1
|
Mukherjee S, Chopra A, Karmakar S, Bhat SG. Periodontitis increases the risk of gastrointestinal dysfunction: an update on the plausible pathogenic molecular mechanisms. Crit Rev Microbiol 2025; 51:187-217. [PMID: 38602474 DOI: 10.1080/1040841x.2024.2339260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/28/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024]
Abstract
Periodontitis is an immuno-inflammatory disease of the soft tissues surrounding the teeth. Periodontitis is linked to many communicable and non-communicable diseases such as diabetes, cardiovascular disease, rheumatoid arthritis, and cancers. The oral-systemic link between periodontal disease and systemic diseases is attributed to the spread of inflammation, microbial products and microbes to distant organ systems. Oral bacteria reach the gut via swallowed saliva, whereby they induce gut dysbiosis and gastrointestinal dysfunctions. Some periodontal pathogens like Porphyromonas. gingivalis, Klebsiella, Helicobacter. Pylori, Streptococcus, Veillonella, Parvimonas micra, Fusobacterium nucleatum, Peptostreptococcus, Haemophilus, Aggregatibacter actinomycetomcommitans and Streptococcus mutans can withstand the unfavorable acidic, survive in the gut and result in gut dysbiosis. Gut dysbiosis increases gut inflammation, and induce dysplastic changes that lead to gut dysfunction. Various studies have linked oral bacteria, and oral-gut axis to various GIT disorders like inflammatory bowel disease, liver diseases, hepatocellular and pancreatic ductal carcinoma, ulcerative colitis, and Crohn's disease. Although the correlation between periodontitis and GIT disorders is well established, the intricate molecular mechanisms by which oral microflora induce these changes have not been discussed extensively. This review comprehensively discusses the intricate and unique molecular and immunological mechanisms by which periodontal pathogens can induce gut dysbiosis and dysfunction.
Collapse
Affiliation(s)
- Sayantan Mukherjee
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Aditi Chopra
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shaswata Karmakar
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Subraya Giliyar Bhat
- Department of Preventive Dental Sciences, Division of Periodontology, College of Dental Surgery, Iman Abdulrahman Bin Faizal University, Dammam, Saudi Arabia
| |
Collapse
|
2
|
Silva de França F, de Oliveira Orsi R, Fernandes DC, Leonel TB, Tambourgi DV. Africanized honeybee venom ( Apis mellifera) promotes human complement activation split products storm. Front Immunol 2024; 15:1463471. [PMID: 39606222 PMCID: PMC11598452 DOI: 10.3389/fimmu.2024.1463471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/14/2024] [Indexed: 11/29/2024] Open
Abstract
Introduction Complement activation split products are signatures of many immunopathological disorders. Among the laboratory findings observed in these diseases, a reduction in the level of circulating intact complement components can be mentioned, and this change has also been detected in envenomation by multiple Africanized honeybee (Apis mellifera) stings. Although envenomation by these animals elicits diverse life-threatening reactions, the capacity of bee venom (AmV) to activate the human complement system remains elusive. Methods and findings By coupling immunochemical and functional approaches, it was observed that AmV strongly consumes components of the alternative pathway (AP) of the complement system in normal human serum (NHS). Additionally, AmV interfered with classical (CP) and lectin pathways (LP) activities. In parallel, a high increase in Ba fragment levels was detected, suggesting that the changes in AP activity were due to its activation. Furthermore, an increase in the level of the C1s-C1INH complex and a decrease in the physiological level of MASP1-C1INH suggested that CP and LP were also activated in the presence of AmV. Strikingly, NHS exposed to increasing AmV concentrations varying from 5 to 1000 µg/mL presented a high generation of C3a, C4a and C5a anaphylatoxins, and sC5b-9 complexes assembly, thus reinforcing that AmV triggers complement activation. Conclusion These results show that AmV is a strong complement activator. This activation presents a mixed profile, with a predominance of AP activation. This suggests that complement split products can play important roles in the envenomation by Africanized honeybee, as they could induce diverse immunopathological events observed in patients and may also dictate patient clinical prognosis.
Collapse
Affiliation(s)
- Felipe Silva de França
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| | - Ricardo de Oliveira Orsi
- Center of Education, Science and Technology in Rational Beekeeping (NECTAR), College of Veterinary Medicine and Animal Sciences, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Dayanne Carla Fernandes
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| | - Thyago Bispo Leonel
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| | - Denise V. Tambourgi
- Immunochemistry Laboratory, Butantan Institute, São Paulo, Brazil
- Center of Toxins, Cell Signaling and Immune Response (CeTICS) – CEPID - FAPESP, São Paulo, Brazil
| |
Collapse
|
3
|
Nagarajan A, Scoggin K, Adams LG, Threadgill D, Andrews-Polymenis H. Identification of a genetic region linked to tolerance to MRSA infection using Collaborative Cross mice. PLoS Genet 2024; 20:e1011378. [PMID: 39178306 PMCID: PMC11407622 DOI: 10.1371/journal.pgen.1011378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 09/17/2024] [Accepted: 07/29/2024] [Indexed: 08/25/2024] Open
Abstract
Staphylococcus aureus (S. aureus) colonizes humans asymptomatically but can also cause opportunistic infections, ranging from mild skin infections to severe life-threatening conditions. Resistance and tolerance are two ways a host can survive an infection. Resistance is limiting the pathogen burden, while tolerance is limiting the health impact of a given pathogen burden. In previous work, we established that collaborative cross (CC) mouse line CC061 is highly susceptible to Methicillin-resistant S. aureus infection (MRSA, USA300), while CC024 is tolerant. To identify host genes involved in tolerance after S. aureus infection, we crossed CC061 mice and CC024 mice to generate F1 and F2 populations. Survival after MRSA infection in the F1 and F2 generations was 65% and 55% and followed a complex dominant inheritance pattern for the CC024 increased survival phenotype. Colonization in F2 animals was more extreme than in their parents, suggesting successful segregation of genetic factors. We identified a Quantitative Trait Locus (QTL) peak on chromosome 7 for survival and weight change after infection. In this QTL, the WSB/EiJ (WSB) allele was present in CC024 mice and contributed to their MRSA tolerant phenotype. Two genes, C5ar1 and C5ar2, have high-impact variants in this region. C5ar1 and C5ar2 are receptors for the complement factor C5a, an anaphylatoxin that can trigger a massive immune response by binding to these receptors. We hypothesize that C5a may have altered binding to variant receptors in CC024 mice, reducing damage caused by the cytokine storm and resulting in the ability to tolerate a higher pathogen burden and longer survival.
Collapse
Affiliation(s)
- Aravindh Nagarajan
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Kristin Scoggin
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - David Threadgill
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas, United States of America
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| | - Helene Andrews-Polymenis
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
4
|
Ghosh M, Rana S. The anaphylatoxin C5a: Structure, function, signaling, physiology, disease, and therapeutics. Int Immunopharmacol 2023; 118:110081. [PMID: 36989901 DOI: 10.1016/j.intimp.2023.110081] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/06/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
The complement system is one of the oldest known tightly regulated host defense systems evolved for efficiently functioning cell-based immune systems and antibodies. Essentially, the complement system acts as a pivot between the innate and adaptive arms of the immune system. The complement system collectively represents a cocktail of ∼50 cell-bound/soluble glycoproteins directly involved in controlling infection and inflammation. Activation of the complement cascade generates complement fragments like C3a, C4a, and C5a as anaphylatoxins. C5a is the most potent proinflammatory anaphylatoxin, which is involved in inflammatory signaling in a myriad of tissues. This review provides a comprehensive overview of human C5a in the context of its structure and signaling under several pathophysiological conditions, including the current and future therapeutic applications targeting C5a.
Collapse
Affiliation(s)
- Manaswini Ghosh
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India
| | - Soumendra Rana
- Chemical Biology Laboratory, School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Odisha 752050, India.
| |
Collapse
|
5
|
Guo Z, Fan X, Yao J, Tomlinson S, Yuan G, He S. The role of complement in nonalcoholic fatty liver disease. Front Immunol 2022; 13:1017467. [PMID: 36248852 PMCID: PMC9562907 DOI: 10.3389/fimmu.2022.1017467] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/13/2022] [Indexed: 11/14/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become a leading cause of chronic liver diseases globally. NAFLD includes a range of hepatic manifestations, starting with liver steatosis and potentially evolving towards nonalcoholic steatohepatitis, cirrhosis or even hepatocellular carcinoma. Although the pathogenesis of NAFLD is incompletely understood, insulin resistance and lipid metabolism disorder are implicated. The complement system is an essential part of the immune system, but it is also involved in lipid metabolism. In particular, activation of the alternative complement pathway and the production of complement activation products such as C3a, C3adesArg (acylation stimulating protein or ASP) and C5a, are strongly associated with insulin resistance, lipid metabolism disorder, and hepatic inflammation. In this review, we briefly summarize research on the role of the complement system in NAFLD, aiming to provide a basis for the development of novel therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Zhenya Guo
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, China
| | - Xiude Fan
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianni Yao
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, China
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Guandou Yuan
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, China
| | - Songqing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Tremblay JA, Peron F, Kreitmann L, Textoris J, Brengel-Pesce K, Lukaszewicz AC, Quemeneur L, Vedrine C, Tan LK, Venet F, Rimmele T, Monneret G. A stratification strategy to predict secondary infection in critical illness-induced immune dysfunction: the REALIST score. Ann Intensive Care 2022; 12:76. [PMID: 35976460 PMCID: PMC9382015 DOI: 10.1186/s13613-022-01051-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/03/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Although multiple individual immune parameters have been demonstrated to predict the occurrence of secondary infection after critical illness, significant questions remain with regards to the selection, timing and clinical utility of such immune monitoring tests. RESEARCH QUESTION As a sub-study of the REALISM study, the REALIST score was developed as a pragmatic approach to help clinicians better identify and stratify patients at high risk for secondary infection, using a simple set of relatively available and technically robust biomarkers. STUDY DESIGN AND METHODS This is a sub-study of a single-centre prospective cohort study of immune profiling in critically ill adults admitted after severe trauma, major surgery or sepsis/septic shock. For the REALIST score, five immune parameters were pre-emptively selected based on their clinical applicability and technical robustness. Predictive power of different parameters and combinations of parameters was assessed. The main outcome of interest was the occurrence of secondary infection within 30 days. RESULTS After excluding statistically redundant and poorly predictive parameters, three parameters remained in the REALIST score: mHLA-DR, percentage of immature (CD10- CD16-) neutrophils and serum IL-10 level. In the cohort of interest (n = 189), incidence of secondary infection at day 30 increased from 8% for patients with REALIST score of 0 to 46% in patients with a score of 3 abnormal parameters, measured ad D5-7. When adjusted for a priori identified clinical risk factors for secondary infection (SOFA score and invasive mechanical ventilation at D5-7), a higher REALIST score was independently associated with increased risk of secondary infection (42 events (22.2%), adjusted HR 3.22 (1.09-9.50), p = 0.034) and mortality (10 events (5.3%), p = 0.001). INTERPRETATION We derived and presented the REALIST score, a simple and pragmatic stratification strategy which provides clinicians with a clear assessment of the immune status of their patients. This new tool could help optimize care of these individuals and could contribute in designing future trials of immune stimulation strategies.
Collapse
Affiliation(s)
- Jan-Alexis Tremblay
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France. .,Critical Care Service, Hôpital Maisonneuve-Rosemont, 5415 Boulevard de l'Assomption, Montréal, H1T2M4, Canada.
| | - Florian Peron
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France
| | - Louis Kreitmann
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France
| | - Julien Textoris
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France
| | - Karen Brengel-Pesce
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France.,Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Laurence Quemeneur
- Sanofi Pasteur, Sanofi 1541 avenue Marcel Mérieux, 69280, Marcy l'Etoile, France
| | | | - Lionel K Tan
- GSK, 980 Great West Road, Brentford, Middlesex, TW8 9GS, UK
| | - Fabienne Venet
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France.,Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Team 'NLRP3 Inflammation and Immune Response to Sepsis', Université Claude Bernard-Lyon 1, Lyon, France
| | - Thomas Rimmele
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France.,Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Guillaume Monneret
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Herriot Hospital, 5 place d'Arsonval, 69003, Lyon, France.,Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | | |
Collapse
|
7
|
Chen K, Lin Y, Liu Y, Liao S, Yang R, Huang J, Xu M, He J. Investigation of Association of Complement 5 Genetic Polymorphisms with Sepsis and Sepsis-Induced Inflammatory Responses. J Inflamm Res 2021; 14:6461-6475. [PMID: 34880647 PMCID: PMC8648101 DOI: 10.2147/jir.s340446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/17/2021] [Indexed: 11/23/2022] Open
Abstract
Background Complement 5 (C5) and C5a production play a pivotal role in the pathophysiology of sepsis. Strong evidence demonstrates an association of C5 gene polymorphisms with various inflammatory diseases. However, no current studies have explored the clinical relevance of C5 polymorphisms in sepsis. Methods Two C5 gene polymorphisms, rs17611 and rs2269067, were identified by genotyping in 636 sepsis patients and 753 controls in a Han Chinese population. C5 gene expression was detected via quantitative real-time PCR. C5a and proinflammatory cytokine production was measured by enzyme-linked immunosorbent assay. An Annexin V apoptosis assay was performed to assess cell apoptosis. Results Our results showed significantly lower frequencies of rs2269067 GC/CC genotypes or C allele in sepsis patients than healthy controls. The frequencies of rs17611 CC/CT genotypes or C allele were significantly overrepresented in both the septic shock and non-survivor subgroups. Patients with this sepsis-associated high-risk rs17611 C allele exhibited a significant increase in C5a, TNF-α and IL-6 production. However, no significant difference in C5a and downstream proinflammatory cytokine production was observed among patients with different rs2269067 genotypes. In addition, in vitro experiments showed an effect of recombinant C5a on enhancing LPS-stimulated IL-1β, IL-6 and TNF-α production and cell apoptosis in THP-1 monocytes. Conclusion The rs2269067 polymorphism conferred protection against sepsis susceptibility. The rs17611 polymorphism was associated with increased C5a production, which ultimately potentiated the secretion of downstream proinflammatory cytokines and conferred susceptibility to sepsis progression and poor prognosis.
Collapse
Affiliation(s)
- Kaidian Chen
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Yao Lin
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Yuchun Liu
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Shuanglin Liao
- The Intensive Care Unit, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, People's Republic of China
| | - Ruoxuan Yang
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Jiefeng Huang
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Mingwei Xu
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| | - Junbing He
- The Intensive Care Unit, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, Guangdong, People's Republic of China
| |
Collapse
|
8
|
Clinicopathological features and prognostic significance of C5aR in human solid tumors: a Meta-analysis. BMC Cancer 2021; 21:1136. [PMID: 34688269 PMCID: PMC8540875 DOI: 10.1186/s12885-021-08883-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/13/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND C5aR has been extensively studied in recent years as an essential component of the complement system. However, the role of C5aR in tumors has not been sufficiently investigated and summarized. The aim of this meta-analysis was to investigate the prognostic value of C5aR in solid tumors as well as the correlation between C5aR and clinicopathological features. METHODS Relevant study collection was performed in PubMed, Embase, Web of Science, BIOSIS Previews, Cochrane Library until July 10, 2021. Pooled hazard ratios (HRs), odds ratios (ORs), and 95% confidence intervals (CIs) were calculated. Sensitivity analyses were performed to assess the robustness of this study, while publication bias was tested by Begg's and Egger's tests. RESULTS A total of 11 studies involving 1577 patients were included in the study. Our results suggest that the high-level C5aR expression in tumor tissue predicted unsatisfactory overall survival (OS) (HR = 1.92, 95% CI:1.47-2.50, P < 0.001) and recurrence-free survival (RFS) (HR = 2.19, 95% CI:1.47-3.27, P < 0.001). Besides, a higher level of C5aR expression was associated with larger tumor size (OR = 1.58, 95% CI: 1.18-2.10, P = 0.002) and the occurrence of metastases in lymph nodes (OR = 1.99, 95% CI: 1.46-2.72, P<0.001), whereas it was independent of tumor stage, vascular invasion and tumor differentiation. CONCLUSION In conclusion, C5aR may be a potential biomarker for evaluating tumor prognosis and treatment.
Collapse
|
9
|
Lehnert T, Leonhardt I, Timme S, Thomas-Rüddel D, Bloos F, Sponholz C, Kurzai O, Figge MT, Hünniger K. Ex vivo immune profiling in patient blood enables quantification of innate immune effector functions. Sci Rep 2021; 11:12039. [PMID: 34103589 PMCID: PMC8187451 DOI: 10.1038/s41598-021-91362-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/25/2021] [Indexed: 02/05/2023] Open
Abstract
The assessment of a patient's immune function is critical in many clinical situations. In complex clinical immune dysfunction like sepsis, which results from a loss of immune homeostasis due to microbial infection, a plethora of pro- and anti-inflammatory stimuli may occur consecutively or simultaneously. Thus, any immunomodulatory therapy would require in-depth knowledge of an individual patient's immune status at a given time. Whereas lab-based immune profiling often relies solely on quantification of cell numbers, we used an ex vivo whole-blood infection model in combination with biomathematical modeling to quantify functional parameters of innate immune cells in blood from patients undergoing cardiac surgery. These patients experience a well-characterized inflammatory insult, which results in mitigation of the pathogen-specific response patterns towards Staphylococcus aureus and Candida albicans that are characteristic of healthy people and our patients at baseline. This not only interferes with the elimination of these pathogens from blood, but also selectively augments the escape of C. albicans from phagocytosis. In summary, our model could serve as a valuable functional immune assay for recording and evaluating innate responses to infection.
Collapse
Affiliation(s)
- Teresa Lehnert
- grid.418398.f0000 0001 0143 807XResearch Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology ‐ Hans Knöll Institute, Jena, Germany ,grid.275559.90000 0000 8517 6224Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | - Ines Leonhardt
- grid.275559.90000 0000 8517 6224Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany ,grid.418398.f0000 0001 0143 807XResearch Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology ‐ Hans Knöll Institute, Jena, Germany
| | - Sandra Timme
- grid.418398.f0000 0001 0143 807XResearch Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology ‐ Hans Knöll Institute, Jena, Germany
| | - Daniel Thomas-Rüddel
- grid.275559.90000 0000 8517 6224Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany ,grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Frank Bloos
- grid.275559.90000 0000 8517 6224Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany ,grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Christoph Sponholz
- grid.275559.90000 0000 8517 6224Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Oliver Kurzai
- grid.275559.90000 0000 8517 6224Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany ,grid.418398.f0000 0001 0143 807XResearch Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology ‐ Hans Knöll Institute, Jena, Germany ,grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| | - Marc Thilo Figge
- grid.418398.f0000 0001 0143 807XResearch Group Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology ‐ Hans Knöll Institute, Jena, Germany ,grid.275559.90000 0000 8517 6224Center for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany ,grid.9613.d0000 0001 1939 2794Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Jena, Germany
| | - Kerstin Hünniger
- grid.418398.f0000 0001 0143 807XResearch Group Fungal Septomics, Leibniz Institute for Natural Product Research and Infection Biology ‐ Hans Knöll Institute, Jena, Germany ,grid.8379.50000 0001 1958 8658Institute for Hygiene and Microbiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Zetoune FS, Ward PA. Role of Complement and Histones in Sepsis. Front Med (Lausanne) 2020; 7:616957. [PMID: 33425963 PMCID: PMC7785970 DOI: 10.3389/fmed.2020.616957] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
The wide use of the mouse model of polymicrobial sepsis has provided important evidence for events occurring in infectious sepsis involving septic mice and septic humans. Nearly 100 clinical trials in humans with sepsis have been completed, yet there is no FDA-approved drug. Our studies of polymicrobial sepsis have highlighted the role of complement activation products (especially C5a anaphylatoxin and its receptors C5aR1 and C5aR2) in adverse effects of sepsis. During sepsis, the appearance of these complement products is followed by appearance of extracellular histones in plasma, which have powerful proinflammatory and prothrombotic activities that cause cell injury and multiorgan dysfunction in septic mice. Similar responses occur in septic humans. Histone appearance in plasma is related to complement activation and appearance of C5a and its interaction with its receptors. Development of the cardiomyopathy of sepsis also depends on C5a, C5a receptors and histones. Neutralization of C5a with antibody or absence of C5aR1 blocks appearance of extracellular histones and cell and organ failure in sepsis. Survival rates in septic mice are greatly improved after blockade of C5a with antibody. We also review the various strategies in sepsis that greatly reduce the development of life-threatening events of sepsis.
Collapse
Affiliation(s)
- Firas S Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
11
|
Carvelli J, Demaria O, Vély F, Batista L, Chouaki Benmansour N, Fares J, Carpentier S, Thibult ML, Morel A, Remark R, André P, Represa A, Piperoglou C, Cordier PY, Le Dault E, Guervilly C, Simeone P, Gainnier M, Morel Y, Ebbo M, Schleinitz N, Vivier E. Association of COVID-19 inflammation with activation of the C5a-C5aR1 axis. Nature 2020; 588:146-150. [PMID: 32726800 PMCID: PMC7116884 DOI: 10.1038/s41586-020-2600-6] [Citation(s) in RCA: 363] [Impact Index Per Article: 72.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a disease caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and has resulted in a pandemic1. The C5a complement factor and its receptor C5aR1 (also known as CD88) have a key role in the initiation and maintenance of several inflammatory responses by recruiting and activating neutrophils and monocytes1. Here we provide a longitudinal analysis of immune responses, including phenotypic analyses of immune cells and assessments of the soluble factors that are present in the blood and bronchoalveolar lavage fluid of patients at various stages of COVID-19 severity, including those who were paucisymptomatic or had pneumonia or acute respiratory distress syndrome. The levels of soluble C5a were increased in proportion to the severity of COVID-19 and high expression levels of C5aR1 receptors were found in blood and pulmonary myeloid cells, which supports a role for the C5a-C5aR1 axis in the pathophysiology of acute respiratory distress syndrome. Anti-C5aR1 therapeutic monoclonal antibodies prevented the C5a-mediated recruitment and activation of human myeloid cells, and inhibited acute lung injury in human C5aR1 knock-in mice. These results suggest that blockade of the C5a-C5aR1 axis could be used to limit the infiltration of myeloid cells in damaged organs and prevent the excessive lung inflammation and endothelialitis that are associated with acute respiratory distress syndrome in patients with COVID-19.
Collapse
Affiliation(s)
- Julien Carvelli
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Réanimation des Urgences, Marseilles, France
- Aix Marseille Université, Marseilles, France
| | | | - Frédéric Vély
- Aix Marseille Université, CNRS, INSERM, CIML, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Immunology, Marseille Immunopole, Marseilles, France
| | | | - Nassima Chouaki Benmansour
- Hôpital d'Instruction des Armées Laveran, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Marseilles, France
| | | | | | | | | | | | | | | | - Christelle Piperoglou
- Aix Marseille Université, CNRS, INSERM, CIML, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Immunology, Marseille Immunopole, Marseilles, France
| | | | - Erwan Le Dault
- Hôpital d'Instruction des Armées Laveran, Marseilles, France
| | - Christophe Guervilly
- Aix Marseille Université, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital Nord, Réanimation des Détresses Respiratoires et Infections Sévères, Aix-Marseille Université, Marseilles, France
| | - Pierre Simeone
- Aix Marseille Université, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Réanimation Polyvalente, Aix-Marseille Université, Marseilles, France
| | - Marc Gainnier
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Réanimation des Urgences, Marseilles, France
- Aix Marseille Université, Marseilles, France
| | | | - Mikael Ebbo
- Aix Marseille Université, CNRS, INSERM, CIML, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Internal Medicine, Marseilles, France
| | - Nicolas Schleinitz
- Aix Marseille Université, CNRS, INSERM, CIML, Marseilles, France
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Internal Medicine, Marseilles, France
| | - Eric Vivier
- Innate Pharma, Marseilles, France.
- Aix Marseille Université, CNRS, INSERM, CIML, Marseilles, France.
- Assistance Publique des Hôpitaux de Marseille, Hôpital de la Timone, Immunology, Marseille Immunopole, Marseilles, France.
| |
Collapse
|
12
|
McConnel CS, Crisp SA, Biggs TD, Ficklin SP, Parrish LM, Trombetta SC, Sischo WM, Adams-Progar A. A Fixed Cohort Field Study of Gene Expression in Circulating Leukocytes From Dairy Cows With and Without Mastitis. Front Vet Sci 2020; 7:559279. [PMID: 33195534 PMCID: PMC7554338 DOI: 10.3389/fvets.2020.559279] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/03/2020] [Indexed: 12/04/2022] Open
Abstract
Specifically designed gene expression studies can be used to prioritize candidate genes and identify novel biomarkers affecting resilience against mastitis and other diseases in dairy cattle. The primary goal of this study was to assess whether specific peripheral leukocyte genes expressed differentially in a previous study of dairy cattle with postpartum disease, also would be expressed differentially in peripheral leukocytes from a diverse set of different dairy cattle with moderate to severe clinical mastitis. Four genes were selected for this study due to their differential expression in a previous transcriptomic analysis of circulating leukocytes from dairy cows with and without evidence of early postpartum disease. An additional 15 genes were included based on their cellular, immunologic, and inflammatory functions associated with resistance and tolerance to mastitis. This fixed cohort study was conducted on a conventional dairy in Washington state. Cows >50 days in milk (DIM) with mastitis (n = 12) were enrolled along with healthy cows (n = 8) selected to match the DIM and lactation numbers of mastitic cows. Blood was collected for a complete blood count (CBC), serum biochemistry, leukocyte isolation, and RNA extraction on the day of enrollment and twice more at 6 to 8-days intervals. Latent class analysis was performed to discriminate healthy vs. mastitic cows and to describe disease resolution. RNA samples were processed by the Primate Diagnostic Services Laboratory (University of Washington, Seattle, WA). Gene expression analysis was performed using the Nanostring System (Nanostring Technologies, Seattle, Washington, USA). Of the four genes (C5AR1, CATHL6, LCN2, and PGLYRP1) with evidence of upregulation in cows with mastitis, three of those genes (CATHL6, LCN2, and PGLYRP1) were investigated due to their previously identified association with postpartum disease. These genes are responsible for immunomodulatory molecules that selectively enhance or alter host innate immune defense mechanisms and modulate pathogen-induced inflammatory responses. Although further research is warranted to explain their functional mechanisms and bioactivity in cattle, our findings suggest that these conserved elements of innate immunity have the potential to bridge disease states and target tissues in diverse dairy populations.
Collapse
Affiliation(s)
- Craig S McConnel
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sierra A Crisp
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Tyler D Biggs
- Department of Horticulture, College of Agriculture, Human, and Natural Resource Sciences, Washington State University, Pullman, WA, United States
| | - Stephen P Ficklin
- Department of Horticulture, College of Agriculture, Human, and Natural Resource Sciences, Washington State University, Pullman, WA, United States
| | - Lindsay M Parrish
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Sophie C Trombetta
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - William M Sischo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA, United States
| | - Amber Adams-Progar
- Department of Animal Sciences, College of Agriculture, Human, and Natural Resource Sciences, Washington State University, Pullman, WA, United States
| |
Collapse
|
13
|
Tromp AT, Zhao Y, Jongerius I, Heezius ECJM, Abrial P, Ruyken M, van Strijp JAG, de Haas CJC, Spaan AN, van Kessel KPM, Henry T, Haas PJA. Pre-existing antibody-mediated adverse effects prevent the clinical development of a bacterial anti-inflammatory protein. Dis Model Mech 2020; 13:dmm045534. [PMID: 32471891 PMCID: PMC7541340 DOI: 10.1242/dmm.045534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022] Open
Abstract
Bacterial pathogens have evolved to secrete strong anti-inflammatory proteins that target the immune system. It was long speculated whether these virulence factors could serve as therapeutics in diseases in which abnormal immune activation plays a role. We adopted the secreted chemotaxis inhibitory protein of Staphylococcus aureus (CHIPS) as a model virulence factor-based therapeutic agent for diseases in which C5AR1 stimulation plays an important role. We show that the administration of CHIPS in human C5AR1 knock-in mice successfully dampens C5a-mediated neutrophil migration during immune complex-initiated inflammation. Subsequent CHIPS toxicology studies in animal models were promising. However, during a small phase I trial, healthy human volunteers showed adverse effects directly after CHIPS administration. Subjects showed clinical signs of anaphylaxis with mild leukocytopenia and increased C-reactive protein concentrations, which are possibly related to the presence of relatively high circulating anti-CHIPS antibodies and suggest an inflammatory response. Even though our data in mice show CHIPS as a potential anti-inflammatory agent, safety issues in human subjects temper the use of CHIPS in its current form as a therapeutic candidate. The use of staphylococcal proteins, or other bacterial proteins, as therapeutics or immune-modulators in humans is severely hampered by pre-existing circulating antibodies.
Collapse
Affiliation(s)
- Angelino T Tromp
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Yuxi Zhao
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Ilse Jongerius
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
- Sanquin Research, Department of Immunopathology, 1006AD Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, 1105AZ Amsterdam, The Netherlands
| | - Erik C J M Heezius
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Pauline Abrial
- Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, Ecole Normale Supérieure de Lyon, Université Lyon, 69007 Lyon, France
| | - Maartje Ruyken
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Jos A G van Strijp
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Carla J C de Haas
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - András N Spaan
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065, USA
| | - Kok P M van Kessel
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| | - Thomas Henry
- Sanquin Research, Department of Immunopathology, 1006AD Amsterdam, The Netherlands
| | - Pieter-Jan A Haas
- Department of Medical Microbiology, University Medical Center Utrecht, 3584CX Utrecht, The Netherlands
| |
Collapse
|
14
|
Metzemaekers M, Gouwy M, Proost P. Neutrophil chemoattractant receptors in health and disease: double-edged swords. Cell Mol Immunol 2020; 17:433-450. [PMID: 32238918 PMCID: PMC7192912 DOI: 10.1038/s41423-020-0412-0] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are frontline cells of the innate immune system. These effector leukocytes are equipped with intriguing antimicrobial machinery and consequently display high cytotoxic potential. Accurate neutrophil recruitment is essential to combat microbes and to restore homeostasis, for inflammation modulation and resolution, wound healing and tissue repair. After fulfilling the appropriate effector functions, however, dampening neutrophil activation and infiltration is crucial to prevent damage to the host. In humans, chemoattractant molecules can be categorized into four biochemical families, i.e., chemotactic lipids, formyl peptides, complement anaphylatoxins and chemokines. They are critically involved in the tight regulation of neutrophil bone marrow storage and egress and in spatial and temporal neutrophil trafficking between organs. Chemoattractants function by activating dedicated heptahelical G protein-coupled receptors (GPCRs). In addition, emerging evidence suggests an important role for atypical chemoattractant receptors (ACKRs) that do not couple to G proteins in fine-tuning neutrophil migratory and functional responses. The expression levels of chemoattractant receptors are dependent on the level of neutrophil maturation and state of activation, with a pivotal modulatory role for the (inflammatory) environment. Here, we provide an overview of chemoattractant receptors expressed by neutrophils in health and disease. Depending on the (patho)physiological context, specific chemoattractant receptors may be up- or downregulated on distinct neutrophil subsets with beneficial or detrimental consequences, thus opening new windows for the identification of disease biomarkers and potential drug targets.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Rega Institute, KU Leuven, Herestraat 49 bus 1042, B-3000, Leuven, Belgium.
| |
Collapse
|
15
|
Abstract
There is abundant evidence that infectious sepsis both in humans and mice with polymicrobial sepsis results in robust activation of complement. Major complement activation products involved in sepsis include C5a anaphylatoxin and its receptors (C5aR1 and C5aR2) and, perhaps, the terminal complement activation product, C5b-9. These products (and others) also cause dysfunction of the innate immune system, with exaggerated early proinflammatory responses, followed by decline of the innate immune system, leading to immunosuppression and multiorgan dysfunction. Generation of C5a during sepsis also leads to activation of neutrophils and macrophages and ultimate appearance of extracellular histones, which have powerful proinflammatory and prothrombotic activities. The distal complement activation product, C5b-9, triggers intracellular Ca fluxes in epithelial and endothelial cells. Histones activate the NLRP3 inflammasome, products of which can damage cells. C5a also activates MAPKs and Akt signaling pathways in cardiomyocytes, causing buildup of [Ca]i, defective action potentials and substantial cell dysfunction, resulting in cardiac and other organ dysfunction. Cardiac dysfunction can be quantitated by ECHO-Doppler parameters. In vivo interventions that block these complement-dependent products responsible for organ dysfunction in sepsis reduce the intensity of sepsis. The obvious targets in sepsis are C5a and its receptors, histones, and perhaps the MAPK pathways. Blockade of C5 has been considered in sepsis, but the FDA-approved antibody (eculizumab) is known to compromise defenses against neisseria and pneumonococcal bacteria, and requires immunization before the mAb to C5 can be used clinically. Small molecular blocking agents for C5aRs are currently in development and may be therapeutically effective for treatment of sepsis.
Collapse
|
16
|
Abstract
OBJECTIVES Complement activation product C5a plays a critical role in systemic inflammatory response syndrome induced by viruses, bacteria, and toxic agents including paraquat poisoning. This study is to explore the efficiency of anti-C5a-based intervention on systemic inflammatory responses induced by paraquat poisoning. DESIGN Study of cynomolgus macaque model and plasma from paraquat-poisoning patients. SETTING Laboratory investigation. SUBJECTS Cynomolgus macaque (n = 12) and samples of plasma from patients (n = 16). INTERVENTIONS The neutralizing antihuman C5a antibody (IFX-1) was administered to investigate the new treatment strategy for paraquat-induced systemic inflammatory responses in cynomolgus macaque model. In addition, C5a activation in plasma of paraquat patients was blocked by IFX-1 to investigate the blockade role of anti-C5a antibody in activation of inflammatory cells. MEASUREMENTS AND MAIN RESULTS Dysregulated complement activation and the subsequent cytokine storm were found in patients with acute lung injury and in a primate model of paraquat poisoning. Targeted inhibition of C5a by IFX-1 led to marked alleviation of systemic inflammatory responses and multiple organ damage in the primate model. In addition, blockade of C5a activity in plasma from patients completely inhibited activation of CD11b on blood granulocytes from normal donors, suggesting that IFX-1 may alleviate the excessive activation of inflammatory responses and have clinical utility for patients with acute lung injury. CONCLUSIONS Anti-C5a antibodies such as IFX-1 may be used as effective therapeutics for treatment of those suffering from systemic inflammatory responses induced by chemical poisoning like paraquat.
Collapse
|
17
|
Li Y, Zhao Q, Liu B, Dixon A, Cancio L, Dubick M, Dalle Lucca J. Early complementopathy predicts the outcomes of patients with trauma. Trauma Surg Acute Care Open 2019; 4:e000217. [PMID: 31058236 PMCID: PMC6461142 DOI: 10.1136/tsaco-2018-000217] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 02/08/2019] [Accepted: 02/28/2019] [Indexed: 01/04/2023] Open
Abstract
Background Complementopathy (rapid complement activation and consumption after trauma) has been reported in trauma patients, but the underlying mechanism of these phenomena and their clinical significance remain unclear. This study aimed to determine the complement/complement pathway activation and identify the association of complement activation with clinical outcomes in trauma patients. Methods We studied 33 trauma patients with mean Injury Severity Score of 25, and 25 healthy volunteers. Sera were collected on patients’ arrival at the emergency department, as well as 1, 2, 3, 5, and 7 days after trauma, to measure the levels of terminal complement activation product soluble C5b-9 (sC5b-9) by ELISA. In addition, the functional complement activation pathway was evaluated using a commercial complement system screening kit. Results Serum concentrations of sC5b-9 (complement terminal pathway activity) were significantly increased in trauma patients throughout the entire observation period except on day 1. Complement terminal activities were significantly higher in 27 of 33 patients with systemic inflammatory response syndrome (SIRS) than non-SIRS patients on day 2, day 5, and day 7. Increased serum levels of sC5b-9 positively correlated with SIRS. Functional complement analysis revealed that the classical pathway was the predominant pathway responsible for complement activation. Burn patients tended to have a greater and prolonged classical pathway activation than non-burn patients, and burn injury and blunt injury were associated with higher blood levels of sC5b-9 than penetrating injury. Discussion Early complement activation through the classical pathway after trauma is observed and positively correlated with the development of SIRS. Thus, monitoring of the complement system might be beneficial in the care of critically injured patients. Level of evidence III. Study type Prognostic.
Collapse
Affiliation(s)
- Yansong Li
- Multiple Organ Support Technology, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Qingwei Zhao
- Burn Research, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Bin Liu
- Blood Research, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Alexander Dixon
- Multiple Organ Support Technology, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Leopoldo Cancio
- Multiple Organ Support Technology, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Michael Dubick
- Damage Control Resuscitation, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Jurandir Dalle Lucca
- Viral Therapeutics, Chemical and Biological Technologies, Fort Belvoir, Virginia, USA
| |
Collapse
|
18
|
Ward PA, Fattahi F. New strategies for treatment of infectious sepsis. J Leukoc Biol 2019; 106:187-192. [PMID: 30821872 DOI: 10.1002/jlb.4mir1118-425r] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
In this mini review, we describe the molecular mechanisms in polymicrobial sepsis that lead to a series of adverse events including activation of inflammatory and prothrombotic pathways, a faulty innate immune system, and multiorgan dysfunction. Complement activation is a well-established feature of sepsis, especially involving generation of C5a and C5b-9, along with engagement of relevant receptors for C5a. Activation of neutrophils by C5a leads to extrusion of DNA, forming neutrophil extracellular traps that contain myeloperoxidase and oxidases, along with extracellular histones. Generation of the distal complement activation product, C5b-9 (known as the membrane attack complex, MAC), also occurs in sepsis. C5b-9 activates the NLRP3 inflammasome, which damages mitochondria, together with appearance in plasma of IL-1β and IL-18. Histones are strongly proinflammatory as well as being prothrombotic, leading to activation of platelets and development of venous thrombosis. Multiorgan dysfunction is also a feature of sepsis. It is well known that septic cardiomyopathy, which if severe, can lead to death. This complication in sepsis is linked to reduced levels in cardiomyocytes of three critical proteins (SERCA2, NCX, Na+ /K+ -ATPase). The reductions in these three key proteins are complement- and histone-dependent. Dysfunction of these ATPases is linked to the cardiomyopathy of sepsis. These data suggest novel targets in the setting of sepsis in humans.
Collapse
Affiliation(s)
- Peter A Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Fatemeh Fattahi
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Fu Z, Xia L, De J, Zhu M, Li H, Lu Y, Chen D. Beneficial effects on H1N1-induced acute lung injury and structure characterization of anti-complementary acidic polysaccharides from Juniperus pingii var. wilsonii. Int J Biol Macromol 2019; 129:246-253. [PMID: 30708019 DOI: 10.1016/j.ijbiomac.2019.01.163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 01/20/2019] [Accepted: 01/28/2019] [Indexed: 01/20/2023]
Abstract
Juniperus pingii var. wilsonii has been traditionally used in Tibetan medicine for the treatment of inflammatory diseases. In the present study, J. pingii var. wilsonii polysaccharides (JPWP), with high content of d‑galacturonic acid, showed potent anti-complementary activity in vitro and significantly attenuated acute lung injury (ALI) induced by H1N1 influenza virus in vivo through reducing the inflammatory responses, alleviating oxidative stress and inhibiting the activation of complement. Thus, anti-complementary activity-guided fractionation of JPWP led to the isolation of an acidic homogeneous polysaccharide, JPWP-PS, whose structure was further elucidated by acid hydrolysis, PMP derivation, methylation and NMR analysis. JPWP-PS had potent anti-complementary activity with the CH50 value of 0.073 ± 0.009 mg/mL, and was characterized by the residues of T-Araf-(1→, →3)-Araf-(1→, →3,5)-Araf-(1→, →3)-Galp-(1→ and →4)-GalpA-(1→.
Collapse
Affiliation(s)
- Zelong Fu
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Long Xia
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Ji De
- College of Science, Tibet University, Lhasa 850000, Tibet, PR China
| | - Mengxia Zhu
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Hong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Yan Lu
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| | - Daofeng Chen
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| |
Collapse
|
20
|
Serum Amyloid P and IgG Exhibit Differential Capabilities in the Activation of the Innate Immune System in Response to Bacillus anthracis Peptidoglycan. Infect Immun 2018. [PMID: 29531132 PMCID: PMC5913848 DOI: 10.1128/iai.00076-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
We showed that human IgG supported the response by human innate immune cells to peptidoglycan (PGN) from Bacillus anthracis and PGN-induced complement activation. However, other serum constituents have been shown to interact with peptidoglycan, including the IgG-like soluble pattern recognition receptor serum amyloid P (SAP). Here, we compared the abilities of SAP and of IgG to support monocyte and complement responses to PGN. Utilizing in vitro methods, we demonstrate that SAP is superior to IgG in supporting monocyte production of cytokines in response to PGN. Like IgG, the response supported by SAP was enhanced by phagocytosis and signaling kinases, such as Syk, Src, and phosphatidylinositol 3-kinase, that are involved in various cellular processes, including Fc receptor signaling. Unlike IgG, SAP had no effect on the activation of complement in response to PGN. These data demonstrate an opsonophagocytic role for SAP in response to PGN that propagates a cellular response without propagating the formation of the terminal complement complex.
Collapse
|
21
|
Karsten CM, Beckmann T, Holtsche MM, Tillmann J, Tofern S, Schulze FS, Heppe EN, Ludwig RJ, Zillikens D, König IR, Köhl J, Schmidt E. Tissue Destruction in Bullous Pemphigoid Can Be Complement Independent and May Be Mitigated by C5aR2. Front Immunol 2018; 9:488. [PMID: 29599777 PMCID: PMC5862877 DOI: 10.3389/fimmu.2018.00488] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 02/23/2018] [Indexed: 12/12/2022] Open
Abstract
Bullous pemphigoid (BP), the most frequent autoimmune bullous disorder, is a paradigmatic autoantibody-mediated disease associated with autoantibodies against BP180 (type XVII collagen, Col17). Several animal models have been developed that reflect important clinical and immunological features of human BP. Complement activation has been described as a prerequisite for blister formation, however, the recent finding that skin lesions can be induced by anti-Col17 F(ab')2 fragments indicates complement-independent mechanisms to contribute to blister formation in BP. Here, C5-/- mice injected with anti-Col17 IgG showed a reduction of skin lesions by about 50% associated with significantly less skin-infiltrating neutrophils compared to wild-type mice. Reduction of skin lesions and neutrophil infiltration was seen independently of the employed anti-Col17 IgG dose. Further, C5ar1-/- mice were protected from disease development, whereas the extent of skin lesions was increased in C5ar2-/- animals. Pharmacological inhibition of C5a receptor 1 (C5aR1) by PMX53 led to reduced disease activity when applied in a prophylactic setting. In contrast, PMX-53 treatment had no effect when first skin lesions had already developed. While C5aR1 was critically involved in neutrophil migration in vitro, its role for Col17-anti-Col17 IgG immune complex-mediated release of reactive oxygen species from neutrophils was less pronounced. Our data demonstrate that complement-dependent and -independent mechanisms coexist in anti-Col17-autoantibody-mediated tissue destruction. C5aR1 and C5aR2 seem to play opposing roles in this process with C5aR1 exerting its primary effect in recruiting inflammatory cells to the skin during the early phase of the disease. Further studies are required to fully understand the role of C5aR2 in autoantibody-mediated skin inflammation.
Collapse
MESH Headings
- Animals
- Autoantibodies/genetics
- Autoantibodies/immunology
- Autoantigens/genetics
- Autoantigens/immunology
- Complement C5/genetics
- Complement C5/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neutrophil Infiltration
- Neutrophils/immunology
- Neutrophils/pathology
- Non-Fibrillar Collagens/genetics
- Non-Fibrillar Collagens/immunology
- Pemphigoid, Bullous/chemically induced
- Pemphigoid, Bullous/genetics
- Pemphigoid, Bullous/immunology
- Pemphigoid, Bullous/pathology
- Peptides, Cyclic/pharmacology
- Reactive Oxygen Species/immunology
- Receptor, Anaphylatoxin C5a/antagonists & inhibitors
- Receptor, Anaphylatoxin C5a/genetics
- Receptor, Anaphylatoxin C5a/immunology
- Skin/immunology
- Skin/pathology
- Collagen Type XVII
Collapse
Affiliation(s)
| | - Tina Beckmann
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | | | - Jenny Tillmann
- Institute of Systemic Inflammation, University of Lübeck, Lübeck, Germany
| | - Sabrina Tofern
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Franziska S. Schulze
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Eva Nina Heppe
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Detlef Zillikens
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Inke R. König
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Jörg Köhl
- Institute of Systemic Inflammation, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital and College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| |
Collapse
|
22
|
Li Z, Wang X, Chen M, Wang Y, Sun R, Qu H, Sun Y, Gao W, Li B, Dong X, Zhang Y, Xia Z. Effectiveness of C5a aptamers in a TNBS-induced colitis mouse model. Exp Ther Med 2017; 14:6119-6124. [PMID: 29285167 DOI: 10.3892/etm.2017.5277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022] Open
Abstract
The complement-activated product, complement component 5a (C5a), is a potent inflammatory peptide with a broad spectrum of functions. In vivo and in vitro studies have demonstrated that C5a serves an important role in inflammation; however, the role of C5a in the pathogenesis of inflammatory bowel disease (IBD) is not known. The purpose of the current study was to investigate the role of C5a in IBD using an experimental mouse model of colitis. Colitis was induced in mice using 2,4,6-trinitrobenzene sulfonic acid (TNBS), and C5a aptamers were subsequently administered via intraperitoneal injection. Clinical symptoms of the disease, histopathological analysis of the colon and the level of inflammatory components were examined. The symptoms of colitis, including changes in behavior, weight loss, colon damage and an increase in inflammatory cytokines, were attenuated following the treatment of mice with TNBS-induced colitis with C5a aptamers. The aptamer-treated mice exhibited a marked attenuation of colitis when compared with untreated mice, as demonstrated by the phenotypic observations, histological examinations and inflammatory cytokine levels. Colitis is characterized by an imbalance between pro-inflammatory and anti-inflammatory mediators. The results of the current study suggest that C5a may serve a critical role in inflammation in IBD.
Collapse
Affiliation(s)
- Zhiping Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Xiwen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Man Chen
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Yuanyuan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Rui Sun
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Han Qu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Yu Sun
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Weicun Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Bo Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Xiaolin Dong
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| | - Yandong Zhang
- Department of Rheumatology, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhiping Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Military Veterinary Institute, Academy of Military Medical Sciences, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
23
|
Karsten CM, Wiese AV, Mey F, Figge J, Woodruff TM, Reuter T, Scurtu O, Kordowski A, Almeida LN, Briukhovetska D, Quell KM, Sun J, Ender F, Schmudde I, Vollbrandt T, Laumonnier Y, Köhl J. Monitoring C5aR2 Expression Using a Floxed tdTomato-C5aR2 Knock-In Mouse. THE JOURNAL OF IMMUNOLOGY 2017; 199:3234-3248. [PMID: 28864475 DOI: 10.4049/jimmunol.1700710] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/10/2017] [Indexed: 12/23/2022]
Abstract
The biological significance of C5a receptor [(C5aR)2/C5L2], a seven-transmembrane receptor binding C5a and C5adesArg, remains ill-defined. Specific ligation of C5aR2 inhibits C5a-induced ERK1/2 activation, strengthening the view that C5aR2 regulates C5aR1-mediated effector functions. Although C5aR2 and C5aR1 are often coexpressed, a detailed picture of C5aR2 expression in murine cells and tissues is still lacking. To close this gap, we generated a floxed tandem dye (td)Tomato-C5aR2 knock-in mouse that we used to track C5aR2 expression in tissue-residing and circulating immune cells. We found the strongest C5aR2 expression in the brain, bone marrow, and airways. All myeloid-derived cells expressed C5aR2, although with different intensities. C5aR2 expression in blood and tissue neutrophils was strong and homogeneous. Specific ligation of C5aR2 in neutrophils from tdTomato-C5aR2 mice blocked C5a-driven ERK1/2 phosphorylation, demonstrating functionality of C5aR2 in the reporter mice. In contrast to neutrophils, we found tissue-specific differences in C5aR2 expression in eosinophils, macrophages, and dendritic cell subsets. Naive and activated T cells stained negative for C5aR2, whereas B cells from different tissues homogeneously expressed C5aR2. Also, NK cell subsets in blood and spleen strongly expressed C5aR2. Activation of C5aR2 in NK cells suppressed IL-12/IL-18-induced IFN-γ production. Intratracheal IL-33 challenge resulted in decreased C5aR2 expression in pulmonary eosinophils and monocyte-derived dendritic cells. In summary, we provide a detailed map of murine C5aR2 immune cell expression in different tissues under steady-state conditions and upon pulmonary inflammation. The C5aR2 knock-in mouse will help to reliably track and conditionally delete C5aR2 expression in experimental models of inflammation.
Collapse
Affiliation(s)
- Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany;
| | - Anna V Wiese
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Fabian Mey
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Julia Figge
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Tom Reuter
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Olga Scurtu
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Anna Kordowski
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Larissa N Almeida
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Daria Briukhovetska
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Katharina M Quell
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Jing Sun
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Inken Schmudde
- Institute of Anatomy, University of Lübeck, Lübeck 23562, Germany
| | - Tillman Vollbrandt
- Cell Analysis Core Facility, University of Lübeck, Lübeck 23562, Germany; and
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck 23562, Germany; .,Division of Immunobiology, Cincinnati Children's Hospital and College of Medicine, University of Cincinnati, Cincinnati, OH 45229
| |
Collapse
|
24
|
Hajishengallis G, Lambris JD. More than complementing Tolls: complement-Toll-like receptor synergy and crosstalk in innate immunity and inflammation. Immunol Rev 2017; 274:233-244. [PMID: 27782328 DOI: 10.1111/imr.12467] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Complement and Toll-like receptors (TLRs) play key roles in the host immune response and are swiftly activated by infection or other types of immunological stress. This review focuses on the capacity of complement and TLRs to engage in signaling crosstalk, ostensibly to coordinate immune and inflammatory responses through synergistic or antagonistic (regulatory) interactions. However, overactivation or dysregulation of either system may lead-often synergistically-to exaggerated inflammation and host tissue injury. Intriguingly, moreover, certain pathogens can manipulate complement-TLR crosstalk pathways in ways that undermine host immunity and favor their persistence. In the setting of polymicrobial inflammatory disease, subversion of complement-TLR crosstalk by keystone pathogens can promote dysbiosis. Knowledge of the molecular mechanisms underlying complement-TLR crosstalk pathways can, therefore, be used productively for tailored therapeutic approaches, such as, to enhance host immunity, mitigate destructive inflammation, or counteract microbial subversion of the host response.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Microbiology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA.
| | - John D Lambris
- Perelman School of Medicine, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
25
|
Shahar E, Gorodetsky R, Aizenshtein E, Lalush L, Pitcovski J. Modulating the innate immune activity in murine tumor microenvironment by a combination of inducer molecules attached to microparticles. Cancer Immunol Immunother 2015; 64:1137-49. [PMID: 26031575 PMCID: PMC11028936 DOI: 10.1007/s00262-015-1719-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 05/21/2015] [Indexed: 12/15/2022]
Abstract
Targeted cancer immunotherapy is challenging due to the cellular diversity and imposed immune tolerance in the tumor microenvironment (TME). A promising route to overcome those drawbacks may be by activating innate immune cells (IIC) in the TME, toward tumor destruction. Studies have shown the ability to "re-educate" pro-tumor-activated IIC toward antitumor responses. The current research aims to stimulate such activation using a combination of innate activators loaded onto microparticles (MP). Four inducers of Toll-like receptors 4 and 7, complement C5a receptor (C5aR) and gamma Fc receptor and their combinations were loaded on MP, and their influence on immune cell activation evaluated. MP stimulation of immune cell activation was tested in vitro and in vivo using a subcutaneous B16-F10 melanoma model induced in C57BL6 mice. Exposure to the TLR4 ligand lipopolysaccharide (LPS) bound to MP-induced acute inflammatory cytokine and chemokine activity in vitro and in vivo, with the elevation of CD45(+) leukocytes in particular GR-1(+) neutrophils and F4/80 macrophages in the TME. Nevertheless, LPS alone on MP was insufficient to significantly delay tumor progression. LPS combined with the C5aR ligand C5a-pep on the same MP resulted in a similar inflammation activation pattern. However, interleukin-10 levels were lower, and tumor growth was significantly delayed. Mixtures of these two ligands on separate MP did not yield the same cytokine activation pattern, demonstrating the importance of the cells' dual activation. The results suggest that combining inducers of distinct innate immune activation pathways holds promise for successful redirection of TME-residing IIC toward anti-tumoral activation.
Collapse
Affiliation(s)
- Ehud Shahar
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
- Lab of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah – Hebrew University Medical Center, Jerusalem, Israel
| | - Raphael Gorodetsky
- Lab of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah – Hebrew University Medical Center, Jerusalem, Israel
| | - Elina Aizenshtein
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
| | - Lior Lalush
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
- Tel Hai Academic College, Upper Galilee, Israel
| | - Jacob Pitcovski
- MIGAL – Galilee Research Institute, P.O. Box 831, 11016 Kiryat Shmona, Israel
- Tel Hai Academic College, Upper Galilee, Israel
| |
Collapse
|
26
|
Sogkas G, Vögtle T, Rau E, Gewecke B, Stegner D, Schmidt RE, Nieswandt B, Gessner JE. Orai1 controls C5a-induced neutrophil recruitment in inflammation. Eur J Immunol 2015; 45:2143-53. [PMID: 25912155 DOI: 10.1002/eji.201445337] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 03/05/2015] [Accepted: 04/25/2015] [Indexed: 11/12/2022]
Abstract
Stromal interaction molecule 1 (STIM1)-dependent store operated calcium-entry (SOCE) through Orai1-mediated calcium (Ca(2+) ) influx is considered a major pathway of Ca(2+) signaling, serving T-cell, mast cell, and platelet responses. Here, we show that Orai1 is critical for neutrophil function. Orai1-deficient neutrophils present defects in fMLP and complement C5a-induced Ca(2+) influx and migration, although they respond normally to another chemoattractant, CXCL2. Up until now, no specific contribution of Orai1 independent from STIM1 or SOCE has been recognized in immune cells. Here, we observe that Orai1-deficient neutrophils exhibit normal STIM1-dependent SOCE and STIM1-deficient neutrophils respond to fMLP and C5a efficiently. Despite substantial cytokine production, Orai1(-/-) chimeric mice show impaired neutrophil recruitment in LPS-induced peritonitis. Moreover, Orai1 deficiency results in profoundly defective C5a-triggered neutrophil lung recruitment in hypersensitivity pneumonitis. Comparative evaluation of inflammation in Stim1(-/-) chimeras reveals a distinct pathogenic contribution of STIM1, including its involvement in IgG-induced C5a production. Our data establish Orai1 as key signal mediator of C5aR activation, contributing to inflammation by a STIM1-independent pathway of Ca(2+) -influx in neutrophils.
Collapse
Affiliation(s)
- Georgios Sogkas
- Clinical Department of Immunology and Rheumatology, Hannover Medical School, Germany
| | - Timo Vögtle
- Chair of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | - Eduard Rau
- Clinical Department of Immunology and Rheumatology, Hannover Medical School, Germany
| | - Britta Gewecke
- Clinical Department of Immunology and Rheumatology, Hannover Medical School, Germany
| | - David Stegner
- Chair of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | - Reinhold E Schmidt
- Clinical Department of Immunology and Rheumatology, Hannover Medical School, Germany
| | - Bernhard Nieswandt
- Chair of Experimental Biomedicine, University Hospital and Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, University of Würzburg, Germany
| | - J Engelbert Gessner
- Clinical Department of Immunology and Rheumatology, Hannover Medical School, Germany
| |
Collapse
|
27
|
Karsten CM, Laumonnier Y, Eurich B, Ender F, Bröker K, Roy S, Czabanska A, Vollbrandt T, Figge J, Köhl J. Monitoring and cell-specific deletion of C5aR1 using a novel floxed GFP-C5aR1 reporter knock-in mouse. THE JOURNAL OF IMMUNOLOGY 2015; 194:1841-55. [PMID: 25589074 DOI: 10.4049/jimmunol.1401401] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Many of the biological properties of C5a are mediated through activation of its receptor (C5aR1), the expression of which has been demonstrated convincingly on myeloid cells, such as neutrophils, monocytes, and macrophages. In contrast, conflicting results exist regarding C5aR1 expression in dendritic cells (DCs) and lymphoid lineage cells. In this article, we report the generation of a floxed GFP-C5aR1 reporter knock-in mouse. Using this mouse strain, we confirmed strong C5aR1 expression in neutrophils from bone marrow, blood, lung, and spleen, as well as in peritoneal macrophages. Further, we show C5aR1 expression in lung eosinophils, lung- and lamina propria-resident and alveolar macrophages, bone marrow-derived DCs, and lung-resident CD11b(+) and monocyte-derived DCs, whereas intestinal and pulmonary CD103(+) DCs stained negative. Also, some splenic NKT cells expressed GFP, whereas naive NK cells and B2 cells lacked GFP expression. Finally, we did not observe any C5aR1 expression in naive or activated CD4(+) Th cells in vitro or in vivo. Mating the floxed GFP-C5aR1 mouse strain with LysMCre mice, we were able to specifically delete C5aR1 in neutrophils and macrophages, whereas C5aR1 expression was retained in DCs. In summary, our findings suggest that C5aR1 expression in mice is largely restricted to cells of the myeloid lineage. The novel floxed C5aR1 reporter knock-in mouse will prove useful to track C5aR1 expression in experimental models of acute and chronic inflammation and to conditionally delete C5aR1 in immune cells.
Collapse
Affiliation(s)
- Christian M Karsten
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Benjamin Eurich
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Katharina Bröker
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Sreeja Roy
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Anna Czabanska
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | | | - Julia Figge
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, 23538 Lübeck, Germany; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| |
Collapse
|
28
|
Abstract
Every year, more cases of sepsis appear in intensive care units. The most frequent complication of sepsis is septic encephalopathy (SE), which is also the essential determinant of mortality. Despite many years of research, it still is not known at which stage of sepsis the first signs of SE appear; however, it is considered the most frequent form of encephalopathy. Patients have dysfunction of cognitive abilities and consciousness, and sometimes even epileptic seizures. Despite intensive treatment, the effects of SE remain for many years and constitute an important social problem. Numerous studies indicate that changes in the brain involve free radicals, nitric oxide, increased synthesis of inflammatory factors, disturbances in cerebral circulation, microthromboses, and ischemia, which cause considerable neuronal destruction in different areas of the brain. To determine at what point during sepsis the first signs of SE appear, different experimental models are needed to detect the aforementioned changes and to select the proper therapy for this syndrome.
Collapse
Affiliation(s)
- Marek Ziaja
- Department of Histology, Jagiellonian University Medical College, Kraków, Poland.
| |
Collapse
|
29
|
Abstract
Sepsis associated encephalopathy (SAE) is a common but poorly understood neurological complication of sepsis. It is characterized by diffuse brain dysfunction secondary to infection elsewhere in the body without overt CNS infection. The pathophysiology of SAE is complex and multifactorial including a number of intertwined mechanisms such as vascular damage, endothelial activation, breakdown of the blood brain barrier, altered brain signaling, brain inflammation, and apoptosis. Clinical presentation of SAE may range from mild symptoms such as malaise and concentration deficits to deep coma. The evaluation of cognitive dysfunction is made difficult by the absence of any specific investigations or biomarkers and the common use of sedation in critically ill patients. SAE thus remains diagnosis of exclusion which can only be made after ruling out other causes of altered mentation in a febrile, critically ill patient by appropriate investigations. In spite of high mortality rate, management of SAE is limited to treatment of the underlying infection and symptomatic treatment for delirium and seizures. It is important to be aware of this condition because SAE may present in early stages of sepsis, even before the diagnostic criteria for sepsis can be met. This review discusses the diagnostic approach to patients with SAE along with its epidemiology, pathophysiology, clinical presentation, and differential diagnosis.
Collapse
|
30
|
Pro-inflammatory effect of cystic fibrosis sputum microparticles in the murine lung. J Cyst Fibros 2013; 12:721-8. [DOI: 10.1016/j.jcf.2013.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 02/20/2013] [Accepted: 03/05/2013] [Indexed: 12/25/2022]
|
31
|
Sun S, Zhao G, Liu C, Wu X, Guo Y, Yu H, Song H, Du L, Jiang S, Guo R, Tomlinson S, Zhou Y. Inhibition of complement activation alleviates acute lung injury induced by highly pathogenic avian influenza H5N1 virus infection. Am J Respir Cell Mol Biol 2013; 49:221-30. [PMID: 23526211 DOI: 10.1165/rcmb.2012-0428oc] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The acute lung injury (ALI) that occurs after the highly pathogenic avian influenza H5N1 virus infection is associated with an abnormal host innate immune response. Because the complement system plays a central role in innate immunity and because aberrant complement activation is associated with a variety of autoimmune and inflammatory diseases, we investigated the complement involvement in the pathogenesis of ALI induced by H5N1 virus infection. We showed that ALI in H5N1-infected mice was caused by excessive complement activation, as demonstrated by deposition of C3, C5b-9, and mannose-binding lectin (MBL)-C in lung tissue, and by up-regulation of MBL-associated serine protease-2 and the complement receptors C3aR and C5aR. Treatment of H5N1-infected mice with a C3aR antagonist led to significantly reduced inflammation in lungs, alleviating ALI. Furthermore, complement inhibition with an anti-C5a antibody or complement depletion with cobra venom factor after H5N1 challenge resulted in a similar level of protection to that seen in C3aR antagonist-treated mice. These results indicate that excessive complement activation plays an important role in mediating H5N1-induced ALI and that inhibition of complement may be an effective clinical intervention and adjunctive treatment for H5N1-induced ALI.
Collapse
Affiliation(s)
- Shihui Sun
- 1 State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Biofilms production is a central feature of nosocomial infection of catheters and other medical devices used in resuscitation and critical care. However, the very effective biofilm forming pathogen Staphylococcus epidermidis often produces a modest host inflammatory response and few of the signs and symptoms associated with more virulent pathogens. To examine the impact of bacterial biofilm formation on provocation of an innate immune response, we studied the elaboration of the major complement anaphylatoxin C5a by human serum upon contact with S. epidermidis biofilms. Wild-type S. epidermidis and mutants of sarA (a regulatory protein that promotes synthesis of the biofilm-forming polysaccharide intercellular adhesin [PIA]) and icaB (responsible for postexport processing of PIA) were studied. C5a release, as a function of exposed biofilm surface area, was on the order of 1 fmol · cm · s and was dependent on the presence of PIA. Experimental results were used to inform a physiologically based pharmacokinetic model of C5a release by an infected central venous catheter, one of S. epidermidis' primary means of causing human disease. These simulations revealed that the magnitude of C5a release on a superior vena cava catheter completely covered with S. epidermidis would be lower than necessary to alert circulating leukocytes. Combined, the experimental and computational results are highly consistent with clinical observations in which the clinical signs of central line-associated bloodstream infection are often muted in association with this important pathogen.
Collapse
|
33
|
Kumar V, Sharma A. Innate Immunity in Sepsis Pathogenesis and Its Modulation: New Immunomodulatory Targets Revealed. J Chemother 2013; 20:672-83. [DOI: 10.1179/joc.2008.20.6.672] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
34
|
Leslie JD, Mayor R. Complement in animal development: unexpected roles of a highly conserved pathway. Semin Immunol 2013; 25:39-46. [PMID: 23665279 PMCID: PMC3989114 DOI: 10.1016/j.smim.2013.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 04/13/2013] [Indexed: 12/16/2022]
Abstract
The complement pathway is most famous for its role in immunity, orchestrating an exquisitely refined system for immune surveillance. At its core lies a cascade of proteolytic events that ultimately serve to recognise microbes, infected cells or debris and target them for elimination. Mounting evidence has shown that a number of the proteolytic intermediaries in this cascade have, in themselves, other functions in the body, signalling through receptors to drive events that appear to be unrelated to immune surveillance. It seems, then, that the complement system not only functions as an immunological effector, but also has cell-cell signalling properties that are utilised by a number of non-immunological processes. In this review we examine a number of these processes in the context of animal development, all of which share a requirement for precise control of cell behaviour in time and space. As we will see, the scope of the complement system's function is indeed much greater than we might have imagined only a few years ago.
Collapse
Affiliation(s)
- Jonathan D Leslie
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | | |
Collapse
|
35
|
Packiriswamy N, Lee T, Raghavendra PB, Durairaj H, Wang H, Parameswaran N. G-protein-coupled receptor kinase-5 mediates inflammation but does not regulate cellular infiltration or bacterial load in a polymicrobial sepsis model in mice. J Innate Immun 2013; 5:401-13. [PMID: 23485819 DOI: 10.1159/000347002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2012] [Accepted: 01/07/2013] [Indexed: 01/05/2023] Open
Abstract
NFκB-dependent signaling is an important modulator of inflammation in several diseases including sepsis. G-protein-coupled receptor kinase-5 (GRK5) is an evolutionarily conserved regulator of the NFκB pathway. We hypothesized that GRK5 via NFκB regulation plays an important role in the pathogenesis of sepsis. To test this we utilized a clinically relevant polymicrobial sepsis model in mice that were deficient in GRK5. We subjected wild-type (WT) and GRK5 knockout (KO) mice to cecal ligation and puncture (CLP)-induced polymicrobial sepsis and assessed the various events in sepsis pathogenesis. CLP induced a significant inflammatory response in the WT and this was markedly attenuated in the KO mice. To determine the signaling mechanisms and the role of NFκB activation in sepsis-induced inflammation, we assessed the levels of IκBα phosphorylation and expression of NFκB-dependent genes in the liver in the two genotypes. Both IκBα phosphorylation and gene expression were significantly inhibited in the GRK5 KO compared to the WT mice. Interestingly, however, GRK5 did not modulate either immune cell infiltration (to the primary site of infection) or local/systemic bacterial load subsequent to sepsis induction. In contrast GRK5 deficiency significantly inhibited sepsis-induced plasma corticosterone levels and the consequent thymocyte apoptosis in vivo. Associated with these outcomes, CLP-induced mortality was significantly prevented in the GRK5 KO mice in the presence of antibiotics. Together, our studies demonstrate that GRK5 is an important regulator of inflammation and thymic apoptosis in polymicrobial sepsis and implicate GRK5 as a potential molecular target in sepsis.
Collapse
Affiliation(s)
- Nandakumar Packiriswamy
- Division of Human Pathology, Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | | | | | | | | | | |
Collapse
|
36
|
Unnewehr H, Rittirsch D, Sarma JV, Zetoune F, Flierl MA, Perl M, Denk S, Weiss M, Schneider ME, Monk PN, Neff T, Mihlan M, Barth H, Gebhard F, Ward PA, Huber-Lang M. Changes and regulation of the C5a receptor on neutrophils during septic shock in humans. THE JOURNAL OF IMMUNOLOGY 2013; 190:4215-25. [PMID: 23479227 DOI: 10.4049/jimmunol.1200534] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During experimental sepsis, excessive generation of the anaphylatoxin C5a results in reduction of the C5a receptor (C5aR) on neutrophils. These events have been shown to result in impaired innate immunity. However, the regulation and fate of C5aR on neutrophils during sepsis are largely unknown. In contrast to 30 healthy volunteers, 60 patients in septic shock presented evidence of complement activation with significantly increased serum levels of C3a, C5a, and C5b-9. In the septic shock group, the corresponding decrease in complement hemolytic activity distinguished survivors from nonsurvivors. Neutrophils from patients in septic shock exhibited decreased C5aR expression, which inversely correlated with serum concentrations of C-reactive protein (CRP) and clinical outcome. In vitro exposure of normal neutrophils to native pentameric CRP led to a dose- and time-dependent loss of C5aR expression on neutrophils, whereas the monomeric form of CRP, as well as various other inflammatory mediators, failed to significantly alter C5aR levels on neutrophils. A circulating form of C5aR (cC5aR) was detected in serum by immunoblotting and a flow-based capture assay, suggestive of an intact C5aR molecule. Levels of cC5aR were significantly enhanced during septic shock, with serum levels directly correlating with lethality. The data suggest that septic shock in humans is associated with extensive complement activation, CRP-dependent loss of C5aR on neutrophils, and appearance of cC5aR in serum, which correlated with a poor outcome. Therefore, cC5aR may represent a new sepsis marker to be considered in tailoring individualized immune-modulating therapy.
Collapse
Affiliation(s)
- Heike Unnewehr
- Department of Orthopedic Trauma, Hand-, Plastic-, and Reconstructive Surgery, University Hospital Ulm, 89081 Ulm, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
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: 3.1] [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
|
38
|
Abstract
Marked alterations of the innate and adaptive immune response follow invasive infection and generalized inflammatory states. If left unchecked, this state of immune dysregulation contributes to a myriad of maladaptive cellular responses that culminate in multiple organ dysfunction, septic shock, and lethality. The molecular details of the cell-signaling networks that underlie the pathophysiology of systemic inflammation and sepsis are now increasingly well understood. While a vigorous and effective immune response to invasive pathogens is essential for microbial clearance and host survival, nonresolving, generalized inflammation can induce diffuse endovascular damage, increased capillary permeability, coagulopathy, and widespread tissue damage. Current evidence indicates that a state of relative immune suppression often accompanies sepsis and might provide novel therapeutic options in some patients. An expanding number of potential therapeutic options are now in clinical development to reestablish control and promote resolution over sepsis-induced systemic inflammation and organ dysfunction.
Collapse
Affiliation(s)
- Steven P Larosa
- Division of Infectious Disease, Beverly Hospital, 85 Herrick Street, Beverly, MA, 01915, USA
| | | |
Collapse
|
39
|
Bogie JFJ, Timmermans S, Huynh-Thu VA, Irrthum A, Smeets HJM, Gustafsson JÅ, Steffensen KR, Mulder M, Stinissen P, Hellings N, Hendriks JJA. Myelin-derived lipids modulate macrophage activity by liver X receptor activation. PLoS One 2012; 7:e44998. [PMID: 22984598 PMCID: PMC3440367 DOI: 10.1371/journal.pone.0044998] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 08/15/2012] [Indexed: 11/19/2022] Open
Abstract
Multiple sclerosis is a chronic, inflammatory, demyelinating disease of the central nervous system in which macrophages and microglia play a central role. Foamy macrophages and microglia, containing degenerated myelin, are abundantly found in active multiple sclerosis lesions. Recent studies have described an altered macrophage phenotype after myelin internalization. However, it is unclear by which mechanisms myelin affects the phenotype of macrophages and how this phenotype can influence lesion progression. Here we demonstrate, by using genome wide gene expression analysis, that myelin-phagocytosing macrophages have an enhanced expression of genes involved in migration, phagocytosis and inflammation. Interestingly, myelin internalization also induced the expression of genes involved in liver-X-receptor signaling and cholesterol efflux. In vitro validation shows that myelin-phagocytosing macrophages indeed have an increased capacity to dispose intracellular cholesterol. In addition, myelin suppresses the secretion of the pro-inflammatory mediator IL-6 by macrophages, which was mediated by activation of liver-X-receptor β. Our data show that myelin modulates the phenotype of macrophages by nuclear receptor activation, which may subsequently affect lesion progression in demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Jeroen F J Bogie
- Hasselt University/Transnational University Limburg, Biomedical Research Institute, School of Life Sciences, Diepenbeek, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
Complement C5a: Impact on the field of veterinary medicine. Vet J 2012; 192:264-71. [DOI: 10.1016/j.tvjl.2011.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 11/30/2011] [Accepted: 12/14/2011] [Indexed: 01/03/2023]
|
42
|
Genetic analysis of C5a receptors in neutrophils from patients with familial Mediterranean fever. Mol Biol Rep 2011; 39:5503-10. [PMID: 22187344 DOI: 10.1007/s11033-011-1353-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 12/12/2011] [Indexed: 01/09/2023]
Abstract
Familial Mediterranean fever (FMF) is an autoinflammatory disease, characterized by MEFV gene mutations and self-limited recurrent episodes of fever and localized serositis. Complement system is a key regulator of the inflammatory process. The aim of this study was to investigate the genetic alterations and mRNA expression pattern of C5aR and C5L2 genes in neutrophils from attack-free FMF patients. No mutations were observed in the two receptors' genes, while the genetic alteration observed in the C5aR1 gene was identified as N279 K polymorphic variant. Furthermore, lower mRNA expression of C5L2 gene was observed in neutrophils from FMF patients compared to control subjects. The binding capacity of rhC5a and the ability to produce reactive oxygen species was similar in neutrophils from healthy subjects and FMF patients and independent of the presence of N279 K polymorphism or mRNA expression of C5L2.
Collapse
|
43
|
Loor JJ, Moyes KM, Bionaz M. Functional adaptations of the transcriptome to mastitis-causing pathogens: the mammary gland and beyond. J Mammary Gland Biol Neoplasia 2011; 16:305-22. [PMID: 21968536 DOI: 10.1007/s10911-011-9232-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/12/2011] [Indexed: 02/06/2023] Open
Abstract
Application of microarrays to the study of intramammary infections in recent years has provided a wealth of fundamental information on the transcriptomics adaptation of tissue/cells to the disease. Due to its heavy toll on productivity and health of the animal, in vivo and in vitro transcriptomics works involving different mastitis-causing pathogens have been conducted on the mammary gland, primarily on livestock species such as cow and sheep, with few studies in non-ruminants. However, the response to an infectious challenge originating in the mammary gland elicits systemic responses in the animal and encompasses tissues such as liver and immune cells in the circulation, with also potential effects on other tissues such as adipose. The susceptibility of the animal to develop mastitis likely is affected by factors beyond the mammary gland, e.g. negative energy balance as it occurs around parturition. Objectives of this review are to discuss the use of systems biology concepts for the holistic study of animal responses to intramammary infection; providing an update of recent work using transcriptomics to study mammary and peripheral tissue (i.e. liver) as well as neutrophils and macrophage responses to mastitis-causing pathogens; discuss the effect of negative energy balance on mastitis predisposition; and analyze the bovine and murine mammary innate-immune responses during lactation and involution using a novel functional analysis approach to uncover potential predisposing factors to mastitis throughout an animal's productive life.
Collapse
Affiliation(s)
- Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | | | | |
Collapse
|
44
|
Aitken SL, Corl CM, Sordillo LM. Immunopathology of mastitis: insights into disease recognition and resolution. J Mammary Gland Biol Neoplasia 2011; 16:291-304. [PMID: 21938490 DOI: 10.1007/s10911-011-9230-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 09/01/2011] [Indexed: 01/02/2023] Open
Abstract
Mastitis is an inflammation of the mammary gland commonly caused by bacterial infection. The inflammatory process is a normal and necessary immunological response to invading pathogens. The purpose of host inflammatory responses is to eliminate the source of tissue injury, restore immune homeostasis, and return tissues to normal function. The inflammatory cascade results not only in the escalation of local antimicrobial factors, but also in the increased movement of leukocytes and plasma components from the blood that may cause damage to host tissues. A precarious balance between pro-inflammatory and pro-resolving mechanisms is needed to ensure optimal bacterial clearance and the prompt return to immune homeostasis. Therefore, inflammatory responses must be tightly regulated to avoid bystander damage to the milk synthesizing tissues of the mammary gland. The defense mechanisms of the mammary gland function optimally when invading bacteria are recognized promptly, the initial inflammatory response is adequate to rapidly eliminate the infection, and the mammary gland is returned to normal function quickly without any noticeable clinical symptoms. Suboptimal or dysfunctional mammary gland defenses, however, may contribute to the development of severe acute inflammation or chronic mastitis that adversely affects the quantity and quality of milk. This review will summarize critical mammary gland defense mechanisms that are necessary for immune surveillance and the rapid elimination of mastitis-causing organisms. Situations in which diminished efficiency of innate or adaptive mammary gland immune responses may contribute to disease pathogenesis will also be discussed. A better understanding of the complex interactions between mammary gland defenses and mastitis-causing pathogens should prove useful for the future control of intramammary infections.
Collapse
Affiliation(s)
- Stacey L Aitken
- G300 Veterinary Medical Center, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| | | | | |
Collapse
|
45
|
Sun S, Guo Y, Zhao G, Zhou X, Li J, Hu J, Yu H, Chen Y, Song H, Qiao F, Xu G, Yang F, Wu Y, Tomlinson S, Duan Z, Zhou Y. Complement and the alternative pathway play an important role in LPS/D-GalN-induced fulminant hepatic failure. PLoS One 2011; 6:e26838. [PMID: 22069473 PMCID: PMC3206060 DOI: 10.1371/journal.pone.0026838] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 10/04/2011] [Indexed: 01/18/2023] Open
Abstract
Fulminant hepatic failure (FHF) is a clinically severe type of liver injury with an extremely high mortality rate. Although the pathological mechanisms of FHF are not well understood, evidence suggests that the complement system is involved in the pathogenesis of a variety of liver disorders. In the present study, to investigate the role of complement in FHF, we examined groups of mice following intraperitoneal injection of LPS/D-GalN: wild-type C57BL/6 mice, wild-type mice treated with a C3aR antagonist, C5aR monoclonal antibody (C5aRmAb) or CR2-Factor H (CR2-fH, an inhibitor of the alternative pathway), and C3 deficient mice (C3⁻/⁻ mice). The animals were euthanized and samples analyzed at specific times after LPS/D-GalN injection. The results show that intraperitoneal administration of LPS/D-GalN activated the complement pathway, as evidenced by the hepatic deposition of C3 and C5b-9 and elevated serum levels of the complement activation product C3a, the level of which was associated with the severity of the liver damage. C3a receptor (C3aR) and C5a receptor (C5aR) expression was also upregulated. Compared with wild-type mice, C3⁻/⁻ mice survived significantly longer and displayed reduced liver inflammation and attenuated pathological damage following LPS/D-GalN injection. Similar levels of protection were seen in mice treated with C3aR antagonist,C5aRmAb or CR2-fH. These data indicate an important role for the C3a and C5a generated by the alternative pathway in LPS/D-GalN-induced FHF. The data further suggest that complement inhibition may be an effective strategy for the adjunctive treatment of fulminant hepatic failure.
Collapse
Affiliation(s)
- Shihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaojun Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Junfeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Jingya Hu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Hong Yu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Yu Chen
- Beijing You-An Hospital, Artificial Liver Center, Capital University of Medical Sciences, Beijing, China
| | - Hongbin Song
- Institute of Disease Control and Prevention, Academy of Military Medical Science, Beijing, China
| | - Fei Qiao
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Guilian Xu
- Institute of Immunology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Fei Yang
- Institute of Immunology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Zhongping Duan
- Beijing You-An Hospital, Artificial Liver Center, Capital University of Medical Sciences, Beijing, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| |
Collapse
|
46
|
Jacob A, Hack B, Chen P, Quigg RJ, Alexander JJ. C5a/CD88 signaling alters blood-brain barrier integrity in lupus through nuclear factor-κB. J Neurochem 2011; 119:1041-51. [PMID: 21929539 DOI: 10.1111/j.1471-4159.2011.07490.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammation is a key factor in a number of neurodegenerative diseases including systemic lupus erythematosus. The complement system is an important mechanism in initiating and amplifying inflammation. Our recent studies demonstrate that C5a, a protein fragment generated during complement activation could alter the blood-brain barrier integrity, and thereby disturb the brain microenvironment. To understand the mechanism by which this occurs, we examined the effects of C5a on apoptosis, translocation of nuclear factor-κB (NF-κb) and the expression of Iκbα, MAPK, CREB and TJ protein, zona occludens (ZO-1) in mouse brain endothelial cells. Apoptosis was examined by DNA laddering and caspase 3 activity and the distribution of the ZO-1 and the p65 subunit of NF-κB were determined by immunofluorescence. Inhibition of CD88 reduced translocation of NF-κb into the nucleus, altered ZO-1 at the interfaces of neighboring cells, decreased caspase 3 activity and prevented apoptosis in these cells. Our results indicate that signaling through CD88 regulates the blood-brain barrier in a NF-κb-dependent manner. These studies suggest that the C5a receptor, CD88 is a promising therapeutic target that will reduce NF-κb-signaling cascades in inflammatory settings.
Collapse
Affiliation(s)
- Alexander Jacob
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | |
Collapse
|
47
|
Raby AC, Holst B, Davies J, Colmont C, Laumonnier Y, Coles B, Shah S, Hall J, Topley N, Köhl J, Morgan BP, Labéta MO. TLR activation enhances C5a-induced pro-inflammatory responses by negatively modulating the second C5a receptor, C5L2. Eur J Immunol 2011; 41:2741-52. [PMID: 21630250 PMCID: PMC3638321 DOI: 10.1002/eji.201041350] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Revised: 05/03/2011] [Accepted: 05/25/2011] [Indexed: 01/07/2023]
Abstract
TLR and complement activation ensures efficient clearance of infection. Previous studies documented synergism between TLRs and the receptor for the pro-inflammatory complement peptide C5a (C5aR/CD88), and regulation of TLR-induced pro-inflammatory responses by C5aR, suggesting crosstalk between TLRs and C5aR. However, it is unclear whether and how TLRs modulate C5a-induced pro-inflammatory responses. We demonstrate a marked positive modulatory effect of TLR activation on cell sensitivity to C5a in vitro and ex vivo and identify an underlying mechanistic target. Pre-exposure of PBMCs and whole blood to diverse TLR ligands or bacteria enhanced C5a-induced pro-inflammatory responses. This effect was not observed in TLR4 signalling-deficient mice. TLR-induced hypersensitivity to C5a did not result from C5aR upregulation or modulation of C5a-induced Ca2+ mobilization. Rather, TLRs targeted another C5a receptor, C5L2 (acting as a negative modulator of C5aR), by reducing C5L2 activity. TLR-induced hypersensitivity to C5a was mimicked by blocking C5L2 and was not observed in C5L2KO mice. Furthermore, TLR activation inhibited C5L2 expression upon C5a stimulation. These findings identify a novel pathway of crosstalk within the innate immune system that amplifies innate host defense at the TLR-complement interface. Unravelling the mutually regulated activities of TLRs and complement may reveal new therapeutic avenues to control inflammation.
Collapse
Affiliation(s)
- Anne-Catherine Raby
- Department of Infection, Immunity and Biochemistry, School of Medicine, Cardiff University, Cardiff, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Manthey HD, Thomas AC, Shiels IA, Zernecke A, Woodruff TM, Rolfe B, Taylor SM. Complement C5a inhibition reduces atherosclerosis in ApoE-/- mice. FASEB J 2011; 25:2447-55. [PMID: 21490292 DOI: 10.1096/fj.10-174284] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The complement C5a receptor, CD88, is present on many of the cells found within human atherosclerotic plaques, but little is known about the role of C5a in atherogenesis. Using real-time PCR, we determined that ApoE(-/-) mice fed a normal diet express more aortic CD88 mRNA compared with controls, and this increase coincides with atherosclerotic lesion development (P<0.001 for 3- vs. 25-wk-old animals). Conversely, mRNA expression of the alternative C5a receptor, C5L2, in aortas of ApoE(-/-) mice, was lower than controls at all time points. Using immunohistochemistry, we confirmed the presence of CD88 on macrophages, smooth muscle cells, and activated endothelial cells in plaques from brachiocephalic arteries. Treatment of ApoE(-/-) mice with a CD88 antagonist (PMX53; 3 mg/kg s.c. 3 ×/wk plus 1 mg/kg/d p.o.) for 25 wk reduced lesion size and lipid content in the plaque by ∼ 40% (P<0.05). Our study provides evidence for a proatherogenic role for C5a and identifies the CD88 antagonist PMX53 as a potential antiatherosclerotic drug.
Collapse
Affiliation(s)
- Helga D Manthey
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | |
Collapse
|
49
|
Sun S, Wang H, Zhao G, An Y, Guo Y, Du L, Song H, Qiao F, Yu H, Wu X, Atkinson C, Jiang S, Tomlinson S, Zhou Y. Complement inhibition alleviates paraquat-induced acute lung injury. Am J Respir Cell Mol Biol 2011; 45:834-42. [PMID: 21421909 DOI: 10.1165/rcmb.2010-0444oc] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The widely used herbicide, paraquat (PQ), is highly toxic and claims thousands of lives from both accidental and voluntary ingestion. The pathological mechanisms of PQ poisoning-induced acute lung injury (ALI) are not well understood, and the role of complement in PQ-induced ALI has not been elucidated. We developed and characterized a mouse model of PQ-induced ALI and studied the role of complement in the pathogenesis of PQ poisoning. Intraperitoneal administration of PQ caused dose- and time-dependent lung damage and mortality, with associated inflammatory response. Within 24 hours of PQ-induced ALI, there was significantly increased expression of the complement proteins, C1q and C3, in the lung. Expression of the anaphylatoxin receptors, C3aR and C5aR, was also increased. Compared with wild-type mice, C3-deficient mice survived significantly longer and displayed significantly reduced lung inflammation and pathology after PQ treatment. Similar reductions in PQ-induced inflammation, pathology, and mortality were recorded in mice treated with the C3 inhibitors, CR2-Crry, and alternative pathway specific CR2-fH. A similar therapeutic effect was also observed by treatment with either C3a receptor antagonist or a blocking C5a receptor monoclonal antibody. Together, these studies indicate that PQ-induced ALI is mediated through receptor signaling by the C3a and C5a complement activation products that are generated via the alternative complement pathway, and that complement inhibition may be an effective clinical intervention for postexposure treatment of PQ-induced ALI.
Collapse
Affiliation(s)
- Shihui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
|