1
|
Zhang K, Honda A, Sagawa T, Miyasaka N, Qiu B, Ishikawa R, Okuda T, Kameda T, Sadakane K, Ichinose T, Takano H. Complement system is activated in acute inflammatory response to environmental particulates in the lungs. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 297:118259. [PMID: 40315749 DOI: 10.1016/j.ecoenv.2025.118259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 04/01/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Short-term exposure (typically ranging from a few hours to 7 days) to particulate matter (PM) elicits acute inflammatory responses with significant activation of complement component C5. However, the relationship between PM-induced complement system activation, acute inflammation, and the contributing factors remains unclear. In this study, we aimed to investigate the intensity of acute inflammatory responses, as well as the activation levels of complement C5 and its related products, C5aR1 and C5b-9, following exposure to different types of PM. Acute inflammatory responses and complement activation were assessed in mice intratracheally administered four types of PM: titanium dioxide (TiO₂), diesel exhaust particles (DEP), Asian sand dust (ASD), and ambient PM with an aerodynamic diameter ≤ 2.5 μm (PM2.5). Complement system and inflammatory markers were evaluated by analyzing increased C5 protein levels, C5b-9 deposition, and C5aR1 expression. Additionally, dark-field microscopy and Raman microscopy were used to detect PM components adjacent to infiltrating neutrophils, and elemental composition was quantified using ICP-MS and EDXRF. ASD, DEP, and PM2.5 exposure significantly increased C5b-9 deposition in lung tissues. All PM-exposed groups exhibited substantial upregulation of C5aR1 expression, primarily in neutrophils. Raman spectroscopic analysis revealed Si, K, Mg, Al, and Fe adjacent to infiltrating neutrophils in ASD-exposed lungs. Furthermore, elemental analysis identified Si, Mg, Al, and K as the most potent contributors to complement activation and inflammatory responses. Of the four types of PM, ASD induced the most severe acute inflammatory response and complement system activation. Therefore, ASD-induced complement system activation, driven at least partly by its mineral components, may play a critical role in neutrophil activation and acute pulmonary inflammation. These findings highlight the differential impact of PM types on complement system activation and underscore the importance of PM composition in the evaluation of air pollution-related health risks, particularly acute pulmonary inflammation.
Collapse
Affiliation(s)
- Kerui Zhang
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Akiko Honda
- Graduate School of Engineering, Kyoto University, Kyoto, Japan.
| | - Tomoya Sagawa
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Natsuko Miyasaka
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Binyang Qiu
- Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Raga Ishikawa
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Tomoaki Okuda
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Takayuki Kameda
- Graduate School of Energy Science, Kyoto University, Kyoto, Japan
| | - Kaori Sadakane
- Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, Japan
| | - Takamichi Ichinose
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita, Japan
| | - Hirohisa Takano
- Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Institute for International Academic Research, Kyoto University of Advanced Science, Kyoto, Japan; Research Institute for Coexistence and Health Science, Kyoto University of Advanced Science, Kyoto, Japan.
| |
Collapse
|
2
|
Seki K, Sueyoshi K, Miyoshi Y, Nakamura Y, Ishihara T, Kondo Y, Kuroda Y, Yonekura A, Iwabuchi K, Okamoto K, Tanaka H. Complement activation and lung injury in Japanese patients with COVID-19: a prospective observational study. Sci Rep 2024; 14:24895. [PMID: 39438600 PMCID: PMC11496683 DOI: 10.1038/s41598-024-76365-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Background The production of inflammatory cytokines is reportedly increased in patients with coronavirus disease 2019 (COVID-19), causing the migration of neutrophils and monocytes to lung tissues. This disrupts the air-blood barrier by damaging the bronchial epithelial and vascular endothelial cells. As multiorgan dysfunction in sepsis is considered to be partly caused by complement activation, which can cause lung injury in patients with COVID-19. There are limited studies examining the link between complement activation in patients with COVID-19. This study aimed to AQinvestigate the association of complement activation with the pathophysiology of COVID-19. Twenty-seven patients with COVID-19 were enrolled in this study and classified into two groups depending on the indication for mechanical ventilation. Plasma complement factors (C3a, C5a, Ba, and sC5b-9), complement regulators (sCD59 and factor H), interleukin-6 (IL-6), and syndecan-1 levels were measured using Enzyme-linked immunosorbent assay (ELISA). Results: All complement factors and regulators, IL-6, and syndecan-1 levels were significantly elevated in patients with COVID-19 compared with those in healthy controls. C5a and sC5b-9 levels were decreased significantly in the invasive mechanical ventilation (IMV) group compared with those in the non-IMV group. Syndecan-1 levels were significantly increased in the IMV group compared with those in the non-IMV group. Conclusions: Complement activation is an exacerbating factor for lung injury in patients with COVID-19. Complement factors are nonessential predictors of mechanical ventilation; however, syndecan-1 could be a biomarker of COVID-19 severity in patients.
Collapse
Affiliation(s)
- Kentaro Seki
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Koichiro Sueyoshi
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan.
| | - Yukari Miyoshi
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Yuki Nakamura
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Tadashi Ishihara
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Yutaka Kondo
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Yoko Kuroda
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Ayumi Yonekura
- Institute for Environmental and Gender Specific Medicine, Juntendo University, Urayasu, Chiba, Japan
| | - Kazuhisa Iwabuchi
- Institute for Environmental and Gender Specific Medicine, Juntendo University, Urayasu, Chiba, Japan
| | - Ken Okamoto
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| | - Hiroshi Tanaka
- Department of Emergency and Critical Care Medicine, Juntendo University Urayasu Hospital, Urayasu, Chiba, Japan
| |
Collapse
|
3
|
Mrozewski L, Tharmalingam S, Michael P, Kumar A, Tai TC. C5a Induces Inflammatory Signaling and Apoptosis in PC12 Cells through C5aR-Dependent Signaling: A Potential Mechanism for Adrenal Damage in Sepsis. Int J Mol Sci 2024; 25:10673. [PMID: 39409001 PMCID: PMC11477224 DOI: 10.3390/ijms251910673] [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: 08/14/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
The complement system is critically involved in the pathogenesis of sepsis. In particular, complement anaphylatoxin C5a is generated in excess during sepsis, leading to cellular dysfunction. Recent studies have shown that excessive C5a impairs adrenomedullary catecholamine production release and induces apoptosis in adrenomedullary cells. Currently, the mechanisms by which C5a impacts adrenal cell function are poorly understood. The PC12 cell model was used to examine the cellular effects following treatment with recombinant rat C5a. The levels of caspase activation and cell death, protein kinase signaling pathway activation, and changes in inflammatory protein expression were examined following treatment with C5a. There was an increase in apoptosis of PC12 cells following treatment with high-dose C5a. Ten inflammatory proteins, primarily involved in apoptosis, cell survival, and cell proliferation, were upregulated following treatment with high-dose C5a. Five inflammatory proteins, involved primarily in chemotaxis and anti-inflammatory functions, were downregulated. The ERK/MAPK, p38/MAPK, JNK/MAPK, and AKT protein kinase signaling pathways were upregulated in a C5aR-dependent manner. These results demonstrate an apoptotic effect and cellular signaling effect of high-dose C5a. Taken together, the overall data suggest that high levels of C5a may play a role in C5aR-dependent apoptosis of adrenal medullary cells in sepsis.
Collapse
Affiliation(s)
- Lucas Mrozewski
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
| | - Sujeenthar Tharmalingam
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| | - Paul Michael
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
| | - Aseem Kumar
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| | - T. C. Tai
- School of Natural Sciences, Laurentian University, Sudbury, ON P3E 2C6, Canada; (L.M.); (S.T.); (P.M.); (A.K.)
- Medical Science Division, NOSM University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
4
|
Chen PR, Li CY, Yazal T, Chen IC, Liu PL, Chen YT, Liu CC, Lo J, Lin TC, Chang CT, Wu HE, Chen YR, Cheng WC, Chiu CC, Chen CS, Wang SC. Protective effects of nordalbergin against LPS-induced endotoxemia through inhibiting MAPK/NF-κB signaling pathway, NLRP3 inflammasome activation, and ROS production. Inflamm Res 2024; 73:1657-1670. [PMID: 39052062 DOI: 10.1007/s00011-024-01922-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE Nordalbergin is a coumarin extracted from Dalbergia sissoo DC. To date, the biological effects of nordalbergin have not been well investigated. To investigate the anti-inflammatory responses and the anti-oxidant abilities of nordalbergin using lipopolysaccharide (LPS)-activated macrophages and LPS-induced sepsis mouse model. MATERIALS AND METHODS Production of nitrite oxide (NO), prostaglandin E2 (PGE2), pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β), reactive oxygen species (ROS), tissue damage and serum inflammatory markers, and the activation of the NLRP3 inflammasome were examined. RESULTS Our results indicated that nordalbergin reduced the production of NO and pro-inflammatory cytokines in vitro and ex vivo. Nordalbergin also suppressed iNOS and cyclooxygenase-2 expressions, decreased NF-κB activity, and attenuated MAPKs signaling pathway activation by decreasing JNK and p38 phosphorylation by LPS-activated J774A.1 macrophages. Notably, nordalbergin diminished NLRP3 inflammasome activation via repressing the maturation of IL-1β and caspase-1 and suppressing ROS production by LPS/ATP- and LPS/nigericin-activated J774A.1 macrophages. Furthermore, nordalbergin exhibited protective effects against the infiltration of inflammatory cells and also inhibited the levels of organ damage markers (AST, ALT, BUN) by LPS-challenged mice. CONCLUSION Nordalbergin possesses anti-inflammatory effects in macrophage-mediated innate immune responses, alleviates ROS production, decreases NLRP3 activation, and exhibits protective effects against LPS-induced tissue damage in mice.
Collapse
Grants
- NSTC 111-2218-E-037-001, NSTC 111-2314-B-037-071-MY3, NSTC 112-2314-B-037-127, NSTC 112-2314-B-037-128, NSTC 112-2926-I-037-501-G, NSTC 112-2314-B-037-089, NSTC 112-2311-B-039 -001, NSTC 112-2622-E-039-001, NSTC 111-2622-E-039-004 and NSTC 112-2218-E-037-001 National Science and Technology Council, Taiwan, R.O.C.
- KT113P010 NTHU-KMU Joint Research Project
- CMU111-IP-04 and CMU112-MF-25 China Medical University, Taiwan
- DMR-112-056, DMR-113-190, and DMR-113-191 China Medical University Hospital
Collapse
Affiliation(s)
- Pin-Rong Chen
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, 91201, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
| | - Taha Yazal
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - I-Chen Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yi-Ting Chen
- Department of Pathology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 427213, Taiwan
| | - Ching-Chih Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Ophthalmology, Chi Mei Medical Center, Tainan, 71004, Taiwan
| | - Jung Lo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Tzu-Chieh Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ching-Tang Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Hsin-En Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yuan-Ru Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40403, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University, Academia Sinica, Taipei, 40403, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shu-Chi Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
5
|
Yang T, Li J, Cheng X, Lu Q, Farooq Z, Fu Y, Lv S, Nan W, Yu B, Duan J, Zhang Y, Fu Y, Jiang H, McCormick PJ, Li Y, Zhang J. Structural analysis of the human C5a-C5aR1 complex using cryo-electron microscopy. J Struct Biol 2024; 216:108117. [PMID: 39153560 DOI: 10.1016/j.jsb.2024.108117] [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: 05/06/2024] [Revised: 08/07/2024] [Accepted: 08/14/2024] [Indexed: 08/19/2024]
Abstract
The complement system is a complex network of proteins that plays a crucial role in the innate immune response. One important component of this system is the C5a-C5aR1 complex, which is critical in the recruitment and activation of immune cells. In-depth investigation of the activation mechanism as well as biased signaling of the C5a-C5aR1 system will facilitate the elucidation of C5a-mediated pathophysiology. In this study, we determined the structure of C5a-C5aR1-Gi complex at a high resolution of 3 Å using cryo-electron microscopy (Cryo-EM). Our results revealed the binding site of C5a, which consists of a polar recognition region on the extracellular side and an amphipathic pocket within the transmembrane domain. Furthermore, we found that C5a binding induces conformational changes of C5aR1, which subsequently leads to the activation of G protein signaling pathways. Notably, a key residue (M265) located on transmembrane helix 6 (TM6) was identified to play a crucial role in regulating the recruitment of β-arrestin driven by C5a. This study provides more information about the structure and function of the human C5a-C5aR1 complex, which is essential for the proper functioning of the complement system. The findings of this study can also provide a foundation for the design of new pharmaceuticals targeting this receptor with bias or specificity.
Collapse
Affiliation(s)
- Tingting Yang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Jian Li
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, School of Pharmacy, Gannan Medical University, Ganzhou 341000, China; Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Xinyu Cheng
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Qiuyuan Lu
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Department of Chemical Biology, School of Life Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Zara Farooq
- William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ying Fu
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Sijia Lv
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Weiwei Nan
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Boming Yu
- Human Aging Research Institute (HARI), School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Jingjing Duan
- Human Aging Research Institute (HARI), School of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yuting Zhang
- Shenzhen Crystalo Biopharmaceutical Co., Ltd, Shenzhen, Guangdong 518118, China
| | - Yang Fu
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Haihai Jiang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| | - Peter J McCormick
- William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GE, UK.
| | - Yanyan Li
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China; Department of Chemical Biology, School of Life Southern University of Science and Technology, Shenzhen, Guangdong 518055, China.
| | - Jin Zhang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
6
|
Krauklis SA, Hussain J, Murphy KM, Dray EL, Ousley CG, Justyna K, Distefano MD, Steelman AJ, McKim DB. Mononuclear phagocyte morphological response to chemoattractants is dependent on geranylgeranyl pyrophosphate. Am J Physiol Endocrinol Metab 2024; 327:E55-E68. [PMID: 38717364 PMCID: PMC11390116 DOI: 10.1152/ajpendo.00359.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/25/2024] [Accepted: 04/24/2024] [Indexed: 06/22/2024]
Abstract
Statins are used to treat hypercholesterolemia and function by inhibiting the production of the rate-limiting metabolite mevalonate. As such, statin treatment not only inhibits de novo synthesis of cholesterol but also isoprenoids that are involved in prenylation, the posttranslational lipid modification of proteins. The immunomodulatory effects of statins are broad and often conflicting. Previous work demonstrated that statins increased survival and inhibited myeloid cell trafficking in a murine model of sepsis, but the exact mechanisms underlying this phenomenon were unclear. Herein, we investigated the role of prenylation in chemoattractant responses. We found that simvastatin treatment abolished chemoattractant responses induced by stimulation by C5a and FMLP. The inhibitory effect of simvastatin treatment was unaffected by the addition of either farnesyl pyrophosphate (FPP) or squalene but was reversed by restoring geranylgeranyl pyrophosphate (GGPP). Treatment with prenyltransferase inhibitors showed that the chemoattractant response to both chemoattractants was dependent on geranylgeranylation. Proteomic analysis of C15AlkOPP-prenylated proteins identified several geranylgeranylated proteins involved in chemoattractant responses, including RHOA, RAC1, CDC42, and GNG2. Chemoattractant responses in THP-1 human macrophages were also geranylgeranylation dependent. These studies provide data that help clarify paradoxical findings on the immunomodulatory effects of statins. Furthermore, they establish the role of geranylgeranylation in mediating the morphological response to chemoattractant C5a.NEW & NOTEWORTHY The immunomodulatory effect of prenylation is ill-defined. We investigated the role of prenylation on the chemoattractant response to C5a. Simvastatin treatment inhibits the cytoskeletal remodeling associated with a chemotactic response. We showed that the chemoattractant response to C5a was dependent on geranylgeranylation, and proteomic analysis identified several geranylgeranylated proteins that are involved in C5a receptor signaling and cytoskeletal remodeling. Furthermore, they establish the role of geranylgeranylation in mediating the response to chemoattractant C5a.
Collapse
Affiliation(s)
- Steven A Krauklis
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Jamal Hussain
- Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Katherine M Murphy
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Evan L Dray
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Carey G Ousley
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Katarzyna Justyna
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, United States
| | - Mark D Distefano
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota, United States
| | - Andrew J Steelman
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| | - Daniel B McKim
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Department of Animal Sciences,University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States
| |
Collapse
|
7
|
Zemtsovski JD, Tumpara S, Schmidt S, Vijayan V, Klos A, Laudeley R, Held J, Immenschuh S, Wurm FM, Welte T, Haller H, Janciauskiene S, Shushakova N. Alpha1-antitrypsin improves survival in murine abdominal sepsis model by decreasing inflammation and sequestration of free heme. Front Immunol 2024; 15:1368040. [PMID: 38562925 PMCID: PMC10982482 DOI: 10.3389/fimmu.2024.1368040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Background Excessive inflammation, hemolysis, and accumulation of labile heme play an essential role in the pathophysiology of multi-organ dysfunction syndrome (MODS) in sepsis. Alpha1-antitrypsin (AAT), an acute phase protein with heme binding capacity, is one of the essential modulators of host responses to inflammation. In this study, we evaluate the putative protective effect of AAT against MODS and mortality in a mouse model of polymicrobial abdominal sepsis. Methods Polymicrobial abdominal sepsis was induced in C57BL/6N mice by cecal ligation and puncture (CLP). Immediately after CLP surgery, mice were treated intraperitoneally with three different forms of human AAT-plasma-derived native (nAAT), oxidized nAAT (oxAAT), or recombinant AAT (recAAT)-or were injected with vehicle. Sham-operated mice served as controls. Mouse survival, bacterial load, kidney and liver function, immune cell profiles, cytokines/chemokines, and free (labile) heme levels were assessed. In parallel, in vitro experiments were carried out with resident peritoneal macrophages (MPMΦ) and mouse peritoneal mesothelial cells (MPMC). Results All AAT preparations used reduced mortality in septic mice. Treatment with AAT significantly reduced plasma lactate dehydrogenase and s-creatinine levels, vascular leakage, and systemic inflammation. Specifically, AAT reduced intraperitoneal accumulation of free heme, production of cytokines/chemokines, and neutrophil infiltration into the peritoneal cavity compared to septic mice not treated with AAT. In vitro experiments performed using MPMC and primary MPMΦ confirmed that AAT not only significantly decreases lipopolysaccharide (LPS)-induced pro-inflammatory cell activation but also prevents the enhancement of cellular responses to LPS by free heme. In addition, AAT inhibits cell death caused by free heme in vitro. Conclusion Data from the septic CLP mouse model suggest that intraperitoneal AAT treatment alone is sufficient to improve sepsis-associated organ dysfunctions, preserve endothelial barrier function, and reduce mortality, likely by preventing hyper-inflammatory responses and by neutralizing free heme.
Collapse
Affiliation(s)
- Jan D. Zemtsovski
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Srinu Tumpara
- Department of Respiratory Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany
| | | | - Vijith Vijayan
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Andreas Klos
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Robert Laudeley
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Julia Held
- Department of Respiratory Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Florian M. Wurm
- Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Tobias Welte
- Department of Respiratory Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany
| | - Hermann Haller
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, Hannover, Germany
| | - Nelli Shushakova
- Department of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| |
Collapse
|
8
|
Wang T, Han JG, Dong W, Yu YH. LCN2 and ELANE overexpression induces sepsis. Medicine (Baltimore) 2024; 103:e37255. [PMID: 38363924 PMCID: PMC10869048 DOI: 10.1097/md.0000000000037255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/23/2024] [Indexed: 02/18/2024] Open
Abstract
Sepsis is a syndrome characterized by a systemic inflammatory response due to the invasion of pathogenic microorganisms. The relationship between Lipocalin-2 (LCN2), elastase, neutrophil expressed (ELANE) and sepsis remains unclear. The sepsis datasets GSE137340 and GSE154918 profiles were downloaded from gene expression omnibus generated from GPL10558. Batch normalization, differentially expressed Genes (DEGs) screening, weighted gene co-expression network analysis (WGCNA), functional enrichment analysis, Gene Set Enrichment Analysis (GSEA), immune infiltration analysis, construction and analysis of protein-protein interaction (PPI) networks, Comparative Toxicogenomics Database (CTD) analysis were performed. Gene expression heatmaps were generated. TargetScan was used to screen miRNAs of DEGs. 328 DEGs were identified. According to Gene Ontology (GO), in the Biological Process analysis, they were mainly enriched in immune response, apoptosis, inflammatory response, and immune response regulation signaling pathways. In cellular component analysis, they were mainly enriched in vesicles, cytoplasmic vesicles, and secretory granules. In Molecular Function analysis, they were mainly concentrated in hemoglobin binding, Toll-like receptor binding, immunoglobulin binding, and RAGE receptor binding. In Kyoto Encyclopedia of Genes and Genomes (KEGG), they were mainly enriched in NOD-like receptor signaling pathway, Toll-like receptor signaling pathway, TNF signaling pathway, P53 signaling pathway, and legionellosis. Seventeen modules were generated. The PPI network identified 4 core genes (MPO, ELANE, CTSG, LCN2). Gene expression heatmaps revealed that core genes (MPO, ELANE, CTSG, LCN2) were highly expressed in sepsis samples. CTD analysis found that MPO, ELANE, CTSG and LCN2 were associated with sepsis, peritonitis, meningitis, pneumonia, infection, and inflammation. LCN2 and ELANE are highly expressed in sepsis and may serve as molecular targets.
Collapse
Affiliation(s)
- Tao Wang
- Department of Anesthesiology, Tianjin University Chest Hospital, Jinnan District, Tianjin, China
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Jinnan District, Tianjin, China
| | - Jian-Ge Han
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Jinnan District, Tianjin, China
| | - Wei Dong
- Tianjin Key Laboratory of Cardiovascular Emergency and Critical Care, Jinnan District, Tianjin, China
| | - Yong-Hao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Heping District, Tianjin, China
| |
Collapse
|
9
|
Fan Y, Guan B, Xu J, Zhang H, Yi L, Yang Z. Role of toll-like receptor-mediated pyroptosis in sepsis-induced cardiomyopathy. Biomed Pharmacother 2023; 167:115493. [PMID: 37734261 DOI: 10.1016/j.biopha.2023.115493] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/23/2023] Open
Abstract
Sepsis, a life-threatening dysregulated status of the host response to infection, can cause multiorgan dysfunction and mortality. Sepsis places a heavy burden on the cardiovascular system due to the pathological imbalance of hyperinflammation and immune suppression. Myocardial injury and cardiac dysfunction caused by the aberrant host responses to pathogens can lead to cardiomyopathy, one of the most critical complications of sepsis. However, many questions about the specific mechanisms and characteristics of this complication remain to be answered. The causes of sepsis-induced cardiac dysfunction include abnormal cardiac perfusion, myocardial inhibitory substances, autonomic dysfunction, mitochondrial dysfunction, and calcium homeostasis dysregulation. The fight between the host and pathogens acts as the trigger for sepsis-induced cardiomyopathy. Pyroptosis, a form of programmed cell death, plays a critical role in the progress of sepsis. Toll-like receptors (TLRs) act as pattern recognition receptors and participate in innate immune pathways that recognize damage-associated molecular patterns as well as pathogen-associated molecular patterns to mediate pyroptosis. Notably, pyroptosis is tightly associated with cardiac dysfunction in sepsis and septic shock. In line with these observations, induction of TLR-mediated pyroptosis may be a promising therapeutic approach to treat sepsis-induced cardiomyopathy. This review focuses on the potential roles of TLR-mediated pyroptosis in sepsis-induced cardiomyopathy, to shed light on this promising therapeutic approach, thus helping to prevent and control septic shock caused by cardiovascular disorders and improve the prognosis of sepsis patients.
Collapse
Affiliation(s)
- Yixuan Fan
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baoyi Guan
- Department of Internal Medicine-Cardiovascular, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Jianxing Xu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - He Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Liang Yi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhixu Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Intensive Care Unit, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
10
|
彭 智, 颜 海, 卢 秀, 张 新, 黄 娇, 肖 政. [Value of complement component 3 in predicting the prognosis of children with sepsis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:941-946. [PMID: 37718400 PMCID: PMC10511224 DOI: 10.7499/j.issn.1008-8830.2304041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/03/2023] [Indexed: 09/19/2023]
Abstract
OBJECTIVES To investigate changes in complement component 3 (C3) levels in children with sepsis and its correlation with the severity of sepsis and to explore the significance of C3 in predicting mortality in children with sepsis. METHODS A retrospective analysis was conducted on 529 children with sepsis who were admitted to the Pediatric Intensive Care Unit in Hunan Children's Hospital between November 2019 and September 2021. The children were categorized into two groups based on their prognosis at day 28 after sepsis diagnosis: the survival group (n=471) and the death group (n=58). Additionally, the children were divided into normal C3 group (n=273) and reduced C3 group (n=256) based on the median C3 level (0.77 g/L) within 24 hours of admission. Clinical data and laboratory markers were compared between the groups, and assess the predictive value of C3 levels in relation to sepsis-related mortality. RESULTS The death group exhibited significantly lower C3 levels compared to the survival group (P<0.05). Multivariate logistic regression analysis revealed that higher pediatric Sequential Organ Failure Assessment (p-SOFA) scores and lower C3 levels were closely associated with sepsis-related mortality (P<0.05). The receiver operating characteristic curve (ROC) analysis demonstrated that combination of p-SOFA scores and C3 levels yielded an area under the ROC curve of 0.852, which was higher than that of each indicator alone (P<0.05). CONCLUSIONS C3 can serve as an indicator to assess the severity and prognosis of sepsis in children. The combination of p-SOFA scores and C3 levels holds good predictive value for mortality in children with sepsis.
Collapse
|
11
|
Marques A, Torre C, Pinto R, Sepodes B, Rocha J. Treatment Advances in Sepsis and Septic Shock: Modulating Pro- and Anti-Inflammatory Mechanisms. J Clin Med 2023; 12:2892. [PMID: 37109229 PMCID: PMC10142733 DOI: 10.3390/jcm12082892] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Sepsis is currently defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection, and it affects over 25 million people every year. Even more severe, septic shock is a subset of sepsis defined by persistent hypotension, and hospital mortality rates are higher than 40%. Although early sepsis mortality has greatly improved in the past few years, sepsis patients who survive the hyperinflammation and subsequent organ damage often die from long-term complications, such as secondary infection, and despite decades of clinical trials targeting this stage of the disease, currently, no sepsis-specific therapies exist. As new pathophysiological mechanisms have been uncovered, immunostimulatory therapy has emerged as a promising path forward. Highly investigated treatment strategies include cytokines and growth factors, immune checkpoint inhibitors, and even cellular therapies. There is much to be learned from related illnesses, and immunotherapy trials in oncology, as well as the recent COVID-19 pandemic, have greatly informed sepsis research. Although the journey ahead is a long one, the stratification of patients according to their immune status and the employment of combination therapies represent a hopeful way forward.
Collapse
Affiliation(s)
- Adriana Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - Carla Torre
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - Rui Pinto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
- Joaquim Chaves Saúde, Joaquim Chaves Laboratório de Análises Clínicas, Miraflores, 1495-069 Algés, Portugal
| | - Bruno Sepodes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| |
Collapse
|
12
|
Huang FM, Chang YC, Lee MW, Su NY, Yang LC, Kuan YH. Rutin alleviates bisphenol A-glycidyl methacrylate-induced generation of proinflammatory mediators through the MAPK and NF-κB pathways in macrophages. ENVIRONMENTAL TOXICOLOGY 2023; 38:628-634. [PMID: 36413001 DOI: 10.1002/tox.23711] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/31/2022] [Accepted: 11/05/2022] [Indexed: 06/16/2023]
Abstract
Bisphenol A-glycidyl methacrylate (BisGMA) is a methacrylate monomer that is mainly used in three-dimensional structures to reconstruct dental and bony defects. BisGMA has toxic and proinflammatory effects on macrophages. Rutin is a natural flavonol glycoside that is present in various plants and has useful biological effects, such as anti-inflammatory, anticancer, and antioxidative effects. The aim of this study was to investigate the anti-inflammation of rutin in macrophages after exposure to BisGMA. Pretreatment of the RAW264.7 macrophage with rutin at 0, 10, 30, and 100 μM for 30 min before being incubated with BisGMA at 0 or 3 μM. Proinflammatory cytokines and prostaglandin (PG) E2 were detected by enzyme-linked immunosorbent assay (ELISA). Nitric oxide (NO) was detected by the Griess assay. Expression and phosphorylation of proteins were measured by Western blot assay. Pretreatment with rutin inhibited the BisGMA-induced generation of proinflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and PGE2, in macrophages. Rutin also suppressed the BisGMA-induced secretion of NO and expression of inducible nitric oxide synthase (iNOS) in a concentration-dependent manner. Furthermore, rutin suppressed the mitogen-activated protein kinase (MAPK) phosphorylation in a concentration-dependent manner. Finally, rutin suppressed the BisGMA-induced phosphorylation of nuclear factor (NF)-κB p65 and degradation of inhibitor of κB (IκB). These results indicate that the concentration of rutin has an inhibitory effect on proinflammatory mediator generation, MAPK phosphorylation, NF-κB p65 phosphorylation, and IκB degradation. In conclusion, rutin is a potential anti-inflammatory agent for BisGMA-stimulated macrophages through NF-κB p65 phosphorylation and IκB degradation resulting from MAPK phosphorylation.
Collapse
Affiliation(s)
- Fu-Mei Huang
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chao Chang
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Min-Wei Lee
- A Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung, Taiwan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ni-Yu Su
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Li-Chiu Yang
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Hsiang Kuan
- Department of Pharmacology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pharmacy, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
13
|
Sen'kova AV, Savin IA, Odarenko KV, Salomatina OV, Salakhutdinov NF, Zenkova MA, Markov AV. Protective effect of soloxolone derivatives in carrageenan- and LPS-driven acute inflammation: Pharmacological profiling and their effects on key inflammation-related processes. Biomed Pharmacother 2023; 159:114231. [PMID: 36640672 DOI: 10.1016/j.biopha.2023.114231] [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: 10/17/2022] [Revised: 12/21/2022] [Accepted: 01/08/2023] [Indexed: 01/13/2023] Open
Abstract
The anti-inflammatory potential of three cyanoenone-containing triterpenoids, including soloxolone methyl (SM), soloxolone (S) and its novel derivative bearing at the C-30 amidoxime moiety (SAO), was studied in murine models of acute inflammation. It was found that the compounds effectively suppressed the development of carrageenan-induced paw edema and peritonitis as well as lipopolysaccharide (LPS)-driven acute lung injury (ALI) with therapeutic outcomes comparable with that of the reference drugs indomethacin and dexamethasone. Non-immunogenic carrageenan-stimulated inflammation was more sensitive to the transformation of C-30 of SM compared with immunogenic LPS-induced inflammation: the anti-inflammatory properties of the studied compounds against carrageenan-induced paw edema and peritonitis decreased in the order of SAO > S > > SM, whereas the efficiency of these triterpenoids against LPS-driven ALI was similar (SAO ≈ S ≈ SM). Further studies demonstrated that soloxolone derivatives significantly inhibited a range of immune-related processes, including granulocyte influx and the expression of key pro-inflammatory cytokines and chemokines in the inflamed sites as well as the functional activity of macrophages. Moreover, SM was found to prevent inflammation-associated apoptosis of A549 pneumocytes and effectively inhibited the protease activity of thrombin (IC50 = 10.3 µM) tightly associated with rodent inflammatome. Taken together, our findings demonstrate that soloxolone derivatives can be considered as novel promising anti-inflammatory drug candidates with multi-targeted mechanism of action.
Collapse
Affiliation(s)
- Aleksandra V Sen'kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 8, 630090 Novosibirsk, Russia.
| | - Innokenty A Savin
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 9, 630090 Novosibirsk, Russia.
| | - Kirill V Odarenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 8, 630090 Novosibirsk, Russia.
| | - Oksana V Salomatina
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 9, 630090 Novosibirsk, Russia.
| | - Nariman F Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 9, 630090 Novosibirsk, Russia.
| | - Marina A Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 8, 630090 Novosibirsk, Russia.
| | - Andrey V Markov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Lavrent'ev avenue, 8, 630090 Novosibirsk, Russia.
| |
Collapse
|
14
|
Zetoune FS, Ward PA. THE IMMUNOPATHOGENESIS OF POLYMICROBIAL SEPSIS. Shock 2023; 59:311-317. [PMID: 36377404 PMCID: PMC9957923 DOI: 10.1097/shk.0000000000002049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
ABSTRACT This report deals with the advances made in the areas of complement and its role in sepsis, both in mice and in humans. The study relates to work over the past 25 years (late 1990s to October 2022). During this period, there has been very rapid progress in understanding the activation pathways of complement and the activation products of complement, especially the anaphylatoxin C5a and its receptors, C5aR1 and C5aR2. Much has also been learned about these pathways of activation that trigger activation of the innate immune system and the array of strong proinflammatory cytokines that can cause cell and organ dysfunction, as well as complement products that cause immunosuppression. The work in septic humans and mice, along with patients who develop lung dysfunction caused by COVID-19, has taught us that there are many strategies for treatment of humans who are septic or develop COVID-19-related lung dysfunction. To date, treatments in humans with these disorders suggest that we are in the midst of a new and exciting area related to the complement system.
Collapse
Affiliation(s)
- Firas S. Zetoune
- University of Michigan Medical School, Department of Pathology, Ann Arbor, MI 48109
| | - Peter A. Ward
- University of Michigan Medical School, Department of Pathology, Ann Arbor, MI 48109
| |
Collapse
|
15
|
Han Z, Yuan Z, Shu L, Li T, Yang F, Chen L. Extracellular histone H3 facilitates ferroptosis in sepsis through ROS/JNK pathway. Immun Inflamm Dis 2023; 11:e754. [PMID: 36705411 PMCID: PMC9795329 DOI: 10.1002/iid3.754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/27/2022] [Accepted: 12/02/2022] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Previous evidence realized the critical role of histone in disease control. The anti-inflammatory function of estradiol (E2) in sepsis has been documented. We here intended to unveil the role of extracellular histone H3 in sepsis regarding cell ferroptosis and the role of E2 in a such mechanism. METHODS Clinical sample, cecal ligation and puncture (CLP)-induced animal models and lipopolysaccharides (LPS)-induced cell models were prepared for testing relative expression of extracellular histone H3 and E2 as well as analyzing the role of extracellular histone H3 and E2 in sepsis concerning cell viability, reactive oxygen species (ROS), and ferroptosis. RESULTS Under sepsis, we found increased ferroptosis and extracellular histone H3 content, but reduced E2 concentration. Extracellular histone H3 facilitated ferroptosis of human umbilical vein endothelial cells (HUVECs) induced by LPS through activating the ROS/c-Jun N-terminal kinase (JNK) pathway. Moreover, E2 antagonized the effect of extracellular histone H3 on LPS-induced HUVEC ferroptosis and sepsis injury in CLP-induced animal models. CONCLUSION We highlighted that extracellular histone H3 facilitated lipopolysaccharides-induced HUVEC ferroptosis via activating ROS/JNK pathway, and such an effect could be antagonized by E2.
Collapse
Affiliation(s)
- Zhijun Han
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Zhizhou Yuan
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Linfei Shu
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Tao Li
- The Second Affiliated Hospital of Hainan Medical UniversityHaikouHainan ProvinceChina
| | - Fan Yang
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| | - Lei Chen
- Department of Urology SurgeryZhuzhou Central HospitalZhuzhouHunan ProvinceChina
| |
Collapse
|
16
|
Complement Activation in Patients With Heat-Related Illnesses: Soluble CD59 Is a Novel Biomarker Indicating Severity of Heat-Related Illnesses. Crit Care Explor 2022; 4:e0678. [PMID: 35474654 PMCID: PMC9029987 DOI: 10.1097/cce.0000000000000678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Although multiple organ dysfunction syndrome (MODS) is the main cause of death in patients with heat-related illnesses, its underlying pathophysiological mechanism remains elusive. Complement activation is considered one of the main causes of MODS in patients with sepsis and trauma. Considering the pathophysiological similarity of heat related-illnesses with sepsis and trauma, the complement system might be activated in patients with heat-related illnesses as well. Our aim was to investigate whether excessive complement activation occurs in patients with heat-related illnesses.
Collapse
|
17
|
van Etten ES, Kaushik K, Jolink WMT, Koemans EA, Ekker MS, Rasing I, Voigt S, Schreuder FHBM, Cannegieter SC, Rinkel GJE, Lijfering WM, Klijn CJM, Wermer MJH. Trigger Factors for Spontaneous Intracerebral Hemorrhage: A Case-Crossover Study. Stroke 2021; 53:1692-1699. [PMID: 34911344 DOI: 10.1161/strokeaha.121.036233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Whether certain activities can trigger spontaneous intracerebral hemorrhage (ICH) remains unknown. Insights into factors that trigger vessel rupture resulting in ICH improves knowledge on the pathophysiology of ICH. We assessed potential trigger factors and their risk for ICH onset. METHODS We included consecutive patients diagnosed with ICH between July 1, 2013, and December 31, 2019. We interviewed patients on their exposure to 12 potential trigger factors (eg, Valsalva maneuvers) in the (hazard) period soon before onset of ICH and their normal exposure to these trigger factors in the year before the ICH. We used the case-crossover design to calculate relative risks (RR) for potential trigger factors. RESULTS We interviewed 149 patients (mean age 64, 66% male) with ICH. Sixty-seven (45%) had a lobar hemorrhage, 60 (40%) had a deep hemorrhage, 19 (13%) had a cerebellar hemorrhage, and 3 (2%) had an intraventricular hemorrhage. For ICH in general, there was an increased risk within an hour after caffeine consumption (RR=2.5 [95% CI=1.8-3.6]), within an hour after coffee consumption alone (RR=4.8 [95% CI=3.3-6.9]), within an hour after lifting >25 kg (RR=6.6 [95% CI=2.2-19.9]), within an hour after minor head trauma (RR=10.1 [95% CI=1.7-60.2]), within an hour after sexual activity (RR=30.4 [95% CI=16.8-55.0]), within an hour after straining for defecation (RR=37.6 [95% CI=22.4-63.4]), and within an hour after vigorous exercise (RR=21.8 [95% CI=12.6-37.8]). Within 24 hours after flu-like disease or fever, the risk for ICH was also increased (RR=50.7 [95% CI=27.1-95.1]). Within an hour after Valsalva maneuvers, the RR for deep ICH was 3.5 (95% CI=1.7-6.9) and for lobar ICH the RR was 2.0 (95% CI=0.9-4.2). CONCLUSIONS We identified one infection and several blood pressure related trigger factors for ICH onset, providing new insights into the pathophysiology of vessel rupture resulting in ICH.
Collapse
Affiliation(s)
- Ellis S van Etten
- Department of Neurology, Leiden University Medical Center, the Netherlands. (E.S.v.E., K.K., E.A.K., I.R., S.V., M.J.H.W.)
| | - Kanishk Kaushik
- Department of Neurology, Leiden University Medical Center, the Netherlands. (E.S.v.E., K.K., E.A.K., I.R., S.V., M.J.H.W.)
| | - Wilmar M T Jolink
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, Utrecht University, the Netherlands (W.M.T.J., G.J.E.R., C.J.M.K.)
| | - Emma A Koemans
- Department of Neurology, Leiden University Medical Center, the Netherlands. (E.S.v.E., K.K., E.A.K., I.R., S.V., M.J.H.W.)
| | - Merel S Ekker
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Centre for Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands (M.S.E., F.H.B.M.S., C.J.M.K.)
| | - Ingeborg Rasing
- Department of Neurology, Leiden University Medical Center, the Netherlands. (E.S.v.E., K.K., E.A.K., I.R., S.V., M.J.H.W.)
| | - Sabine Voigt
- Department of Neurology, Leiden University Medical Center, the Netherlands. (E.S.v.E., K.K., E.A.K., I.R., S.V., M.J.H.W.)
| | - Floris H B M Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Centre for Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands (M.S.E., F.H.B.M.S., C.J.M.K.)
| | - Suzanne C Cannegieter
- Department of Epidemiology, Leiden University Medical Center, the Netherlands. (S.C.C., W.M.L.).,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands. (S.C.C., W.M.L.).,Department of Internal Medicine, Section Thrombosis and Hemostasis, Leiden University Medical Center, the Netherlands. (S.C.C.)
| | - Gabriël J E Rinkel
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, Utrecht University, the Netherlands (W.M.T.J., G.J.E.R., C.J.M.K.)
| | - Willem M Lijfering
- Department of Epidemiology, Leiden University Medical Center, the Netherlands. (S.C.C., W.M.L.).,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands. (S.C.C., W.M.L.)
| | - Catharina J M Klijn
- Department of Neurology and Neurosurgery, University Medical Center Utrecht Brain Center, Utrecht University, the Netherlands (W.M.T.J., G.J.E.R., C.J.M.K.).,Department of Neurology, Donders Institute for Brain, Cognition and Behavior, Centre for Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands (M.S.E., F.H.B.M.S., C.J.M.K.)
| | - Marieke J H Wermer
- Department of Neurology, Leiden University Medical Center, the Netherlands. (E.S.v.E., K.K., E.A.K., I.R., S.V., M.J.H.W.)
| |
Collapse
|
18
|
Xu H, An X, Tian J, Fu M, Wang Q, Li C, He X, Niu L. Angiotensin-(1-7) protects against sepsis-associated left ventricular dysfunction induced by lipopolysaccharide. Peptides 2021; 144:170612. [PMID: 34298021 DOI: 10.1016/j.peptides.2021.170612] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/11/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022]
Abstract
Sepsis-induced myocardial dysfunction is a major cause of death. The present study explored whether angiotensin (Ang)-(1-7), an important biologically active peptide of the renin-angiotensin system, could improve cardiac dysfunction and attenuate inflammation and apoptosis. Experiments were carried out in mice and in neonatal rat cardiomyocytes (NRCMs) treated with lipopolysaccharide (LPS) or Ang-(1-7). Angiotensin converting enzyme 2 (ACE2), Ang-(1-7) and Mas receptor (MasR) expressions were reduced in the mouse left ventricular and NRCM treated with LPS. Ang-(1-7) increased the ejection fraction and fractional shortening of left ventricular, which were reduced upon LPS injection in mice. Ang-(1-7) pre-treatment reversed LPS-induced decreases of α-myosin heavy chain (MHC) and β-MHC, and increases of S100 calcium binding protein A8 (S100A8) and S100A9 in the mouse left ventricular. The LPS-induced increases of tumor necrosis factor (TNF)-α and interleukin (IL)-1β in the mouse left ventricular and NRCMs were inhibited by Ang-(1-7) administration. Ang-(1-7) treatment reversed the increases of cleaved-caspase 3, cleaved-caspase 8 and Bax, and the decrease of Bcl2 induced by LPS in the mouse left ventricular and NRCMs. The increases of MAPKs pathway induced by LPS in NRCMs were inhibited by Ang-(1-7). These results indicate that Ang-(1-7) protects against sepsis-associated left ventricular dysfunction induced by LPS, and increases cardiac contractility via attenuating inflammation and apoptosis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xinjiang An
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jing Tian
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Mingyu Fu
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qingwen Wang
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chunli Li
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiuhua He
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ling Niu
- Department of Cardiology, Xuzhou Children's Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
19
|
More than a Pore: Nonlytic Antimicrobial Functions of Complement and Bacterial Strategies for Evasion. Microbiol Mol Biol Rev 2021; 85:85/1/e00177-20. [PMID: 33504655 DOI: 10.1128/mmbr.00177-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The complement system is an evolutionarily ancient defense mechanism against foreign substances. Consisting of three proteolytic activation pathways, complement converges on a common effector cascade terminating in the formation of a lytic pore on the target surface. The classical and lectin pathways are initiated by pattern recognition molecules binding to specific ligands, while the alternative pathway is constitutively active at low levels in circulation. Complement-mediated killing is essential for defense against many Gram-negative bacterial pathogens, and genetic deficiencies in complement can render individuals highly susceptible to infection, for example, invasive meningococcal disease. In contrast, Gram-positive bacteria are inherently resistant to the direct bactericidal activity of complement due to their thick layer of cell wall peptidoglycan. However, complement also serves diverse roles in immune defense against all bacteria by flagging them for opsonization and killing by professional phagocytes, synergizing with neutrophils, modulating inflammatory responses, regulating T cell development, and cross talk with coagulation cascades. In this review, we discuss newly appreciated roles for complement beyond direct membrane lysis, incorporate nonlytic roles of complement into immunological paradigms of host-pathogen interactions, and identify bacterial strategies for complement evasion.
Collapse
|
20
|
Bothrops lanceolatus snake (Fer-de-lance) venom triggers inflammatory mediators' storm in human blood. Arch Toxicol 2021; 95:1129-1138. [PMID: 33398417 DOI: 10.1007/s00204-020-02959-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/26/2020] [Indexed: 12/20/2022]
Abstract
Systemic increased inflammatory mediators' levels are a hallmark in a plethora of pathological conditions, including thrombotic diseases as the envenomation by Bothrops lanceolatus snake. Multiple organ infarctions, which are not prevented by anticoagulant therapy, are the main cause of death on this envenomation. However, the potential mechanisms involved in these systemic reactions are underexplored. This study aimed to explore the potential systemic events which could contribute to thrombotic reactions on the envenomation by B. lanceolatus in an ex vivo human whole-blood model. B. lanceolatus venom elicited an inflammatory reaction, which was characterized by a strong complement activation, since we detected high C3a, C4a and C5a anaphylatoxins levels. Besides, the venom promoted soluble Terminal Complement Complex (sTCC) assembly. Complement activation was accompanied by intense lipid mediators' release, which included LTB4, PGE2 and TXB2. In addition, in the blood exposed to B. lanceolatus venom, we detected IL-1β, IL-6 and TNF-α interleukins production. Chemokines, including CCL2, CCL5 and CXCL8 were upregulated in the venom presence. These outcomes show that B. lanceolatus venom causes a strong inflammatory reaction in the blood favoring a potential setting to thrombi formation. Thus, inhibiting inflammatory mediators or their receptors may help in the envenomed patients' management.
Collapse
|
21
|
Elyaspour Z, Zibaeenezhad MJ, Razmkhah M, Razeghian-Jahromi I. Is It All About Endothelial Dysfunction and Thrombosis Formation? The Secret of COVID-19. Clin Appl Thromb Hemost 2021; 27:10760296211042940. [PMID: 34693754 PMCID: PMC8543709 DOI: 10.1177/10760296211042940] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/12/2021] [Indexed: 01/08/2023] Open
Abstract
The world is in a hard battle against COVID-19. Endothelial cells are among the most critical targets of SARS-CoV-2. Dysfunction of endothelium leads to vascular injury following by coagulopathies and thrombotic conditions in the vital organs increasing the risk of life-threatening events. Growing evidences revealed that endothelial dysfunction and consequent thrombotic conditions are associated with the severity of outcomes. It is not yet fully clear that these devastating sequels originate directly from the virus or a side effect of virus-induced cytokine storm. Due to endothelial dysfunction, plasma levels of some biomarkers are changed and relevant clinical manifestations appear as well. Stabilization of endothelial integrity and supporting its function are among the promising therapeutic strategies. Other than respiratory, COVID-19 could be called a systemic vascular disease and this aspect should be scrutinized in more detail in order to reduce related mortality. In the present investigation, the effects of COVID-19 on endothelial function and thrombosis formation are discussed. In this regard, critical players, laboratory findings, clinical manifestation, and suggestive therapies are presented.
Collapse
Affiliation(s)
- Zahra Elyaspour
- Cardiovascular Research Center, Shiraz
University of Medical Sciences, Shiraz, Iran
| | | | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research,
Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
22
|
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
|
23
|
What's New in Shock, November 2020? Shock 2020; 54:583-585. [PMID: 33048866 DOI: 10.1097/shk.0000000000001674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
24
|
Marchetti M. COVID-19-driven endothelial damage: complement, HIF-1, and ABL2 are potential pathways of damage and targets for cure. Ann Hematol 2020; 99:1701-1707. [PMID: 32583086 PMCID: PMC7312112 DOI: 10.1007/s00277-020-04138-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023]
Abstract
COVID-19 pandemia is a major health emergency causing hundreds of deaths worldwide. The high reported morbidity has been related to hypoxia and inflammation leading to endothelial dysfunction and aberrant coagulation in small and large vessels. This review addresses some of the pathways leading to endothelial derangement, such as complement, HIF-1α, and ABL tyrosine kinases. This review also highlights potential targets for prevention and therapy of COVID-19-related organ damage and discusses the role of marketed drugs, such as eculizumab and imatinib, as suitable candidates for clinical trials.
Collapse
Affiliation(s)
- Monia Marchetti
- Hematology Department, Az Osp SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy.
| |
Collapse
|