenvironment, including alveolar stretch, cyclical atelectasis, changes in oxygen tension, and respiratory tract microbiota, substantially impact antibacterial host responses. Mechanisms accounting for dysregulated immune homeostasis are incompletely understood, but likely involve: (1) alterations in the balance of pro- and anti-inflammatory cytokines; (2) changes in pathogen recognition receptor and G-protein coupled receptor expression and downstream signaling cascades; and (3) dysregulated cell death responses. Antibiotics and preventive strategies are the mainstay of therapy in patients with VAP. However, novel approaches are needed to reverse immunological reprogramming that occurs during critical illness and/or mechanical ventilation, and to identify patients who are most likely to benefit from immunomodulatory therapy.
Collapse
|
31
|
Abstract
Multiorgan failure (MOF) represents the leading cause of death in patients with sepsis and systemic inflammatory response syndrome (SIRS) following severe trauma. The underlying immune response is highly complex and involves activation of the complement system as a crucial entity of innate immunity. Uncontrolled activation of the complement system during sepsis and SIRS with in excessive generation of complement activation products contributes to an ensuing dysfunction of various organ systems. In the present review, mechanisms of the inflammatory response in the development of MOF in sepsis and SIRS with particular focus on the complement system are discussed.
Collapse
|
32
|
Yan C, Gao H. New insights for C5a and C5a receptors in sepsis. Front Immunol 2012; 3:368. [PMID: 23233853 PMCID: PMC3518060 DOI: 10.3389/fimmu.2012.00368] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 11/19/2012] [Indexed: 11/24/2022] Open
Abstract
The complement system plays a central role in inflammation and immunity. Among the complement activation products, C5a is one of the most potent inflammatory peptides with a broad spectrum of functions. There is strong evidence for complement activation including elevated plasma level of C5a in humans and animals with sepsis. C5a exerts its effects through the C5a receptors. Of the two receptors that bind C5a, the C5aR (CD88) is known to mediate signaling activity, whereas the function of another C5a binding receptor, C5L2, remains largely unknown. Here, we review the critical role of C5a in sepsis and summarize evidence indicating that both C5aR and C5L2 act as regulating receptors for C5a during sepsis.
Collapse
Affiliation(s)
| | - Hongwei Gao
- Department of Anesthesiology, Perioperative and Pain Medicine, Center for Experimental Therapeutics and Reperfusion Injury, Brigham and Women's Hospital, Harvard Medical School, Harvard Institutes of MedicineBoston, MA, USA
| |
Collapse
|
33
|
Abstract
Complement activation usually results in the formation of complement fragment 5a (C5a) that interacts with its two receptors, C5aR and C5L2. These receptors belong to the rhodopsin family of G protein-coupled seven transmembrane-containing receptors. C5aR and C5L2 are expressed on/in a wide variety of cells and tissues. Interaction of C5a with C5aR leads to many pleiotropic effects, including the release of cytokines and chemokines and recruitment of inflammatory cells. In certain circumstances, C5a-C5aR interactions can also result in pathophysiological changes as seen in sepsis, rheumatoid arthritis, asthma, acute lung injury and ischemia-reperfusion injury. This overview of the C5a-C5aR interactions describes how such interactions facilitate the pivotal role the complement system plays in the host's innate and adaptive responses.
Collapse
Affiliation(s)
- J Vidya Sarma
- University of Michigan Medical School, Department of Pathology, Ann Arbor, MI, USA
| | | |
Collapse
|
34
|
p38MAPK, ERK and PI3K signaling pathways are involved in C5a-primed neutrophils for ANCA-mediated activation. PLoS One 2012; 7:e38317. [PMID: 22675451 PMCID: PMC3365028 DOI: 10.1371/journal.pone.0038317] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/08/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The complement system is one of the important contributing factors in the development of antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV). C5a and the neutrophil C5a receptor play a central role in antineutrophil cytoplasmic antibody (ANCA)-mediated neutrophil recruitment and activation. The current study further investigated the signaling pathways of C5a-mediated priming of human neutrophils for ANCA-induced neutrophil activation. METHODOLOGY/PRINCIPAL FINDINGS The effects of the p38 mitogen-activated protein kinase (p38MAPK) inhibitor (SB202190), extracellular signal-regulated kinase (ERK) inhibitor (PD98059), c-Jun N-terminal kinase (JNK) inhibitor (6o) and phosphoinositol 3-kinase (PI3K) inhibitor (LY294002) were tested on respiratory burst and degranulation of C5a-primed neutrophils activated with ANCA, as well as on C5a-induced increase in expression of membrane-bound PR3 (mPR3) on neutrophils. For C5a-primed neutrophils for MPO-ANCA-induced respiratory burst, the mean fluorescence intensity (MFI) value was 254.8±67.1, which decreased to 203.6±60.3, 204.4±36.7, 202.4±49.9 and 188±47.9 upon pre-incubation with SB202190, PD98059, LY294002 and the mixture of above-mentioned three inhibitors (compared with that without inhibitors, P<0.01, P<0.05, P<0.01 and P<0.05), respectively. For PR3-ANCA-positive IgG, the MFI value increased in C5a-primed neutrophils, which decreased upon pre-incubation with above-mentioned inhibitors. The lactoferrin concentration increased in C5a-primed neutrophils induced by MPO or PR3-ANCA-positive IgG supernatant and decreased upon pre-incubation with above-mentioned three inhibitors. mPR3 expression increased from 923.3±182.4 in untreated cells to 1278.3±299.3 after C5a treatment and decreased to 1069.9±188.9, 1100±238.2, 1092.3±231.8 and 1053.9±200.3 by SB202190, PD98059, LY294002 and the mixture of above-mentioned three inhibitors (compared with that without inhibitors, P<0.01, P<0.05, P<0.01 and P<0.01), respectively. CONCLUSIONS/SIGNIFICANCE Activation of p38MAPK, ERK and PI3K are important steps in the translocation of ANCA antigens and C5a-induced activation of neutrophils by ANCA.
Collapse
|
35
|
Fortin CF, Cloutier A, Ear T, Sylvain-Prévost S, Mayer TZ, Bouchelaghem R, McDonald PP. A class IA PI3K controls inflammatory cytokine production in human neutrophils. Eur J Immunol 2011; 41:1709-19. [DOI: 10.1002/eji.201040945] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 02/15/2011] [Accepted: 03/11/2011] [Indexed: 12/30/2022]
|
36
|
Abstract
Phosphoinositide-3 kinases (PI3Ks) are critical regulatory proteins in the immunologic defense system against sepsis. The PI3K mechanism helps modulate cellular survival, innate and adaptive immunities, inflammation, nuclear factor-κB transcription, and may, in turn, play a protective role in sepsis. Animal studies confirm its role in the prevention of organ dysfunction and improvement of survival outcomes. Further advances in the understanding of this key immunomodulatory pathway may provide valuable insights into the manipulation of cellular function for therapeutic treatment of sepsis and other inflammatory diseases.
Collapse
|
37
|
C5a-mediated neutrophil dysfunction is RhoA-dependent and predicts infection in critically ill patients. Blood 2011; 117:5178-88. [PMID: 21292772 DOI: 10.1182/blood-2010-08-304667] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Critically ill patients are at heightened risk for nosocomial infections. The anaphylatoxin C5a impairs phagocytosis by neutrophils. However, the mechanisms by which this occurs and the relevance for acquisition of nosocomial infection remain undetermined. We aimed to characterize mechanisms by which C5a inhibits phagocytosis in vitro and in critically ill patients, and to define the relationship between C5a-mediated dysfunction and acquisition of nosocomial infection. In healthy human neutrophils, C5a significantly inhibited RhoA activation, preventing actin polymerization and phagocytosis. RhoA inhibition was mediated by PI3Kδ. The effects on RhoA, actin, and phagocytosis were fully reversed by GM-CSF. Parallel observations were made in neutrophils from critically ill patients, that is, impaired phagocytosis was associated with inhibition of RhoA and actin polymerization, and reversed by GM-CSF. Among a cohort of 60 critically ill patients, C5a-mediated neutrophil dysfunction (as determined by reduced CD88 expression) was a strong predictor for subsequent acquisition of nosocomial infection (relative risk, 5.8; 95% confidence interval, 1.5-22; P = .0007), and remained independent of time effects as assessed by survival analysis (hazard ratio, 5.0; 95% confidence interval, 1.3-8.3; P = .01). In conclusion, this study provides new insight into the mechanisms underlying immunocompromise in critical illness and suggests novel avenues for therapy and prevention of nosocomial infection.
Collapse
|
38
|
Dahlke K, Wrann CD, Sommerfeld O, Sossdorf M, Recknagel P, Sachse S, Winter SW, Klos A, Stahl GL, Ma YX, Claus RA, Reinhart K, Bauer M, Riedemann NC. Distinct different contributions of the alternative and classical complement activation pathway for the innate host response during sepsis. THE JOURNAL OF IMMUNOLOGY 2011; 186:3066-75. [PMID: 21263075 DOI: 10.4049/jimmunol.1002741] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Complement activation represents a crucial innate defense mechanism to invading microorganisms, but there is an eminent lack of understanding of the separate contribution of the different complement activation pathways to the host response during sepsis. We therefore investigated different innate host immune responses during cecal ligation and puncture (CLP)-induced sepsis in mice lacking either the alternative (fD(-/-)) or classical (C1q(-/-)) complement activation pathway. Both knockout mice strains showed a significantly reduced survival and increased organ dysfunction when compared with control mice. Surprisingly, fD(-/-) mice demonstrated a compensated bacterial clearance capacity as control mice at 6 h post CLP, whereas C1q(-/-) mice were already overwhelmed by bacterial growth at this time point. Interestingly, at 24 h after CLP, fD(-/-) mice failed to clear bacteria in a way comparable to control mice. However, both knockout mice strains showed compromised C3 cleavage during sepsis. Investigating potential causes for this discrepancy, we were able to demonstrate that despite normal bacterial clearance capacity early during the onset of sepsis, fD(-/-) mice displayed increased inflammatory cytokine generation and neutrophil recruitment into lungs and blood when compared with both control- and C1q(-/-) mice, indicating a potential loss of control over these immune responses. Further in vitro experiments revealed a strongly increased Nf-κB activation capacity in isolated neutrophils from fD(-/-) mice, supporting this hypothesis. Our results provide evidence for the new concept that the alternative complement activation pathway exerts a distinctly different contribution to the innate host response during sepsis when compared with the classical pathway.
Collapse
Affiliation(s)
- Katja Dahlke
- Department of Anesthesiology and Intensive Care Therapy, Jena University Hospital, Friedrich Schiller University Jena, 07747 Jena, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Cohen G, Raupachova J, Ilic D, Werzowa J, Hörl WH. Effect of leptin on polymorphonuclear leucocyte functions in healthy subjects and haemodialysis patients. Nephrol Dial Transplant 2011; 26:2271-81. [PMID: 21216885 DOI: 10.1093/ndt/gfq731] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Dysfunction of polymorphonuclear leucocytes (PMNLs) in end-stage renal disease (ESRD) patients contributes to a diminished immune defence. The serum levels of leptin are elevated in patients with ESRD. We analysed in vitro effects of leptin on PMNLs from healthy subjects (HS; n = 12) and haemodialysis (HD) patients (n = 15) before and after HD. METHODS PMNL oxidative burst and phagocytosis were tested by flow cytometry in whole blood. Chemotaxis of isolated PMNLs was assessed by the under-agarose method. To assess the involvement of leptin in PMNL signalling pathways, signal transduction inhibitors were used and the activity of intracellular kinases was investigated by western blotting, in vitro kinase assays and the Luminex technology. RESULTS Increasing the leptin level in the blood of HS leads to a reduced activation of the oxidative burst by Escherichia coli and phorbol 12-myristate 13-acetate. Activation of the oxidative burst is reduced in the blood of HD patients and the addition of leptin does not lead to further PMNL inhibition. Leptin at a concentration measured in HD patients significantly reduces the chemotaxis of PMNLs from HS but had no effect on PMNLs from ESRD patients before and also after HD treatment with high-flux dialysers. The phosphoinositide 3-kinase/Akt pathway is involved in the inhibitory effects of leptin. CONCLUSIONS In the presence of leptin, PMNLs from HS and HD patients respond differently to stimuli. The lack of response to leptin in PMNLs from HD patients cannot be influenced by HD.
Collapse
Affiliation(s)
- Gerald Cohen
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Cincinnati, Cincinnati, OH, USA.
| | | | | | | | | |
Collapse
|
40
|
Mitroulis I, Kourtzelis I, Kambas K, Rafail S, Chrysanthopoulou A, Speletas M, Ritis K. Regulation of the autophagic machinery in human neutrophils. Eur J Immunol 2010; 40:1461-72. [PMID: 20162553 DOI: 10.1002/eji.200940025] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The induction of the autophagy machinery, a process for the catabolism of cytosolic proteins and organelles, constitutes a crucial mechanism in innate immunity. However, the involvement of autophagy in human neutrophils and the possible inducers of this process have not been completely elucidated. In this study, the induction of autophagy was examined in human neutrophils treated with various activators and detected by the formation of acidified autophagosomes through monodansylcadaverine staining and via LC-3B conversion screened by immunoblotting and immunofluorescence confocal microscopy. In addition, the expression of the ATG genes was assessed by real-time RT-PCR. We provide evidence that autophagy is implicated in human neutrophils in both a phagocytosis-independent (rapamycin, TLR agonists, PMA) and phagocytosis (Escherichia coli)-dependent initiation manner. ROS activation is a positive mechanism for autophagy induction in the case of PMA, TLR activation and phagocytosis. Furthermore, LC3B gene expression was uniformly upregulated, indicating a transcriptional level of regulation for the autophagic machinery. This study provides a stepping stone toward further investigation of autophagy in neutrophil-driven inflammatory disorders.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- First Department of Internal Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | | | | | | | | |
Collapse
|
41
|
He P. Leucocyte/endothelium interactions and microvessel permeability: coupled or uncoupled? Cardiovasc Res 2010; 87:281-90. [PMID: 20472564 DOI: 10.1093/cvr/cvq140] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In response to infections or tissue injury, circulating leucocytes adhere to and migrate from the vessel lumen to interstitial inflammatory sites to combat invading pathogens. However, these defensive actions may also cause host tissue injury and microvascular dysfunction through oxidative bursts or enzyme release. For decades, the interaction between leucocytes and microvessel walls has been considered as a critical event leading to organ dysfunction. Extensive investigations have therefore focused on blocking specific adhesive ligands to prevent tissue injury. However, anti-adhesion therapies have shown limited success in preventing vascular dysfunction in clinical trials. Numerous studies have demonstrated temporal and spatial dissociations of leucocyte adhesion and/or emigration from permeability increases. The mechanisms that initiate the adhesion cascade have been found to be distinct from those that trigger the leucocyte oxidative burst responsible for increasing microvessel permeability. Recent studies demonstrated that endothelial activation by inflammatory mediators is critical for initiating platelet adhesion and platelet-dependent leucocyte recruitment resulting in augmented increases in microvessel permeability. These new developments suggest that targeting endothelial activation via directly enhancing endothelial barrier function might be a more efficient strategy than focusing on anti-adhesion or platelet/leucocyte depletion to prevent vascular damage during inflammation. Owing to space limitations and the wide range of studies in the field, this article will not serve as a comprehensive review. Instead, it will highlight the emerging evidence of adhesion-uncoupled permeability changes and establish a basis for re-evaluating the coupled relationship between leucocyte/platelet activation and microvessel permeability to achieve a better understanding of permeability regulation during inflammation.
Collapse
Affiliation(s)
- Pingnian He
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV 26506-9229, USA.
| |
Collapse
|
42
|
Reutershan J, Saprito MS, Wu D, Rückle T, Ley K. Phosphoinositide 3-kinase gamma required for lipopolysaccharide-induced transepithelial neutrophil trafficking in the lung. Eur Respir J 2009; 35:1137-47. [PMID: 19797129 DOI: 10.1183/09031936.00085509] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phosphoinositide 3-kinase gamma(PI3Kgamma) is a critical mediator of directional cell movement. Here, we sought to characterise the role of PI3Kgamma in mediating the different steps of polymorphonuclear leukocyte (PMN) trafficking in the lung. In a murine model of lipopolysaccharide (LPS)-induced lung injury, PMN migration into the different lung compartments was determined in PI3Kgamma gene-deficient (PI3Kgamma(-/-)) and wild-type mice. Bone marrow chimeras were created to characterise the role of PI3Kgamma on haematopoietic versus nonhaematopoietic cells. A small-molecule PI3Kgamma inhibitor was tested in vitro and in vivo. PMN adhesion to the pulmonary endothelium and transendothelial migration into the lung interstitium was enhanced in PI3Kgamma(-/-) mice. However, transepithelial migration into the alveolar space was reduced in these mice. When irradiated PI3Kgamma(-/-) mice were reconstituted with bone marrow from wild-type mice, migratory activity into the alveolar space was restored partially. A small-molecule PI3Kgamma inhibitor reduced chemokine-induced PMN migration in vitro when PMNs or epithelial cells, but not when endothelial cells, were treated. The inhibitor also reduced LPS-induced PMN migration in vivo. We conclude that PI3Kgamma is required for transepithelial but not for transendothelial migration in LPS-induced lung injury. Inhibition of PI3Kgamma activity may be effective at curbing excessive PMN infiltration in lung injury.
Collapse
Affiliation(s)
- J Reutershan
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
| | | | | | | | | |
Collapse
|
43
|
Shin OS, Miller LS, Modlin RL, Akira S, Uematsu S, Hu LT. Downstream signals for MyD88-mediated phagocytosis of Borrelia burgdorferi can be initiated by TRIF and are dependent on PI3K. THE JOURNAL OF IMMUNOLOGY 2009; 183:491-8. [PMID: 19542460 DOI: 10.4049/jimmunol.0900724] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We previously have shown that MyD88 is important for uptake of Borrelia burgdorferi by bone marrow derived macrophages (BMDMs). The mechanism by which MyD88 is involved in uptake of B. burgdorferi is currently is not well characterized. Here, we report that MyD88-mediated defect in the phagocytosis of B. burgdorferi can be complemented by TLR3/Toll/IL-1R domain-containing adaptor-inducing IFN-beta (TRIF) activation in BMDMs from MyD88(-/-) mice. This effect of TLR3/TRIF activation was not due to its induction of type I IFNs, suggesting instead a convergence of signaling pathways downstream of MyD88 and TRIF. To characterize signaling pathways involved in MyD88-mediated phagocytosis of B. burgdorferi, BMDMs were treated with specific inhibitors of MAPK, protein kinase C, JAK/STAT, or PI3K. Only inhibition of PI3K resulted in a significant decrease of B. burgdorferi uptake. Consistent with this, B. burgdorferi activation of MyD88 or TLR3/TRIF signaling resulted in increased activity of PI3K. Additionally, association of B. burgdorferi with actin-related protein (Arp2/3) complexes, which facilitate actin rearrangements during phagocytosis, was similarly reduced in MyD88(-/-) BMDMs and in BMDMs treated with a PI3K inhibitor. Taken together, these findings define an essential pathway whereby downstream signals from MyD88 or TRIF converge on PI3K, which triggers actin polymerization to initiate the phagocytosis of B. burgdorferi.
Collapse
Affiliation(s)
- Ok S Shin
- Department of Pathology/Immunology, Tufts University, Boston, MA 02111, USA
| | | | | | | | | | | |
Collapse
|
44
|
Conway Morris A, Kefala K, Wilkinson TS, Dhaliwal K, Farrell L, Walsh T, Mackenzie SJ, Reid H, Davidson DJ, Haslett C, Rossi AG, Sallenave JM, Simpson AJ. C5a mediates peripheral blood neutrophil dysfunction in critically ill patients. Am J Respir Crit Care Med 2009; 180:19-28. [PMID: 19324972 PMCID: PMC2948533 DOI: 10.1164/rccm.200812-1928oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
RATIONALE Critically ill patients are highly susceptible to hospital-acquired infection. Neutrophil function in critical illness remains poorly understood. OBJECTIVES To characterize and define mechanisms of peripheral blood neutrophil (PBN) dysfunction in critically ill patients. To determine whether the inflamed lung contributes additional phagocytic impairment. METHODS Prospective collection of blood and bronchoalveolar lavage fluid from patients with suspected ventilator-associated pneumonia and from age- and sex-matched volunteers; laboratory analysis of neutrophil functions. MEASUREMENTS AND MAIN RESULTS Seventy-two patients and 21 volunteers were included. Phagocytic capacity of PBNs was 36% lower in patients than in volunteers (P < 0.0001). From several biologically plausible candidates only activated complement was significantly associated with impaired PBN phagocytosis (P < 0.0001). Phagocytosis was negatively correlated with serum C3a and positively correlated with expression of C5a receptor type 1 (CD88) on PBNs. C5a recapitulated impaired PBN phagocytosis and significantly down-regulated CD88 expression in vitro. C5a-mediated phagocytic impairment was prevented by blocking either CD88 or phosphoinositide 3-kinase, and completely reversed by granulocyte-macrophage colony-stimulating factor. C5a also impaired killing of Pseudomonas aeruginosa by, and migration of, PBNs, indicating that effects were not restricted to phagocytosis. Bronchoalveolar lavage fluid leukocytes from patients also demonstrated significantly impaired function, and lavage supernatant reduced phagocytosis in healthy neutrophils by 43% (P = 0.0001). However, lavage fluid did not affect CD88 expression and lavage-mediated impairment of phagocytosis was not blocked by anti-CD88 antibody. CONCLUSIONS Critically ill patients have significant dysfunction of PBNs, which is mediated predominantly by activated complement. Further, profound complement-independent neutrophil dysfunction occurs in the inflamed lung.
Collapse
Affiliation(s)
- Andrew Conway Morris
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh, Scotland, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
BACKGROUND Remarkable progress has been made during the last decade in defining the molecular mechanisms that underlie septic shock. This rapidly expanding field is leading to new therapeutic opportunities in the management of severe sepsis. AIM To provide the clinician with a timely summary of the molecular biology of sepsis and to better understand recent advances in sepsis research. DATA SELECTION Medline search of relevant publications in basic mechanisms of sepsis/severe sepsis/septic shock, and selected literature review of other manuscripts about the signalosome, inflammasome, apoptosis, or mechanisms of shock. DATA SYNTHESIS AND FINDINGS: The identification of the toll-like receptors and the associated concept of innate immunity based upon pathogen- or damage-associated molecular pattern molecules allowed significant advances in our understanding of the pathophysiology of sepsis. The essential elements of the inflammasome and signal transduction networks responsible for activation of the host response have now been characterized. Apoptosis, mitochondrial dysfunction, sepsis-related immunosuppression, late mediators of systemic inflammation, control mechanisms for coagulation, and reprogramming of immune response genes all have critical roles in the development of sepsis. CONCLUSIONS Many of these basic discoveries have direct implications for the clinical management of sepsis. The translation of these "bench-to-bedside" findings into new therapeutic strategies is already underway. This brief review provides the clinician with a primer into the basic mechanisms responsible for the molecular biology of sepsis, severe sepsis, and septic shock.
Collapse
|
46
|
Dommisch H, Steglich M, Eberhard J, Winter J, Jepsen S. Phosphatidylinositol-3-kinase inhibitor LY 294002 blocksStreptococcus mutans-induced interleukin (IL)-6 and IL-8 gene expression in odontoblast-like cells. Int Endod J 2008; 41:763-71. [DOI: 10.1111/j.1365-2591.2008.01429.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
47
|
Brekke OL, Christiansen D, Fure H, Pharo A, Fung M, Riesenfeld J, Mollnes TE. Combined inhibition of complement and CD14 abolish E. coli-induced cytokine-, chemokine- and growth factor-synthesis in human whole blood. Mol Immunol 2008; 45:3804-13. [PMID: 18606453 DOI: 10.1016/j.molimm.2008.05.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 05/24/2008] [Accepted: 05/25/2008] [Indexed: 10/21/2022]
Abstract
The relative role of complement and CD14 in E. coli-induced cytokine synthesis in an in vitro human whole blood model of sepsis was examined. Fresh lepirudin-anticoagulated whole blood was incubated with E. coli for 2h. Monoclonal antibodies or a C5a receptor antagonist were used to block complement. Inflammatory mediators (n=27) were measured by multiplex technology, selected cytokine mRNA by real time PCR, and CD11b, oxidative burst and phagocytosis by flow cytometry. E. coli significantly increased 18 of the 27 inflammatory mediators, including proinflammatory cytokines (TNF-alpha, IL-6, INF-gamma and IL-1beta), chemokines (IL-8, MCP-1, MIP-1alpha, MIP-1beta, eotaxin and IP-10), growth factors (VEGF, FGF-basic, G-CSF and GM-CSF) and other interleukins (IL-9, IL-15 and IL-17). Notably, the increases in all mediators were abolished by a combined inhibition of CD14 and complement using anti-C2 and anti-factor D in combination, whereas the relative effect of the inhibition of complement and CD14 varied. In comparison, a C5a receptor antagonist and anti-CD14 in combination reduced cytokine synthesis less efficiently. Real time PCR analysis confirmed that the cytokine synthesis was blocked at the mRNA level. Similarly, E. coli-induced CD11b up-regulation, oxidative burst and phagocytosis was totally inhibited by CD14, anti-C2 and anti-factor D in combination after 2h incubation. In conclusion, the combined inhibition of complement using anti-C2, anti-factor D and CD14 almost completely inhibits the E. coli-induced inflammatory response. The combined approach may therefore be a new treatment regimen in Gram-negative sepsis.
Collapse
Affiliation(s)
- Ole-Lars Brekke
- Department of Laboratory Medicine, Nordland Hospital, Bodø, Norway.
| | | | | | | | | | | | | |
Collapse
|
48
|
Liu M. Ventilator-induced lung injury and mechanotransduction: why should we care? CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 11:168. [PMID: 18001490 PMCID: PMC2556754 DOI: 10.1186/cc6131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mechanotransduction holds the underlying mechanisms of ventilator-induced lung injury. Research on this subject, however, could be difficult for clinicians, especially when results are controversial. A recent study by Li and co-workers is used as an example, to explain how to critically read literatures related to basic science and how to understand the limitation of experimental studies.
Collapse
|
49
|
Bates JT, Honko AN, Graff AH, Kock ND, Mizel SB. Mucosal adjuvant activity of flagellin in aged mice. Mech Ageing Dev 2008; 129:271-81. [PMID: 18367233 DOI: 10.1016/j.mad.2008.01.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Revised: 12/11/2007] [Accepted: 01/25/2008] [Indexed: 01/22/2023]
Abstract
We evaluated the ability of flagellin, a highly effective mucosal adjuvant in mice and non-human primates, to promote mucosal innate and adaptive immunity in aged mice. We found that intratracheal instillation of flagellin induced a stronger respiratory innate response in aged mice than in young mice, and that intranasal instillation of flagellin was equally effective at triggering recruitment of T and B lymphocytes to the draining lymph nodes of young and aged mice. Intranasal immunization of aged mice with flagellin and the Yersinia pestis protein F1 promoted specific IgG and IgA production, but at lower levels and lower avidities than in young mice. Although intranasal instillation of flagellin and F1 antigen increased germinal center formation and size in young mice, it did not do so in aged mice. Our findings are consistent with the conclusion that flagellin can promote adaptive immune responses in aged mice, but at a less robust level than in young mice.
Collapse
Affiliation(s)
- John T Bates
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | |
Collapse
|