1
|
Hook JS, Matheis AD, Kavanaugh JS, Horswill AR, Moreland JG. Role for IRAK-4 and p38 in Neutrophil Signaling in Response to Bacterial Lipoproteins from Staphylococcus aureus. Inflammation 2024:10.1007/s10753-024-02147-7. [PMID: 39302496 DOI: 10.1007/s10753-024-02147-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Neutrophils, polymorphonuclear leukocytes (PMN), express numerous pattern recognition receptors, including TLRs, capable of recognizing a wide variety of pathogens. Receptor engagement initiates a cascade of PMN responses with some occurring in seconds, and some requiring de novo protein synthesis over the course of many hours. Although numerous species of bacteria and bacterial products have been shown to activate PMN via TLRs, the signaling intermediates required for distinct PMN responses have not been well-defined in human PMN. Given the potential for host tissue damage by overexuberant PMN activity, a better understanding of neutrophil signaling is needed to generate effective therapies. We hypothesized that PMN responses to a lipoprotein-containing cell membrane preparation from methicillin-resistant S. aureus (MRSA-CMP) would activate signaling via IRAK4 and p38, with potentially distinct pathways for early vs. late responses. Using human PMN we investigated MRSA-CMP-elicited reactive oxygen species (ROS) production, elastase activity, NET formation, IL-8 production, and the role of IRAK4 and p38 activation. MRSA-CMP elicited ROS in a concentration and lipoprotein-dependent manner. MRSA-CMP elicited phosphorylation of p38 MAPK, and MRSA-CMP-elicited ROS production was partially dependent on p38 MAPK and IRAK4 activation. Inhibition of IRAK4 resulted in a reduction of p38 phosphorylation. MRSA-CMP-elicited elastase activity and NET formation was partially dependent on p38 MAPK activation, but independent of IRAK4 activation. MRSA-CMP-elicited IL-8 production required both p38 and IRAK4 activation. In conclusion, MRSA-CMP elicits PMN responses via distinct signaling pathways. There is potential to target components of the neutrophil inflammatory response without compromising critical pathogen-specific immune functions.
Collapse
Affiliation(s)
- Jessica S Hook
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Austin D Matheis
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jeffrey S Kavanaugh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, USA
- Department of Veterans Affairs, Eastern Colorado Healthcare System, Aurora, CO, USA
| | - Jessica G Moreland
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Microbiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-8504, USA.
| |
Collapse
|
2
|
Liu X, Li Y, Zhang W, Gao N, Chen J, Xiao C, Zhang G. Inhibition of cIAP1/2 reduces RIPK1 phosphorylation in pulmonary endothelial cells and alleviate sepsis-induced lung injury and inflammatory response. Immunol Res 2024; 72:841-850. [PMID: 38748318 DOI: 10.1007/s12026-024-09491-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/08/2024] [Indexed: 06/01/2024]
Abstract
Acute respiratory distress syndrome (ARDS)/acute lung injury (ALI) is a severe complication of sepsis characterized by acute respiratory distress, hypoxemia, and diffuse bilateral pulmonary infiltrates. The regulation of RIPK1 is an important part of the inflammatory response, and cIAP1/2 serves as the E3 ubiquitin ligase for RIPK1. In this study, we investigated the effect and mechanism of cIAP1/2 inhibition on sepsis-induced lung injury. Our results showed that cIAP1/2 inhibition can alleviate sepsis-induced lung injury and reduce the inflammatory response, which is accompanied by downregulation of RIPK1 phosphorylation and ubiquitination. Additionally, cIAP1/2 inhibition led to the up-regulation of programmed cell death, including apoptosis, necroptosis, and pyroptosis, and inhibiting these three cell death pathways can further reduce the inflammatory response, which is similar to the recently discovered programmed cell death pathway PANoptosis. Our findings suggest that cIAP1/2 and PANoptosis inhibition may be a new strategy for treating sepsis-induced lung injury and provide important references for further exploring the mechanism of sepsis-induced lung injury and identifying new therapeutic targets.
Collapse
Affiliation(s)
- Xiaoyu Liu
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Yan Li
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Weijian Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
- Peking University, China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Nan Gao
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| | - Cheng Xiao
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, China.
| | - Guoqiang Zhang
- Department of Emergency, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
3
|
Ishikawa F, Matsubara T, Koyama T, Iwamoto H, Miyaji K. Whey protein hydrolysate mitigates both inflammation and endotoxin tolerance in THP-1 human monocytic leukemia cells. Immun Inflamm Dis 2022; 10:e737. [PMID: 36444621 PMCID: PMC9639455 DOI: 10.1002/iid3.737] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 10/10/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION It is important to control both inflammation and immunosuppression after severe insults, such as sepsis, trauma, and surgery. Endotoxin tolerance is one of the immunosuppressive conditions and it has been known that endotoxin tolerance relates to poorer clinical outcomes in patients with severe insults. This study investigated whether whey protein hydrolysate (WPH) mitigates inflammation and endotoxin tolerance in THP-1 human monocytic leukemia cells. METHODS Endotoxin tolerance can be experimentally reproduced by two consecutive stimulations with lipopolysaccharide (LPS). THP-1 cells were incubated with LPS and WPH (first stimulation). After collecting the culture supernatant to evaluate the effect on inflammation, the cells were washed and restimulated by 100 ng/ml LPS (second stimulation). The culture supernatant was again collected to evaluate the effect on endotoxin tolerance. Concentrations of LPS and WPH in the first stimulation were adjusted to evaluate their dose dependency. Cytokine levels in the supernatant were determined by enzyme-linked immunosorbent assay. Statistical analysis was performed using the student's t-test or Dunnett's test. RESULTS Five mg/ml WPH significantly decreased interleukin (IL)-6 (p = .006) and IL-10 (p < .001) levels after the first LPS stimulation (1000 ng/ml). WPH significantly increased tumor necrosis factor-alpha (p < .001) and IL-10 (p = .014) levels after the second LPS stimulation. The suppressive effect of WPH on inflammation and endotoxin tolerance was dependent on the concentrations of LPS and WPH. The effective dose of WPH for endotoxin tolerance was lower than its effective dose for inflammation. CONCLUSION WPH mitigated both inflammation and endotoxin tolerance. Therefore, WPH might be a candidate for valuable food ingredients to control both inflammation and immunosuppression after severe insults.
Collapse
Affiliation(s)
- Fuka Ishikawa
- Health Care & Nutrition Science InstituteR&D Division, Morinaga Milk Industry Co. Ltd.KanagawaZamaJapan
| | - Takeshi Matsubara
- Health Care & Nutrition Science InstituteR&D Division, Morinaga Milk Industry Co. Ltd.KanagawaZamaJapan
| | - Takahiro Koyama
- Health Care & Nutrition Science InstituteR&D Division, Morinaga Milk Industry Co. Ltd.KanagawaZamaJapan
| | - Hiroshi Iwamoto
- Health Care & Nutrition Science InstituteR&D Division, Morinaga Milk Industry Co. Ltd.KanagawaZamaJapan
| | - Kazuhiro Miyaji
- Health Care & Nutrition Science InstituteR&D Division, Morinaga Milk Industry Co. Ltd.KanagawaZamaJapan
| |
Collapse
|
4
|
Çakırlar FK. Application of Biomarkers in the Diagnostic Distinction of Bacterial and Viral Infections. Biomark Med 2022. [DOI: 10.2174/9789815040463122010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Infectious diseases, which pose a great threat worldwide, have a significant
impact on public health and the world economy. It contributes to increased healthcare
costs, unnecessary drug-related side effects, and increased antimicrobial resistance. It is
not always easy to distinguish the etiological differentiation of diseases that can
develop with bacteria and viruses. Therefore, one of the biggest challenges in medicine
is how to correctly distinguish between the different causes of these infections and how
to manage the patient. Because bacterial and viral infections often present similar
symptoms. The real decision is whether the infection is caused by bacteria or viruses
and whether to treat the patient with antibiotics. There are many different
methodological approaches to diagnosing infections. Biomarkers have been used in the
diagnosis of diseases and other conditions for many years. Biomarkers are molecules
found in blood and body fluids in measurable amounts, which can evaluate biological
and pathological processes. These key indicators can provide vital information in
determining disease prognosis, predicting response to treatments, adverse events and
drug interactions, and identifying key risks. An effective biomarker is extremely
important for the early diagnosis of various diseases. The explosion of interest in
biomarker research is driving the development of new predictive, diagnostic, and
prognostic products in modern medical practice. The purpose of this review is to
demonstrate the use and diagnostic potential of current and investigational biomarkers
in the distinction between bacterial and viral infections.
Collapse
Affiliation(s)
- Fatma Köksal Çakırlar
- Faculty of Cerrahpaşa Medicine, University of İstanbul- Cerrahpaşa,Department of Medical Microbiology,Department of Medical Microbiology, Faculty of Cerrahpaşa Medicine, University of İstanbul- Cerrahpaşa, Istanbul, Turkey,Istanbul,Turkey
| |
Collapse
|
5
|
Lin Y, Wongkrajang K, Shen X, Wang P, Zhou Z, Chuprajob T, Sornkaew N, Yang N, Yang L, Lu X, Chokchaisiri R, Suksamrarn A, Zhang G, Wang F. Discovery of diarylheptanoids that activate α7 nAchR-JAK2-STAT3 signaling in macrophages with anti-inflammatory activity in vitro and in vivo. Bioorg Med Chem 2022; 66:116811. [PMID: 35576655 DOI: 10.1016/j.bmc.2022.116811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 11/16/2022]
Abstract
Acute inflammatory diseases, such as sepsis, are life-threatening illnesses. Regulating the α7 nicotinic acetylcholine receptor (α7 nAchR)-mediated signaling may be a promising strategy to treat sepsis. Diarylheptanoids have long been found to exhibit anti-inflammatory properties. However, the possible mechanism of diarylheptanoids has rarely been investigated. In this study, we isolated and synthesized 49 diarylheptanoids and analogues and evaluated their anti-inflammatory activities. Among them, compounds 28 and 40 markedly blocked lipopolysaccharide (LPS)-induced production of nitric oxide (NO), interleukin-1β (IL-1β) and interleukin-6 in murine RAW264.7 cells. Furthermore, compounds 28 and 40 also effectively attenuated LPS-induced sepsis, acute lung injury, and cytokines release in vivo. Mechanistically, compounds 28 and 40 significantly induced phosphorylation of janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling and suppression of nuclear factor-κB (NF-κB) pathway. Furthermore, blocking α7 nAchR could effectively abolish compounds 28 and 40-mediated activation of JAK2-STAT3 signaling as well as inhibition of NF-κB activation and NO production in LPS-exposed RAW264.7 cells. Collectively, our findings have identified a new diarylheptanoid, compound 28, as an agonist of α7 nAchR-JAK2-STAT3 signaling, which can be potentially developed as a valuable candidate for the treatment of sepsis, and provide a new lead structure for the development of anti-inflammatory agents targeting α7 nAchR-JAK2-STAT3 signaling.
Collapse
Affiliation(s)
- Yuan Lin
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China; Sichuan Xincheng Biological Co., LTD, Chengdu, China
| | - Kanjana Wongkrajang
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand; Department of Chemistry, Faculty of Science and Technology, Pibulsongkram Rajabhat University, Phitsanulok, Thailand
| | - Xiaofei Shen
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China; Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zongyuan Zhou
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Thipphawan Chuprajob
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand; Department of Chemistry, Faculty of Science, Siam University, Bangkok, Thailand
| | - Nilubon Sornkaew
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Na Yang
- West China-Frontier PharmaTech Co., Ltd, Chengdu, China
| | - Lijuan Yang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Xiaoxia Lu
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | | | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand.
| | - Guolin Zhang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.
| | - Fei Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.
| |
Collapse
|
6
|
Kim HG, Lee C, Yoon JH, Kim JH, Cho JY. BN82002 alleviated tissue damage of septic mice by reducing inflammatory response through inhibiting AKT2/NF-κB signaling pathway. Pharmacotherapy 2022; 148:112740. [DOI: 10.1016/j.biopha.2022.112740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 01/20/2023]
|
7
|
Madonna G, Sale S, Capone M, De Falco C, Santocchio V, Di Matola T, Fiorentino G, Pirozzi C, D’Antonio A, Sabatino R, Atripaldi L, Atripaldi U, Raffone M, Curvietto M, Grimaldi AM, Vanella V, Festino L, Scarpato L, Palla M, Spatarella M, Perna F, Cerino P, Botti G, Parrella R, Montesarchio V, Ascierto PA, Atripaldi L. Clinical Outcome Prediction in COVID-19 Patients by Lymphocyte Subsets Analysis and Monocytes' iTNF-α Expression. BIOLOGY 2021; 10:735. [PMID: 34439967 PMCID: PMC8389652 DOI: 10.3390/biology10080735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022]
Abstract
In December 2019, a novel coronavirus, "SARS-CoV-2", was recognized as the cause of coronavirus disease 2019 (COVID-19). Several studies have explored the changes and the role of inflammatory cells and cytokines in the immunopathogenesis of the disease, but until today, the results have been controversial. Based on these premises, we conducted a retrospective assessment of monocyte intracellular TNF-α expression (iTNF-α) and on the frequencies of lymphocyte sub-populations in twenty-five patients with moderate/severe COVID-19. We found lymphopenia in all COVID-19 infected subjects compared to healthy subjects. On initial observation, in patients with favorable outcomes, we detected a high absolute eosinophil count and a high CD4+/CD8+ T lymphocytes ratio, while in the Exitus Group, we observed high neutrophil and CD8+ T lymphocyte counts. During infection, in patients with favorable outcomes, we observed a rise in the lymphocyte count, in the monocyte and in Treg lymphocyte counts, and in the CD4+ and in CD8+ T lymphocytes count but a reduction in the CD4+/CD8+ T lymphocyte ratio. Instead, in the Exitus Group, we observed a reduction in the Treg lymphocyte counts and a decrease in iTNF-α expression. Our preliminary findings point to a modulation of the different cellular mediators of the immune system, which probably play a key role in the outcomes of COVID-19.
Collapse
Affiliation(s)
- Gabriele Madonna
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Silvia Sale
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Chiara De Falco
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Valentina Santocchio
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Tiziana Di Matola
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Giuseppe Fiorentino
- UOC Fisiopatologia e Riabilitazione Respiratoria, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy;
| | - Caterina Pirozzi
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Anna D’Antonio
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Rocco Sabatino
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| | - Lidia Atripaldi
- Dipartimento di Scienze Mediche Traslazionali, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.A.); (U.A.)
| | - Umberto Atripaldi
- Dipartimento di Scienze Mediche Traslazionali, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.A.); (U.A.)
| | - Marcello Raffone
- UOC Microbiologia e Virologia, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy;
| | - Marcello Curvietto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Antonio Maria Grimaldi
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Lucia Festino
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Luigi Scarpato
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Marco Palla
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Michela Spatarella
- UOSD di Farmacia, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy;
| | - Francesco Perna
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli “Federico II”, 80131 Naples, Italy;
| | - Pellegrino Cerino
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy;
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Roberto Parrella
- UOC Malattie Infettive ad Indirizzo Respiratorio, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy;
| | | | - Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Napoli, Italy; (G.M.); (M.C.); (M.C.); (A.M.G.); (V.V.); (L.F.); (L.S.); (M.P.)
| | - Luigi Atripaldi
- UOC Biochimica Clinica, AORN Ospedali dei Colli—Monaldi—Cotugno—CTO, 80131 Napoli, Italy; (S.S.); (C.D.F.); (V.S.); (T.D.M.); (C.P.); (A.D.); (R.S.); (L.A.)
| |
Collapse
|
8
|
Zeng X, Feng J, Yang Y, Zhao R, Yu Q, Qin H, Wei L, Ji P, Li H, Wu Z, Zhang J. Screening of Key Genes of Sepsis and Septic Shock Using Bioinformatics Analysis. J Inflamm Res 2021; 14:829-841. [PMID: 33737824 PMCID: PMC7962593 DOI: 10.2147/jir.s301663] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/26/2021] [Indexed: 12/20/2022] Open
Abstract
Objective Sepsis is a disease associated with high mortality. We performed bioinformatic analysis to identify key biomarkers associated with sepsis and septic shock. Methods The top 20% of genes showing the greatest variance between sepsis and controls in the GSE13904 dataset (children) were screened by co-expression network analysis. The differentially expressed genes (DEGs) were identified through analyzing differential gene expression between sepsis patients and control in the GSE13904 (children) and GSE154918 (adult) data sets. Intersection analysis of module genes and DEGs was performed to identify common DEGs for enrichment analysis, protein-protein interaction network (PPI network) analysis, and Short Time-series Expression Miner (STEM) analysis. The PPI network genes were ranked by degree of connectivity, and the top 100 sepsis-associated genes were identified based on the area under the receiver operating characteristic curve (AUC). In addition, we evaluated differences in immune cell infiltration between sepsis patients and controls in children (GSE13904, GSE25504) and adults (GSE9960, GSE154918). Finally, we analyzed differences in DNA methylation levels between sepsis patients and controls in GSE138074 (adults). Results The common genes were associated mainly with up-regulated inflammatory and metabolic responses, as well as down-regulated immune responses. Sepsis patients showed lower infiltration by most types of immune cells. Genes in the PPI network with AUC values greater than 0.9 in both GSE13904 (children) and GSE154918 (adults) were screened as key genes for diagnosis. These key genes (MAPK14, FGR, RHOG, LAT, PRKACB, UBE2Q2, ITK, IL2RB, and CD247) were also identified in STEM analysis to be progressively dysregulated across controls, sepsis patients and patients with septic shock. In addition, the expression of MAPK14, FGR, and CD247 was modified by methylation. Conclusion This study identified several potential diagnostic genes and inflammatory and metabolic responses mechanisms associated with the development of sepsis.
Collapse
Affiliation(s)
- Xiaoliang Zeng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Jihua Feng
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Yanli Yang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Ruzhi Zhao
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Qiao Yu
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Han Qin
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Lile Wei
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Pan Ji
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Hongyuan Li
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Zimeng Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| | - Jianfeng Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, People's Republic of China
| |
Collapse
|
9
|
Manibalan S, Thirukumaran K, Varshni M, Shobana A, Achary A. Report on biopharmaceutical profile of recent biotherapeutics and insilco docking studies on target bindings of known aptamer biotherapeutics. Biotechnol Genet Eng Rev 2021; 36:57-80. [PMID: 33393433 DOI: 10.1080/02648725.2020.1858395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Accumulated Toxicity, disease recurrence and drug resistivity problems have been observed due to the synthetic and semisynthetic therapeutic practices, which alternatively led to focus on Bio-therapeutics production than xenobiotics. Quick plasma clearance and high potency are the reasons for trending research with huge pharma market of numerous Bio-therapeutics than ever before. Researchers proved that most of the nano and micro Bio-therapeutics have multiple beneficial therapeutic effects. We have analyzed the past, and present scenario of some notable clinically approved Bio-therapeutics to identify the future formulation needs with advanced techniques. Protein-related drugs are the foremost Bio-therapeutics such as antibodies, enzymes, and short, fragmented polypeptides show aggregation properties during storage, naked peptide moieties are resisted by the polar cell membrane, and also the antidrug antibodies were reported. Even though Nucleic acid nano-bodies are excellent target binders than proteins, they had only a few minutes of half-life. Maintaining homogeneousness upon storage of Bio-therapeutics is still a significant challenge in industrial-scale formulation. Notably, plant systems are identified as most useful cost-effective hosts to produce human enzymes than animal systems without any possible viral loads. Irrespective of numerous advancements in routes of administration and additives, subcutaneous is still a golden one to achieve better dynamics. Additionally, the interactions and effective bonds made by each class of well-known aptamer biotherapeutics which are considered as future drugs were studied.
Collapse
Affiliation(s)
- Subramaniyan Manibalan
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Kandasamy Thirukumaran
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Mathimaran Varshni
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Ayyasamy Shobana
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| | - Anant Achary
- Center for Research, Department of Biotechnology, Kamaraj College of Engineering and Technology , Madurai, India
| |
Collapse
|
10
|
Nowill AE, de Campos-Lima PO. Immune Response Resetting as a Novel Strategy to Overcome SARS-CoV-2-Induced Cytokine Storm. THE JOURNAL OF IMMUNOLOGY 2020; 205:2566-2575. [PMID: 32958687 DOI: 10.4049/jimmunol.2000892] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 08/30/2020] [Indexed: 12/15/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which rapidly became a pandemic of global proportions. Sepsis is commonly present with high lethality in the severe forms of the disease. The virus-induced cytokine storm puts the immune system in overdrive at the expense of the pathogen-specific immune response and is likely to underlie the most advanced COVID-19 clinical features, including sepsis-related multiple organ dysfunction as well as the pathophysiological changes found in the lungs. We review the major therapeutic strategies that have been considered for sepsis and might be amenable to repurposing for COVID-19. We also discuss two different immunization strategies that have the potential to confer antiviral heterologous protection: innate-induced trained immunity and adaptive-induced immune response resetting.
Collapse
Affiliation(s)
- Alexandre E Nowill
- Integrated Center for Pediatric OncoHaematological Research, State University of Campinas, Campinas SP 13083-888, Brazil;
| | - Pedro O de Campos-Lima
- Boldrini Children's Center, Campinas SP 13083-210, Brazil; and .,Functional and Molecular Biology Graduate Program, Institute of Biology, State University of Campinas, Campinas SP 13083-865, Brazil
| |
Collapse
|
11
|
Doerflinger M, Reljic B, Menassa J, Nedeva C, Jose I, Faou P, Mackiewicz L, Mansell A, Pellegrini M, Hotchkiss R, Puthalakath H. Circulating BiP/Grp78 is a novel prognostic marker for sepsis-mediated immune cell death. FEBS J 2020; 288:1809-1821. [PMID: 32894892 DOI: 10.1111/febs.15552] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/23/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
Abstract
Sepsis remains to be a major contributor to mortality in ICUs, and immune suppression caused by immune cell apoptosis determines the overall patient survival. However, diagnosis of sepsis-induced lymphopenia remains problematic with no accurate prognostic techniques or biomarkers for cell death available. Developing reliable prognostic tools for sepsis-mediated cell death is not only important for identifying patients at increased risk of immune suppression but also to monitor treatment progress of currently trialed immunotherapy strategies. We have previously shown an important role for endoplasmic reticulum stress (ER stress) in inducing sepsis-mediated cell death and here report on the identification of a secreted form of the ER chaperone BiP (immunoglobulin binding protein) as a novel circulating prognostic biomarker for immune cell death and ER stress during sepsis. Using biochemical purification and mass spectrometry coupled with an established in vitro sepsis cell death assay, we identified BiP/Grp78 as a factor secreted by lipopolysaccharide-activated macrophages that is capable of inducing cell death in target cells. Quantitative ELISA analysis showed significantly elevated levels of circulating BiP in mice undergoing polymicrobial sepsis, which was absent in Bim-/- mice that are protected from sepsis-induced lymphopenia. Using blood serum from human sepsis patients, we could detect a significant difference in levels of secreted BiP in sepsis patients compared to nonseptic controls, suggesting that secreted circulating BiP could indeed be used as a prognostic marker that is directly correlative to immune cell death during sepsis.
Collapse
Affiliation(s)
- Marcel Doerflinger
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia.,Biochemistry and Molecular Biology, Clayton, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Boris Reljic
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia.,Biochemistry and Molecular Biology, Clayton, Vic., Australia
| | - Joseph Menassa
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Christina Nedeva
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Irvin Jose
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Pierre Faou
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| | - Liana Mackiewicz
- Biochemistry and Molecular Biology, Clayton, Vic., Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, Vic., Australia
| | - Ashley Mansell
- Hudson Institute of Medical Research, Clayton, Vic., Australia
| | - Marc Pellegrini
- Biochemistry and Molecular Biology, Clayton, Vic., Australia
| | - Richard Hotchkiss
- Department of Pediatrics and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hamsa Puthalakath
- Department of Biochemistry and Genetics, La Trobe University, Melbourne, Vic., Australia
| |
Collapse
|
12
|
Short SS, Papillon SC, Ford HR. Sepsis. PEDIATRIC SURGERY 2020:461-476. [DOI: 10.1007/978-3-662-43588-5_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
13
|
Infection-induced innate antimicrobial response disorders: from signaling pathways and their modulation to selected biomarkers. Cent Eur J Immunol 2020; 45:104-116. [PMID: 32425688 PMCID: PMC7226557 DOI: 10.5114/ceji.2020.94712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Severe infections are a major public health problem responsible for about 40-65% of hospitalizations in intensive care units (ICU). The high mortality (30-50%) of persons diagnosed with severe infection is caused by largely unknown mechanisms of sepsis-induced immune system response. Severe infections with dynamic progress are accompanied with SIRS (systemic inflammatory reaction syndrome) and CARS (compensatory anti-inflammatory response syndrome), and require a biological treatment appropriate to the phase of immune response. The mechanisms responsible for severe infection related to immune system response particularly attract extensive interest of non-specific defense mechanisms, including signaling pathways of Toll-like receptors (mainly TLR4 and TLR2) that recognize distinct pathogen-associated molecular patterns (PAMP) and play a critical role in innate immune response. There are attempts of treatment, followed by blocking ligand binding with TLR or modulation of intracellular signaling pathways, to inhibit signal transduction. Moreover, researches regarding new and more efficient diagnostics biomarkers were mostly focused on indicators related to innate response to infection as well as connections of pro-inflammatory response with anti-inflammatory response.According to these studies, in case of ICU septic patients with high-risk of mortality, the solution for the problem will require mainly early immune and genetic diagnostics (e.g. cytokines, microRNA, cluster of differentiation-64 [CD64], triggering receptor expressed on myeloid cells-1 [TREM-1], and high mobility group box 1 protein [HMGB1]).
Collapse
|
14
|
The Pathogenesis of Sepsis and Potential Therapeutic Targets. Int J Mol Sci 2019; 20:ijms20215376. [PMID: 31671729 PMCID: PMC6862039 DOI: 10.3390/ijms20215376] [Citation(s) in RCA: 405] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/05/2019] [Accepted: 10/25/2019] [Indexed: 02/06/2023] Open
Abstract
Sepsis is defined as “a life-threatening organ dysfunction caused by a host’s dysfunctional response to infection”. Although the treatment of sepsis has developed rapidly in the past few years, sepsis incidence and mortality in clinical treatment is still climbing. Moreover, because of the diverse manifestations of sepsis, clinicians continue to face severe challenges in the diagnosis, treatment, and management of patients with sepsis. Here, we review the recent development in our understanding regarding the cellular pathogenesis and the target of clinical diagnosis of sepsis, with the goal of enhancing the current understanding of sepsis. The present state of research on targeted therapeutic drugs is also elaborated upon to provide information for the treatment of sepsis.
Collapse
|
15
|
Le KTT, Chu X, Jaeger M, Plantinga JA, Matzaraki V, Withoff S, Joosten LAB, Netea MG, Wijmenga C, Li Y, Moser J, Kumar V. Leukocyte-Released Mediators in Response to Both Bacterial and Fungal Infections Trigger IFN Pathways, Independent of IL-1 and TNF-α, in Endothelial Cells. Front Immunol 2019; 10:2508. [PMID: 31708927 PMCID: PMC6824321 DOI: 10.3389/fimmu.2019.02508] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022] Open
Abstract
In sepsis, dysregulated immune responses to infections cause damage to the host. Previous studies have attempted to capture pathogen-induced leukocyte responses. However, the impact of mediators released after pathogen-leukocyte interaction on endothelial cells, and how endothelial cell responses vary depending on the pathogen-type is lacking. Here, we comprehensively characterized the transcriptomic responses of human leukocytes and endothelial cells to Gram negative-bacteria, Gram positive-bacteria, and fungi. We showed that whole pathogen lysates induced strong activation of leukocytes but not endothelial cells. Interestingly, the common response of leukocytes to various pathogens converges on endothelial activation. By exposing endothelial cells to leukocyte-released mediators, we observed a strong activation of endothelial cells at both transcription and protein levels. By adding IL-1RA and TNF-α antibody in leukocyte-released mediators before exposing to endothelial cells, we identified specific roles for IL-1 and TNF-α in driving the most, but not all, endothelial activation. We also showed for the first time, activation of interferon response by endothelial cells in response to leukocyte-released mediators, independently from IL-1 and TNF-α pathways. Our study therefore, not only provides pathogen-dependent transcriptional changes in leukocytes and endothelial cells during infections, but also reveals a role for IFN, together with IL1 and TNFα signaling, in mediating leukocyte-endothelial interaction in infections.
Collapse
Affiliation(s)
- Kieu T T Le
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Xiaojing Chu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martin Jaeger
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Josée A Plantinga
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Vasiliki Matzaraki
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sebo Withoff
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Immunology, K.G. Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
| | - Yang Li
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jill Moser
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Vinod Kumar
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Internal Medicine and Radboud Centre for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
16
|
Honokiol Increases CD4+ T Cell Activation and Decreases TNF but Fails to Improve Survival Following Sepsis. Shock 2019; 50:178-186. [PMID: 29023360 DOI: 10.1097/shk.0000000000001021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Honokiol is a biphenolic isolate extracted from the bark of the magnolia tree that has been used in traditional Chinese and Japanese medicine, and has more recently been investigated for its anti-inflammatory and antibacterial properties. Honokiol has previously been demonstrated to improve survival in sepsis models that have rapid 100% lethality. The purpose of this study was to determine the impact of Honokiol on the host response in a model of sepsis that more closely approximates human disease. Male and female C57BL/6 mice underwent cecal ligation and puncture to induce polymicrobial intra-abdominal sepsis. Mice were then randomized to receive an injection of either Honokiol (120 mg/kg/day) or vehicle and were sacrificed after 24 h for functional studies or followed 7 days for survival. Honokiol treatment after sepsis increased the frequency of CD4 T cells and increased activation of CD4 T cells as measured by the activation marker CD69. Honokiol also increased splenic dendritic cells. Honokiol simultaneously decreased frequency and number of CD8 T cells. Honokiol decreased systemic tumor necrosis factor without impacting other systemic cytokines. Honokiol did not have a detectable effect on kidney function, lung physiology, liver function, or intestinal integrity. In contrast to prior studies of Honokiol in a lethal model of sepsis, Honokiol did not alter survival at 7 days (70% mortality for Honokiol vs. 60% mortality for vehicle). Honokiol is thus effective in modulating the host immune response and inflammation following a clinically relevant model of sepsis but is not sufficient to alter survival.
Collapse
|
17
|
Inhibition of transmembrane TNF-α shedding by a specific antibody protects against septic shock. Cell Death Dis 2019; 10:586. [PMID: 31383857 PMCID: PMC6683172 DOI: 10.1038/s41419-019-1808-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 01/21/2023]
Abstract
Transmembrane TNF-α (tmTNF-α) and secretory TNF-α (sTNF-α) display opposite effects in septic shock. Reducing tmTNF-α shedding can offset the detrimental effects of sTNF-α and increase the beneficial effect of tmTNF-α. We previously developed a monoclonal antibody that is specific for tmTNF-α and does not cross-react with sTNF-α. In this study, we show that this antibody can specifically suppress tmTNF-α shedding by competing with a TNF-α converting enzyme that cleaves the tmTNF-α ectodomain to release sTNF-α. This tmTNF-α antibody significantly inhibited LPS-induced secretion of interleukin (IL)-1β, IL-6, interferon-β, and nitric oxide by monocytes/macrophages, and protected mice from septic shock induced by lipopolysaccharide (LPS) or cecal ligation and puncture, while reducing the bacterial load. The mechanism associated with the protective effect of this tmTNF-α antibody involved promotion of LPS-induced toll-like receptor 4 (TLR4) internalization and degradation by recruiting Triad3A to TLR4. Moreover, the tmTNF-α antibody inhibited LPS-induced activation of nuclear factor-κB and interferon regulatory factor 3 pathways by upregulating expression of A20 and monocyte chemotactic protein-induced protein 1. Similarly, treatment of macrophages with exogenous tmTNF-α suppressed LPS/TLR4 signaling and release of proinflammatory cytokines, indicating that increased levels of tmTNF-α promoted by the antibody contributed to its inhibitory effect. Thus, use of this tmTNF-α antibody for specific suppression of tmTNF-α shedding may be a promising strategy to treat septic shock.
Collapse
|
18
|
Leisewitz A, Goddard A, De Gier J, Van Engelshoven J, Clift S, Thompson P, Schoeman JP. Disease severity and blood cytokine concentrations in dogs with natural Babesia rossi infection. Parasite Immunol 2019; 41:e12630. [PMID: 31063593 DOI: 10.1111/pim.12630] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/28/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
Abstract
AIMS Babesia rossi causes severe disease in dogs. Here, we describe the association between serum cytokine concentrations and disease severity. METHODS Seventeen controls and 55 PCR confirmed B rossi-infected dogs were included. Diseased dogs were subdivided into 23 critically ill and 32 relatively well cases. Serum concentrations of 11 cytokines and biochemical markers of disease severity were determined. RESULTS Significant differences were detected for IL-6, IL-8, IL-10, MCP-1 and TNF-α between the groups. Generally, the more complicated the disease, the more pro-inflammatory the cytokine milieu. IL-8 showed a reverse trend and was negatively correlated with disease severity. IL-6, MCP-1 and TNF-α were also significantly higher in the dogs that died (n = 9) compared to the dogs that survived (n = 46). IL-8 showed the opposite. MCP-1 and TNF-α were negatively correlated with biochemical markers of severity. Glucose was negatively correlated with IL-6. Cortisol, peripheral parasite density and band neutrophil count were positively correlated, whilst thyroid hormone was negatively correlated with IL-6, MCP-1 and TNF-α. CONCLUSIONS As in malaria and sepsis, B rossi infection induces a pro-inflammatory cytokine storm that correlates with disease severity and adverse outcome. The multiplicity of cytokines involved argues for redundancy in the system once the disease is established.
Collapse
Affiliation(s)
- Andrew Leisewitz
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Amelia Goddard
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Jill De Gier
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Jessica Van Engelshoven
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Sarah Clift
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Peter Thompson
- Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Johan P Schoeman
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
19
|
Batsaikhan B, Wang JY, Scerba MT, Tweedie D, Greig NH, Miller JP, Hoffer BJ, Lin CT, Wang JY. Post-Injury Neuroprotective Effects of the Thalidomide Analog 3,6'-Dithiothalidomide on Traumatic Brain Injury. Int J Mol Sci 2019; 20:ijms20030502. [PMID: 30682785 PMCID: PMC6387371 DOI: 10.3390/ijms20030502] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/18/2019] [Accepted: 01/21/2019] [Indexed: 01/09/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and disability worldwide. Long-term deficits after TBI arise not only from the direct effects of the injury but also from ongoing processes such as neuronal excitotoxicity, inflammation, oxidative stress and apoptosis. Tumor necrosis factor-α (TNF-α) is known to contribute to these processes. We have previously shown that 3,6′-dithiothalidomide (3,6′-DT), a thalidomide analog that is more potent than thalidomide with similar brain penetration, selectively inhibits the synthesis of TNF-α in cultured cells and reverses behavioral impairments induced by mild TBI in mice. In the present study, we further explored the therapeutic potential of 3,6′-DT in an animal model of moderate TBI using Sprague-Dawley rats subjected to controlled cortical impact. A single dose of 3,6′-DT (28 mg/kg, i.p.) at 5 h after TBI significantly reduced contusion volume, neuronal degeneration, neuronal apoptosis and neurological deficits at 24 h post-injury. Expression of pro-inflammatory cytokines in the contusion regions were also suppressed at the transcription and translation level by 3,6′-DT. Notably, neuronal oxidative stress was also suppressed by 3,6′-DT. We conclude that 3,6′-DT may represent a potential therapy to ameliorate TBI-induced functional deficits.
Collapse
Affiliation(s)
- Buyandelger Batsaikhan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.
| | - Jing-Ya Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.
| | - Michael T Scerba
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD 21224, USA.
| | - Jonathan P Miller
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Barry J Hoffer
- Department of Neurological Surgery, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Chih-Tung Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei 11031, Taiwan.
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
20
|
Shirakura K, Ishiba R, Kashio T, Funatsu R, Tanaka T, Fukada SI, Ishimoto K, Hino N, Kondoh M, Ago Y, Fujio Y, Yano K, Doi T, Aird WC, Okada Y. The Robo4-TRAF7 complex suppresses endothelial hyperpermeability in inflammation. J Cell Sci 2019; 132:jcs.220228. [PMID: 30510113 DOI: 10.1242/jcs.220228] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/28/2018] [Indexed: 01/15/2023] Open
Abstract
Roundabout guidance receptor 4 (Robo4) is an endothelial cell-specific receptor that stabilizes the vasculature in pathological angiogenesis. Although Robo4 has been shown to suppress vascular hyperpermeability induced by vascular endothelial growth factor (VEGF) in angiogenesis, the role of Robo4 in inflammation is poorly understood. In this study, we investigated the role of Robo4 in vascular hyperpermeability during inflammation. Endotoxemia models using Robo4 -/- mice showed increased mortality and vascular leakage. In endothelial cells, Robo4 suppressed tumor necrosis factor α (TNFα)-induced hyperpermeability by stabilizing VE-cadherin at cell junctions, and deletion assays revealed that the C-terminus of Robo4 was involved in this suppression. Through binding and localization assays, we demonstrated that in endothelial cells, Robo4 binds to TNF receptor-associated factor 7 (TRAF7) through interaction with the C-terminus of Robo4. Gain- and loss-of-function studies of TRAF7 with or without Robo4 expression showed that TRAF7 is required for Robo4-mediated suppression of hyperpermeability. Taken together, our results demonstrate that the Robo4-TRAF7 complex is a novel negative regulator of inflammatory hyperpermeability. We propose this complex as a potential future target for protection against inflammatory diseases.
Collapse
Affiliation(s)
- Keisuke Shirakura
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Ryosuke Ishiba
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Taito Kashio
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Risa Funatsu
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Toru Tanaka
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - So-Ichiro Fukada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Kenji Ishimoto
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Nobumasa Hino
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Yukio Ago
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Yasushi Fujio
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - Kiichiro Yano
- The Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Takefumi Doi
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| | - William C Aird
- The Center for Vascular Biology Research and Division of Molecular and Vascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0781, Japan
| |
Collapse
|
21
|
Lebedev NV, Klimov AE, Cherepanova ON, Barkhudarov AA. [Inflammatory markers in diagnosis and prognosis of abdominal sepsis]. Khirurgiia (Mosk) 2018:92-98. [PMID: 30531745 DOI: 10.17116/hirurgia201810192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
For today, it is necessary to recognize, that treatment of patients with abdominal sepsis remains the basic problem in urgent surgery due to the invariably high mortality. Early diagnostics and targeted therapy are the key points for improving of sepsis outcome. At present, researchers around the world have proposed a large number of biological markers for diagnosing sepsis and predicting mortality. Ideally, doctors can use biomarkers for risk stratification, diagnosing, monitoring of treatment effectiveness and outcome prediction. The biomarker is a laboratory parameter that can be objectively measured and characterized as an indicator of normal and pathological biological processes. The article presents the modern concept of the sepsis pathogenesis for understanding the role of various biomarkers and inflammatory indicators in its development. We have analyzed literature data and summarized information on the possible use of biological markers and their combinations in the early detection of sepsis, for monitoring sepsis and predicting its outcome.
Collapse
Affiliation(s)
- N V Lebedev
- Chair of Faculty-Based Surgery, Peoples' Friendship University of Russia, Moscow, Russia
| | - A E Klimov
- Chair of Faculty-Based Surgery, Peoples' Friendship University of Russia, Moscow, Russia
| | - O N Cherepanova
- Chair of Faculty-Based Surgery, Peoples' Friendship University of Russia, Moscow, Russia
| | - A A Barkhudarov
- Chair of Faculty-Based Surgery, Peoples' Friendship University of Russia, Moscow, Russia
| |
Collapse
|
22
|
Pulmonary and muscle profile in pneumosepsis: A temporal analysis of inflammatory markers. Cytokine 2018; 114:128-134. [PMID: 30470659 DOI: 10.1016/j.cyto.2018.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/06/2018] [Accepted: 11/12/2018] [Indexed: 01/09/2023]
Abstract
In sepsis, greater understanding of the inflammatory mechanism involved would provide insights into the condition and into its extension to the muscular apparatus in critically ill patients. Therefore, this study evaluates the inflammatory profile of pneumosepsis induced by Klebsiella pneumoniae (K.p.) in lungs and skeletal muscles during the first 72 h. Male BALB/c mice were divided into 4 groups, submitted to intratracheal inoculation of K.p. at a concentration of 2 × 108 (PS) or PBS, and assessed after 24 (PS24), 48 (PS48) and 72 (PS72) hours. The Maximum Physical Capacity Test (MPCT) was performed before and after induction. Pulmonary inflammation was assessed by total cell number, nitric oxide levels (NOx), IL-1β and TNF-α levels in bronchoalveolar lavage fluid (BALF); inflammation and muscle trophism were evaluated by the levels of TNF-α, IL-6, TGF-β and BDNF by ELISA and NF-κB by western blotting in muscle tissue. Cells and colony forming units (CFU) were also analyzed in blood samples. The PS groups showed an increase in total cells in the BALF (p < 0.05), as well in the number of granulocytes in the blood (p < 0.05) and a decrease in performance in the MPCT (p < 0.05). NOx levels showed significant increase in PS72, when compared to Control group (p = 0.03). The PS24 showed a significant increase lung in TNF-α levels (p < 0.001) and in CFU (p = 0.013). We observed an increase in muscular IL-6 and nuclear NF-κB levels in PS24 group, when compared to PS48 and Control groups (p < 0.05). Nevertheless, mild signs of injury in the skeletal muscle tissue does not support the idea of an early muscular injury in this experimental model, suggesting that the low performance of the animals during the MPCT may be related to lung inflammation.
Collapse
|
23
|
Armacki M, Trugenberger AK, Ellwanger AK, Eiseler T, Schwerdt C, Bettac L, Langgartner D, Azoitei N, Halbgebauer R, Groß R, Barth T, Lechel A, Walter BM, Kraus JM, Wiegreffe C, Grimm J, Scheffold A, Schneider MR, Peuker K, Zeißig S, Britsch S, Rose-John S, Vettorazzi S, Wolf E, Tannapfel A, Steinestel K, Reber SO, Walther P, Kestler HA, Radermacher P, Barth TF, Huber-Lang M, Kleger A, Seufferlein T. Thirty-eight-negative kinase 1 mediates trauma-induced intestinal injury and multi-organ failure. J Clin Invest 2018; 128:5056-5072. [PMID: 30320600 DOI: 10.1172/jci97912] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 08/28/2018] [Indexed: 12/17/2022] Open
Abstract
Dysregulated intestinal epithelial apoptosis initiates gut injury, alters the intestinal barrier, and can facilitate bacterial translocation leading to a systemic inflammatory response syndrome (SIRS) and/or multi-organ dysfunction syndrome (MODS). A variety of gastrointestinal disorders, including inflammatory bowel disease, have been linked to intestinal apoptosis. Similarly, intestinal hyperpermeability and gut failure occur in critically ill patients, putting the gut at the center of SIRS pathology. Regulation of apoptosis and immune-modulatory functions have been ascribed to Thirty-eight-negative kinase 1 (TNK1), whose activity is regulated merely by expression. We investigated the effect of TNK1 on intestinal integrity and its role in MODS. TNK1 expression induced crypt-specific apoptosis, leading to bacterial translocation, subsequent septic shock, and early death. Mechanistically, TNK1 expression in vivo resulted in STAT3 phosphorylation, nuclear translocation of p65, and release of IL-6 and TNF-α. A TNF-α neutralizing antibody partially blocked development of intestinal damage. Conversely, gut-specific deletion of TNK1 protected the intestinal mucosa from experimental colitis and prevented cytokine release in the gut. Finally, TNK1 was found to be deregulated in the gut in murine and porcine trauma models and human inflammatory bowel disease. Thus, TNK1 might be a target during MODS to prevent damage in several organs, notably the gut.
Collapse
Affiliation(s)
- Milena Armacki
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | - Ann K Ellwanger
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Tim Eiseler
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Christiane Schwerdt
- Waldkrankenhaus "Rudolph Elle" Eisenberg, Lehrstuhl für Orthopädie Uniklinik Jena, Jena, Germany
| | - Lucas Bettac
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, and
| | - Ninel Azoitei
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Rebecca Halbgebauer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Rüdiger Groß
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Tabea Barth
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - André Lechel
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Benjamin M Walter
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | | | | | | | - Annika Scheffold
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | | | - Kenneth Peuker
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Sebastian Zeißig
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Stefan Britsch
- Institute of Molecular and Cellular Anatomy, Ulm University, Ulm, Germany
| | | | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology, Ulm University, Ulm, Germany
| | | | | | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Bundeswehrkrankenhaus Ulm, Ulm, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, and
| | - Paul Walther
- Central Facility for Electron Microscopy, University of Ulm, Ulm, Germany
| | | | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, Ulm University, Ulm, Germany
| | | | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Alexander Kleger
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
24
|
Neural pathways involved in infection-induced inflammation: recent insights and clinical implications. Clin Auton Res 2018. [PMID: 29541878 DOI: 10.1007/s10286-018-0518-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Although the immune and nervous systems have long been considered independent biological systems, they turn out to mingle and interact extensively. The present review summarizes recent insights into the neural pathways activated by and involved in infection-induced inflammation and discusses potential clinical applications. The simplest activation concerns a reflex action within C-fibers leading to neurogenic inflammation. Low concentrations of pro-inflammatory cytokines or bacterial fragments may also act on these afferent nerve fibers to signal the central nervous system and bring about early fever, hyperalgesia and sickness behavior. In the brain, the preoptic area and the paraventricular hypothalamus are part of a neuronal network mediating sympathetic activation underlying fever while brainstem circuits play a role in the reduction of food intake after systemic exposure to bacterial fragments. A vagally-mediated anti-inflammatory reflex mechanism has been proposed and, in turn, questioned because the major immune organs driving inflammation, such as the spleen, are not innervated by vagal efferent fibers. On the contrary, sympathetic nerves do innervate these organs and modulate immune cell responses, production of inflammatory mediators and bacterial dissemination. Noradrenaline, which is both released by these fibers and often administered during sepsis, along with adrenaline, may exert pro-inflammatory actions through the stimulation of β1 adrenergic receptors, as antagonists of this receptor have been shown to exert anti-inflammatory effects in experimental sepsis.
Collapse
|
25
|
Chen X, Feng Y, Shen X, Pan G, Fan G, Gao X, Han J, Zhu Y. Anti-sepsis protection of Xuebijing injection is mediated by differential regulation of pro- and anti-inflammatory Th17 and T regulatory cells in a murine model of polymicrobial sepsis. JOURNAL OF ETHNOPHARMACOLOGY 2018; 211:358-365. [PMID: 28987599 DOI: 10.1016/j.jep.2017.10.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 10/01/2017] [Accepted: 10/02/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xuebijing injection (XBJ), a Chinese herbal medicine containing extracts from 5 herbs, is frequently used as an add-on with standard therapies to treat sepsis or septic shock with fewer side effects in China. Nonetheless, its mechanism of action on septic shock remains to be unveiled. We explored the differential effects of XBJ on subtypes of CD4+ T cell differentiation and septic shock protection in a murine model to understand the contribution of XBJ to regulation of the inflammation-immune axis function. MATERIALS AND METHODS In vitro T cell differentiation assays were performed to determine the effect of XBJ on CD4+ regulatory T cell and T helper cell differentiation. Besides, 2ml/kg, 6ml/kg- and 18ml/kg of XBJ were administered to different groups of septic mice once/day for 5 days after cecal ligation and puncture (CLP) surgeries. 36h after CLP, serum levels of pro-inflammatory cytokine TNF-α and IL-6 were determined with Elisa. Frequencies of CD4+ T cells were analyzed after staining with Tregs and T helper cell lineage specific antibodies by flow cytometer. RESULTS XBJ at 18ml/kg stimulated Treg differentiation and moderately inhibited Th17 differentiation in vitro. Accordingly, 18ml/kg XBJ facilitated the expansion of IL-10+ Tregs and normalized pro-inflammatory Th17 population in septic mice. This regimen also significantly reduced serum levels of inflammatory cytokines TNF-α and IL-6 in septic mice. Additionally, 18ml/kg XBJ injection effectively prevented neutrophil infiltration into the lung and kidney and improved survival in this septic shock model. CONCLUSIONS In summary, XBJ improves survival in septic shock partially through preventing cytokine storm, inhibiting inflammation and regulating the balance of Tregs and Th17 cells. Thus, higher dose of XBJ is a potential regimen to benefit septic shock patients.
Collapse
Affiliation(s)
- Xi Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Yuxin Feng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Xiya Shen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China; State Key Laboratory of Medicinal Chemical Biology, and Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin 300071, China
| | - Guixiang Pan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China
| | - Guanwei Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China
| | - Jihong Han
- State Key Laboratory of Medicinal Chemical Biology, and Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin 300071, China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Nankai District, Tianjin 300193, China; Research and Development Center of TCM, Tianjin International Joint Academy of Biotechnology & Medicine, 220 Dongting Road, TEDA, Tianjin 300457, China; Molecular Cardiology Research Institute, Tufts Medical Center and Tufts University School of Medicine, 750 Washington Street, Boston, MA 02111, USA.
| |
Collapse
|
26
|
Yadav N, Chandra H. Modulation of alveolar macrophage innate response in proinflammatory-, pro-oxidant-, and infection- models by mint extract and chemical constituents: Role of MAPKs. Immunobiology 2017; 223:49-56. [PMID: 29031422 DOI: 10.1016/j.imbio.2017.10.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/11/2017] [Accepted: 10/03/2017] [Indexed: 12/23/2022]
Abstract
There is a continuing need for discovering novel primary or adjunct therapeutic agents to treat inflammatory conditions and infections. Natural products have inspired the discovery of several modern therapeutics; however, there is a paucity of mechanistic information on their mode of action. This study investigated the therapeutic potential and mode of action of corn mint's (Mentha arvensis) leaf extract (ME) in alveolar macrophages (AMs) challenged with model pro-inflammatory (LPS), pro- oxidant (LPS or H2O2), and infection (Mycobacterium) agents and contribution of its dominant constituents rosmarinic acid, l-menthol, and l-menthone. LPS-induced inflammatory response in the murine AM cell line MH-S was significantly reduced in terms of pro-inflammatory cytokines (TNF-α, IL-1α) and nitric oxide (NO) when pre- or post-treated with ME. The ME pretreatment of macrophages led to a significant increase (P≤0.05) in phagocytic activity toward Mycobacterium smegmatis and a greater pathogen clearance in 24h in both ME pre-treated (P≤0.05) and post-treated cells. Significant attenuation (P≤0.01) of reactive oxygen species (ROS) production in LPS- or H2O2-treated macrophages by pretreatment with whole mint extract (ME) was accounted for in part by the mint constituents rosmarinic acid and l-menthone. Attenuation of pro-inflammatory response by ME pretreatment coincided with the significant reduction in total and phosphorylated JNK1/2, decrease in total p38, and increase in phospho-ERK1/2 thereby implying a role of differential modulation of MAPKs. Taken together, the results demonstrate that corn mint leaf components cause potent anti-inflammatory, anti-oxidant, and anti-infection effects in AMs via suppression of the production of cytokines/soluble mediators and ROS and increased pathogen clearance, respectively. To our knowledge, this is the first report on the mode of action of corn mint targeting the alveolar macrophages and on the potential role of MAPKs in immunomodulation by this product.
Collapse
Affiliation(s)
- Niket Yadav
- Microbial Pathogenesis and Immunotoxicology Laboratory, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0056, USA
| | - Harish Chandra
- Microbial Pathogenesis and Immunotoxicology Laboratory, Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0056, USA.
| |
Collapse
|
27
|
Tumor necrosis factor-α -308 G/A polymorphism and risk of sepsis, septic shock, and mortality: an updated meta-analysis. Oncotarget 2017; 8:94910-94919. [PMID: 29212277 PMCID: PMC5706923 DOI: 10.18632/oncotarget.20862] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/27/2017] [Indexed: 02/05/2023] Open
Abstract
Background The -308G/A polymorphism in the gene encoding tumor necrosis factor-α (TNF-α) has been implicated in sepsis risk in many studies but with variable results. This study aimed to comprehensively assess the evidence of association between this polymorphism and risk of sepsis and sepsis-related mortality. Materials and Methods PubMed, EMBASE and other databases were searched to identify relevant studies, and data were analyzed using Review Manager 5.0 and STATA 12.0. Results Data from 34 publications involving 12,284 subjects were meta-analyzed. Combined analysis revealed an association between TNF-α -308G/A gene polymorphism and risk of sepsis (AA+GA vs. GG, OR 1.35, 95% CI 1.10–1.67, P = 0.005). This association was observed in the Caucasian subgroup (OR 1.50, 95% CI 1.13–2.00, P = 0.006), but not in the Asian subgroup. Across the entire study population, the polymorphism was also significantly related to septic shock risk (OR 1.52, 95% CI 1.18–1.95, P = 0.001) but not to sepsis-related mortality (OR 0.99, 95% CI 0.71–1.40, P = 0.97). Conclusions This meta-analysis suggests that the -308G/A gene polymorphism in the TNF-α gene may contribute to risk of sepsis and septic shock, but not risk of mortality.
Collapse
|
28
|
Therapeutic targeting of HMGB1 during experimental sepsis modulates the inflammatory cytokine profile to one associated with improved clinical outcomes. Sci Rep 2017; 7:5850. [PMID: 28724977 PMCID: PMC5517568 DOI: 10.1038/s41598-017-06205-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/08/2017] [Indexed: 12/29/2022] Open
Abstract
Sepsis remains a significant health burden and a major clinical need exists for therapeutics to dampen the excessive and uncontrolled immune activation. Nuclear protein high mobility group box protein 1 (HMGB1) is released following cell death and is a late mediator in sepsis pathogenesis. While approaches targeting HMGB1 have demonstrated reduced mortality in pre-clinical models of sepsis, the impact of HMGB1 blockade on the complex septic inflammatory milieu and the development of subsequent immunosuppression remain enigmatic. Analysis of plasma samples obtained from septic shock patients established an association between increased HMGB1 and non-survival, higher APACHE II scores, and increased pro-inflammatory cytokine responses. Pre-clinically, administration of neutralising ovine anti-HMGB1 polyclonal antibodies improved survival in murine endotoxaemia and caecal ligation and puncture-induced sepsis models, and altered early cytokine profiles to one which corresponded to patterns observed in the surviving patient cohort. Additionally, anti-HMGB1 treated murine sepsis survivors were significantly more resistant to secondary bacterial infection and exhibited altered innate immune cell phenotypes and cytokine responses. These findings demonstrate that anti-HMGB1 antibodies alter inflammation in murine sepsis models and reduce sepsis mortality without potentiating immunosuppression.
Collapse
|
29
|
Ozer EK, Goktas MT, Kilinc I, Toker A, Bariskaner H, Ugurluoglu C, Iskit AB. Infliximab alleviates the mortality, mesenteric hypoperfusion, aortic dysfunction, and multiple organ damage in septic rats. Can J Physiol Pharmacol 2017; 95:866-872. [DOI: 10.1139/cjpp-2016-0628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Tumor necrosis factor-alpha (TNF-α) is a pivotal mediator that triggers inflammatory process, oxidative stress, and multiple organ injury in sepsis. We investigated the effects of infliximab on survival, mesenteric artery blood flow (MBF), vascular reactivity, and oxidative and inflammatory injuries in cecal ligation and puncture (CLP)-induced sepsis. Wistar rats were divided into Sham, CLP, Sham+infliximab, and CLP+infliximab subgroups. Twenty-four hours before the operations, rats were injected intraperitoneally with infliximab (7 mg/kg) or vehicle (saline; 1 mL/kg). Twenty hours after the operations, MBF and phenylephrine responses of isolated aortic rings were measured. Tissue damages were examined biochemically and histopathologically. Furthermore, survival rates were monitored throughout 96 h. Infliximab improved survival, mesenteric perfusion, and aortic function after CLP. Increases of serum AST, ALT, LDH, BUN, Cr, and inflammatory cytokines (tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6) induced by CLP were blocked by infliximab. Infliximab prevented malondialdehyde elevations in septic liver, lung, spleen, and kidney tissues, as well as glutathione reductions in septic liver, spleen, and kidney tissues. Protective effects of infliximab on multiple organ damage were also observed histopathologically. Infliximab showed protective effects in sepsis due to its improvement effects on mesenteric perfusion, aortic function, and its anti-inflammatory and antioxidative effects.
Collapse
Affiliation(s)
- Erdem Kamil Ozer
- Department of Pharmacology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Mustafa Tugrul Goktas
- Department of Pharmacology, Faculty of Medicine, Yildirim Beyazit University, Ankara, Turkey
| | - Ibrahim Kilinc
- Department of Biochemistry, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Aysun Toker
- Department of Biochemistry, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Hulagu Bariskaner
- Department of Pharmacology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Ceyhan Ugurluoglu
- Department of Pathology, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Alper Bektas Iskit
- Department of Pharmacology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
30
|
Dendoncker K, Libert C. Glucocorticoid resistance as a major drive in sepsis pathology. Cytokine Growth Factor Rev 2017; 35:85-96. [DOI: 10.1016/j.cytogfr.2017.04.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/07/2017] [Accepted: 04/19/2017] [Indexed: 01/07/2023]
|
31
|
Khafaga AF. Exogenous phosphatidylcholine supplementation retrieve aluminum-induced toxicity in male albino rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2017; 24:15589-15598. [PMID: 28523611 DOI: 10.1007/s11356-017-9151-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/01/2017] [Indexed: 05/25/2023]
Abstract
This study investigated the ameliorative potential of exogenous phosphatidylcholine (PC) against aluminum-induced toxicity in male albino rats. Four groups of rats were used for this study (N = 8): group I served as the control, group II (PC treated) received L-α-phosphatidylcholine (egg yolk-derived) 100 mg/kg bwt/day orally, group III (aluminum treated) received aluminum chloride 100 mg/kg bwt/day orally, and group VI (aluminum + PC treated) received similar oral dose of aluminum and PC (100 mg/kg bwt/day). Treatment was continued for 8 weeks. Results revealed that aluminum chloride treatment leading to a significant elevation in serum aspartate aminotransferase, serum alanine aminotransferase, urea, creatinine, malondialdehyde, serum cytokines (tumor necrosis factor-α, interleukin-6), and brain content of acetylcholine, as well as a significant reduction in serum-reduced glutathione, serum testosterone, and brain content of acetylcholinesterase. Moreover, aluminum administration caused significant histopathological alteration in liver, kidney, brain, testes, and epididymis. Co-treatment with exogenous PC resulted in significant improvement in intensity of histopathologic lesions, serum parameters, testosterone level, proinflammatory cytokines, and oxidative/antioxidative status. However, it does not affect the brain content of acetylcholine and acetylcholinesterase. Conclusively, treatment with exogenous PC can retrieve the adverse effect of aluminum toxicities through its antioxidative and anti-inflammatory properties.
Collapse
Affiliation(s)
- Asmaa Fahmy Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina-Rasheed, Behira, Egypt.
| |
Collapse
|
32
|
Borken F, Markwart R, Requardt RP, Schubert K, Spacek M, Verner M, Rückriem S, Scherag A, Oehmichen F, Brunkhorst FM, Rubio I. Chronic Critical Illness from Sepsis Is Associated with an Enhanced TCR Response. THE JOURNAL OF IMMUNOLOGY 2017; 198:4781-4791. [DOI: 10.4049/jimmunol.1700142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/10/2017] [Indexed: 12/15/2022]
|
33
|
Kesselheim AS, Tan YT, Avorn J. The roles of academia, rare diseases, and repurposing in the development of the most transformative drugs. Health Aff (Millwood) 2016; 34:286-93. [PMID: 25646109 DOI: 10.1377/hlthaff.2014.1038] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Transformative drugs, defined as pharmaceuticals that are both innovative and have groundbreaking effects on patient care, are the "holy grail" of drug research and development. The sources of drug innovation are often debated, with pharmaceutical manufacturers arguing that high drug prices support innovative output from their sector. We studied the developmental histories of twenty-six drugs or drug classes approved by the Food and Drug Administration between 1984 and 2009 that were judged by expert physicians to be transformative (in two cases, the first drug in a transformative class was approved before 1984). Most of the twenty-six were first approved early in the study period; only four were approved in 2000 or later. Many were based on discoveries made by academic researchers who were supported by federal government funding. Others were jointly developed in both publicly funded and commercial institutions; the fewest number of drugs had originated solely within pharmaceutical industry research programs. Nine of the twenty-six (35 percent) were repurposed from products developed for other indications, and ten (38 percent) were developed for rare diseases before much broader applicability was found. The insights from these case studies provide an experience-based foundation for policies to encourage the development of future transformative drugs.
Collapse
Affiliation(s)
- Aaron S Kesselheim
- Aaron S. Kesselheim is an associate professor of medicine at Harvard Medical School, in the Division of Pharmacoepidemiology and Pharmacoeconomics at Brigham and Women's Hospital, in Boston, Massachusetts
| | | | - Jerry Avorn
- Jerry Avorn is a professor of medicine at Harvard Medical School and chief of the Division of Pharmacoepidemiology and Pharmacoeconomics at Brigham and Women's Hospital
| |
Collapse
|
34
|
Li Y, Li X, Qu Y, Huang J, Zhu T, Zhao F, Li S, Mu D. Role of HMGB1 translocation to neuronal nucleus in rat model with septic brain injury. Neurosci Lett 2016; 645:90-96. [PMID: 27889435 DOI: 10.1016/j.neulet.2016.11.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/13/2022]
Abstract
High-mobility Group Box-1 (HMGB1) is a central late proinflammatory cytokine that triggers the inflammatory response during sepsis. However, whether HMGB1 is involved in the pathogenesis of septic brain damage is unknown. In this study, we investigated the role of HMGB1 in regulating brain injury in a rat model of sepsis. Wistar rats were subjected to cecal ligation and puncture (CLP) to induce septic brain injury. Hematoxylin and eosin staining was used to detect pathological changes in the cortex. The cellular localization of HMGB1 was determined using immunostaining. Cortical levels of HMGB1, its receptor for advanced glycation end-products (RAGE), and downstream effecter, nuclear factor kappa-B (NF-κB) subunit p65, were detected via western blot.HMGB1was increased in the cytoplasm via translocation from the nucleus predominantly in neurons. Moreover, RAGE and NF-κB p65 were upregulated after septic brain injury. Ethyl pyruvate, an inhibitor of HMGB1, down-regulated the expression of RAGE and NF-κB p65via inhibiting HMGB1 expression in the cytoplasm. Collectively, our findings suggest that HMGB1 and its signaling transduction have critical roles in the pathogenesis of septic brain injury. HMGB1 inhibition might be a potential new therapeutic target for septic brain injury.
Collapse
Affiliation(s)
- Yafei Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China
| | - Xihong Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China.
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China
| | - Jichong Huang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China
| | - Tingting Zhu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China
| | - Fengyan Zhao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China
| | - Shiping Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041,China.
| |
Collapse
|
35
|
Sun X, Liang J, Yao X, Lu C, Zhong T, Hong X, Wang X, Xu W, Gu M, Tang J. The activation of EGFR promotes myocardial tumor necrosis factor-α production and cardiac failure in endotoxemia. Oncotarget 2016; 6:35478-95. [PMID: 26486084 PMCID: PMC4742119 DOI: 10.18632/oncotarget.6071] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/28/2015] [Indexed: 12/29/2022] Open
Abstract
To study the effect of EGFR activation on the generation of TNF-α and the occurrence of cardiac dysfuncetion during sepsis, PD168393 and erlotinib (both are EGFR inhibitors) were applied to decreased the production of TNF-α and phosphrylation of ERK1/2 and p38 induced by LPS in cardiomyocytes. These results were further proved by specifically knocked down the expression of EGFR in vitro. Both TAPI-1, a TNF-α converting enzyme (TACE) inhibitor, and TGF-α neutralizing antibody could inhibit the activation of EGFR and the generation of TNF-α mRNA after LPS treatment. The increase of TGF-α in response to LPS could also be suppressed by TAPI-1. On the other hand, exogenous TGF-α increased the expression of TNF-α mRNA and partially reversed the inhibitory effect of TAPI-1 on expression of TNF-α mRNA in response to LPS indicating that the transactivation of EGFR by LPS in cardiomyocytes needs the help of TACE and TGF-α. In endotoxemic mice, inhibition the activation of EGFR not only decreased TNF-α production in the myocardium but also improved left ventricular pump function and ameliorated cardiac dysfunction and ultimately improved survival rate. All these results provided a new insight of how EGFR regulation the production of TNF-α in cardiomyocytes and a potential new target for the treatment of cardiac dysfunction in sepsis.
Collapse
Affiliation(s)
- Xuegang Sun
- The Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiani Liang
- The Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xueqing Yao
- The Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Science, Guangzhou, Guangdong, China
| | - Chunhua Lu
- The Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Tianyu Zhong
- The Department of Laboratory Medicine, First Affiliated Hospital of Ganna Medical University, Ganzhou, Jiangxi, China
| | - Xiaoyang Hong
- The Department of Intensive Care Unit, BaYi Children's Hospital, Beijing Military General Hospital, Beijing, China
| | - Xiaofei Wang
- The Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenjuan Xu
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Miaoning Gu
- The Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Tang
- The Department of Anesthesia, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
36
|
Abstract
For more than two decades, sepsis was defined as a microbial infection that produces fever (or hypothermia), tachycardia, tachypnoea and blood leukocyte changes. Sepsis is now increasingly being considered a dysregulated systemic inflammatory and immune response to microbial invasion that produces organ injury for which mortality rates are declining to 15-25%. Septic shock remains defined as sepsis with hyperlactataemia and concurrent hypotension requiring vasopressor therapy, with in-hospital mortality rates approaching 30-50%. With earlier recognition and more compliance to best practices, sepsis has become less of an immediate life-threatening disorder and more of a long-term chronic critical illness, often associated with prolonged inflammation, immune suppression, organ injury and lean tissue wasting. Furthermore, patients who survive sepsis have continuing risk of mortality after discharge, as well as long-term cognitive and functional deficits. Earlier recognition and improved implementation of best practices have reduced in-hospital mortality, but results from the use of immunomodulatory agents to date have been disappointing. Similarly, no biomarker can definitely diagnose sepsis or predict its clinical outcome. Because of its complexity, improvements in sepsis outcomes are likely to continue to be slow and incremental.
Collapse
Affiliation(s)
- Richard S Hotchkiss
- Department of Anesthesiology, Washington University of St. Louis, St. Louis, Missouri, USA
| | - Lyle L Moldawer
- Department of Surgery, University of Florida College of Medicine, Shands Hospital, Room 6116, 1600 SW Archer Road, Gainesville, Florida 32610-0019, USA
| | - Steven M Opal
- Department of Infectious Diseases and Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Konrad Reinhart
- Department of Anesthesiology and Intensive Care, Jena University Hospital, Jena, Germany
| | - Isaiah R Turnbull
- Department of Anesthesiology, Washington University of St. Louis, St. Louis, Missouri, USA
| | - Jean-Louis Vincent
- Department of Intensive Care, Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
37
|
Survival benefits of remote ischemic conditioning in sepsis. J Surg Res 2016; 213:131-137. [PMID: 28601305 DOI: 10.1016/j.jss.2016.01.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/20/2016] [Accepted: 01/27/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Sepsis remains the leading cause of death in the surgical intensive care unit. Prior studies have demonstrated a survival benefit of remote ischemic conditioning (RIC) in many disease states. The aim of this study was to determine the effects of RIC on survival in sepsis in an animal model and to assess alterations in inflammatory biochemical profiles. We hypothesized that RIC alters inflammatory biochemical profiles resulting in decreased mortality in a septic mouse model. MATERIALS AND METHODS Eight to 12 week C57BL/6 mice received intra-peritoneal injection of 12.5-mg/kg lipopolysaccharide (LPS). Septic animals in the experimental group underwent RIC at 0, 2, and 6 h after LPS by surgical exploration and alternate clamping of the femoral artery. Six 4-min cycles of ischemia-reperfusion were performed. Primary outcome was survival at 5-d after LPS injection. Secondary outcome was to assess the following serum cytokine levels: interferon-γ (IFN-γ), interleukin (IL)-10, IL-1β, and tumor necrosis factoralpha (TNFα) at the baseline before LPS injection, 0 hour after LPS injection, and at 2, 4, 24 hours after induction of sepsis (RIC was performed at 2 h after LPS injection). Kaplan-Meier survival analysis and log-rank test were used. ANOVA test was used to compare cytokine measurements. RESULTS We performed experiments on 44 mice: 14 sham and 30 RIC mice (10 at each time point). Overall survival was higher in the experimental group compared to the sham group (57% versus 21%; P = 0.02), with the highest survival rate observed in the 2-hour post-RIC group (70%). On Kaplan-Meier analysis, 2-h post-RIC group had increased survival at 5 days after LPS (P = 0.04) with hazard ratio of 0.3 (95% confidence interval = 0.09-0.98). In the RIC group, serum concentrations of IFN-γ, IL-10, IL-1β, and TNFα peaked at 2 h after LPS and then decreased significantly over 24 hours (P < 0.0001) compared to the baseline. CONCLUSIONS RIC improves survival in sepsis and has the potential for implementation in the clinical practice. Early implementation of RIC may play an immune-modulatory role in sepsis. Further studies are necessary to refine understanding of the observed survival benefits and its implications in sepsis management.
Collapse
|
38
|
Indenes and tetralenes analogues attenuates lipopolysaccharide-induced inflammation: An in-vitro and in-vivo study. Chem Biol Interact 2016; 245:12-9. [DOI: 10.1016/j.cbi.2015.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/27/2015] [Accepted: 12/11/2015] [Indexed: 02/06/2023]
|
39
|
Saxena A, Yadav D, Maurya AK, Kumar A, Mohanty S, Gupta MM, Lingaraju MC, Yatoo MI, Thakur US, Bawankule DU. Diarylheptanoids from Alnus nepalensis attenuates LPS-induced inflammation in macrophages and endotoxic shock in mice. Int Immunopharmacol 2015; 30:129-136. [PMID: 26679675 DOI: 10.1016/j.intimp.2015.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 11/06/2015] [Accepted: 12/01/2015] [Indexed: 12/15/2022]
Abstract
Diarylheptanoids, a group of plant secondary metabolites are increasingly recognized as potential therapeutic agents. The aim of study was to ascertain the anti-inflammatory profile of diarylheptanoids from Alnus nepalensis against lipopolysaccharide (LPS)-induced inflammation in macrophages and endotoxic shock in mice. Extracts prepared from dried leaves of A. nepalensis using standard solvents were tested against LPS-induced inflammation in macrophages. Among all, butanol extract (ANB) has shown most significant inhibition of pro-inflammatory cytokines without any cytotoxicity. HPLC analysis of ANB showed the presence of diarylheptanoids. The diarylheptanoids were further isolated and tested in-vitro for anti-inflammatory activity. Treatment of isolated diarylheptanoids (HOG, ORE and PLS) was able to reduce the production and mRNA level of pro-inflammatory cytokines (TNF-α and IL-6). Furthermore, we demonstrated that it inhibited the expression of NF-kB protein in LPS-induced inflammation in macrophages. In-vivo efficacy and safety profile of ANB revealed that oral treatment of ANB was able to improve the survival rate, and inhibited the production of pro-inflammatory cytokines in serum, attenuated vital organ injury in a dose dependent manner without any toxic effect at higher dose in mice. The results suggest that diarylheptanoids from A. nepalensis can be considered as potential therapeutic candidates for the management of inflammation related diseases.
Collapse
Affiliation(s)
- Archana Saxena
- Molecular Bioprospection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Deepti Yadav
- Analytical Chemistry Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Anil K Maurya
- Molecular Bioprospection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Anant Kumar
- Molecular Bioprospection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Shilpa Mohanty
- Molecular Bioprospection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Madan M Gupta
- Analytical Chemistry Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Madhu C Lingaraju
- Department of Pharmacology, Indian Veterinary Research Institute, Izatnagar 243122, India
| | - M I Yatoo
- Department of Veterinary Medicine, Indian Veterinary Research Institute, Izatnagar 243122, India
| | - Uttam S Thakur
- Department of Pharmacology, Indian Veterinary Research Institute, Izatnagar 243122, India
| | - Dnyaneshwar U Bawankule
- Molecular Bioprospection Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India.
| |
Collapse
|
40
|
Chiewchengchol D, Wright HL, Thomas HB, Lam CW, Roberts KJ, Hirankarn N, Beresford MW, Moots RJ, Edwards SW. Differential changes in gene expression in human neutrophils following TNF-α stimulation: Up-regulation of anti-apoptotic proteins and down-regulation of proteins involved in death receptor signaling. IMMUNITY INFLAMMATION AND DISEASE 2015; 4:35-44. [PMID: 27042300 PMCID: PMC4768069 DOI: 10.1002/iid3.90] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/30/2015] [Accepted: 11/04/2015] [Indexed: 01/14/2023]
Abstract
Responses of human neutrophils to TNF‐α are complex and multifactorial. Exposure of human neutrophils to TNF‐α in vitro primes the respiratory burst, delays apoptosis and induces the expression of several genes including chemokines, and TNF‐α itself. This study aimed to determine the impact of TNF‐α exposure on the expression of neutrophil genes and proteins that regulate apoptosis. Quantitative PCR and RNA‐Seq, identified changes in expression of several apoptosis regulating genes in response to TNF‐α exposure. Up‐regulated genes included TNF‐α itself, and several anti‐apoptotic genes, including BCL2A1, CFLAR (cFLIP) and TNFAIP3, whose mRNA levels increased above control values by between 4‐20 fold (n = 3, P < 0.05). In contrast, the expression of pro‐apoptotic genes, including CASP8, FADD and TNFRSF1A and TNFRSF1B, were significantly down‐regulated following TNF‐α treatment. These changes in mRNA levels were paralleled by decreases in protein levels of caspases 8 and 10, TRADD, FADD, TNFRSF1A and TNFRSF1B, and increased cFLIP protein levels, as detected by western blotting. These data indicate that when neutrophils are triggered by TNF‐α exposure, they undergo molecular changes in transcriptional expression to up‐regulate expression of specific anti‐apoptotic proteins and concomitantly decrease expression of specific proteins involved in death receptor signaling which will alter their function in TNF‐α rich environments.
Collapse
Affiliation(s)
- Direkrit Chiewchengchol
- Institutes of Integrative BiologyUniversity of LiverpoolLiverpoolUnited Kingdom; Immunology Unit & Center of Excellence in Immunology and Immune-mediated DiseaseDepartment of Microbiology, Faculty of Medicine, Chulalongkorn UniversityBangkokThailand
| | - Helen L Wright
- Institutes of Integrative Biology University of Liverpool Liverpool United Kingdom
| | - Huw B Thomas
- Institutes of Integrative Biology University of Liverpool Liverpool United Kingdom
| | - Connie W Lam
- Institutes of Integrative Biology University of Liverpool Liverpool United Kingdom
| | - Kate J Roberts
- Institutes of Integrative Biology University of Liverpool Liverpool United Kingdom
| | - Nattiya Hirankarn
- Immunology Unit & Center of Excellence in Immunology and Immune-mediated Disease Department of Microbiology, Faculty of Medicine, Chulalongkorn University Bangkok Thailand
| | | | - Robert J Moots
- Ageing and Chronic Disease University of Liverpool Liverpool United Kingdom
| | - Steven W Edwards
- Institutes of Integrative Biology University of Liverpool Liverpool United Kingdom
| |
Collapse
|
41
|
Potla R, Singh IS, Atamas SP, Hasday JD. Shifts in temperature within the physiologic range modify strand-specific expression of select human microRNAs. RNA (NEW YORK, N.Y.) 2015; 21:1261-1273. [PMID: 26018549 PMCID: PMC4478345 DOI: 10.1261/rna.049122.114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/15/2015] [Indexed: 06/04/2023]
Abstract
Previous studies have revealed that clinically relevant changes in temperature modify clinically relevant gene expression profiles through transcriptional regulation. Temperature dependence of post-transcriptional regulation, specifically, through expression of miRNAs has been less studied. We comprehensively analyzed the effect of 24 h exposure to 32°C or 39.5°C on miRNA expression profile in primary cultured human small airway epithelial cells (hSAECs) and its impact on expression of a targeted protein, protein kinase C α (PKCα). Using microarray, and solution hybridization-based nCounter assays, with confirmation by quantitative RT-PCR, we found significant temperature-dependent changes in expression level of only five mature human miRNAs, representing only 1% of detected miRNAs. Four of these five miRNAs are the less abundant passenger (star) strands. They exhibited a similar pattern of increased expression at 32°C and reduced expression at 39.5°C relative to 37°C. As PKCα mRNA has multiple potential binding sites for three of these miRNAs, we analyzed PKCα protein expression in HEK 293T cells and hSAECs. PKCα protein levels were lowest at 32°C and highest at 39.5°C and specific miRNA inhibitors reduced these effects. Finally, we analyzed cell-cycle progression in hSAECs and found 32°C cells exhibited the greatest G1 to S transition, a process known to be inhibited by PKCα, and the effect was mitigated by specific miRNA inhibitors. These results demonstrate that exposure to clinically relevant hypothermia or hyperthermia modifies expression of a narrow subset of miRNAs and impacts expression of at least one signaling protein involved in multiple important cellular processes.
Collapse
Affiliation(s)
- Ratnakar Potla
- Pulmonary and Critical Care Medicine Division, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Ishwar S Singh
- Pulmonary and Critical Care Medicine Division, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA Medicine and Research Services, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA
| | - Sergei P Atamas
- Medicine and Research Services, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA
| | - Jeffrey D Hasday
- Pulmonary and Critical Care Medicine Division, University of Maryland School of Medicine, Baltimore, Maryland 21201, USA Medicine and Research Services, Baltimore VA Medical Center, Baltimore, Maryland 21201, USA
| |
Collapse
|
42
|
Falasca L, Agrati C, Petrosillo N, Di Caro A, Capobianchi MR, Ippolito G, Piacentini M. Molecular mechanisms of Ebola virus pathogenesis: focus on cell death. Cell Death Differ 2015; 22:1250-9. [PMID: 26024394 PMCID: PMC4495366 DOI: 10.1038/cdd.2015.67] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 12/28/2022] Open
Abstract
Ebola virus (EBOV) belongs to the Filoviridae family and is responsible for a severe disease characterized by the sudden onset of fever and malaise accompanied by other non-specific signs and symptoms; in 30–50% of cases hemorrhagic symptoms are present. Multiorgan dysfunction occurs in severe forms with a mortality up to 90%. The EBOV first attacks macrophages and dendritic immune cells. The innate immune reaction is characterized by a cytokine storm, with secretion of numerous pro-inflammatory cytokines, which induces a huge number of contradictory signals and hurts the immune cells, as well as other tissues. Other highly pathogenic viruses also trigger cytokine storms, but Filoviruses are thought to be particularly lethal because they affect a wide array of tissues. In addition to the immune system, EBOV attacks the spleen and kidneys, where it kills cells that help the body to regulate its fluid and chemical balance and that make proteins that help the blood to clot. In addition, EBOV causes liver, lungs and kidneys to shut down their functions and the blood vessels to leak fluid into surrounding tissues. In this review, we analyze the molecular mechanisms at the basis of Ebola pathogenesis with a particular focus on the cell death pathways induced by the virus. We also discuss how the treatment of the infection can benefit from the recent experience of blocking/modulating cell death in human degenerative diseases.
Collapse
Affiliation(s)
- L Falasca
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - C Agrati
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - N Petrosillo
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - A Di Caro
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - M R Capobianchi
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - G Ippolito
- National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy
| | - M Piacentini
- 1] National Institute for Infectious Diseases, Lazzaro Spallanzani, Rome, Italy [2] Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
43
|
Ren H, Shao X, Zeng L, Wang F, Huang DN, Hou G. Matrix metalloproteinase-mediation of tumor targeting human recombinant tumor necrosis factor-α fusion protein. Mol Med Rep 2015; 12:2035-42. [PMID: 25891416 DOI: 10.3892/mmr.2015.3639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/20/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to use genetic engineering in order to establish an efficient tumor necrosis factor (TNF)-α fusion protein with low toxicity, which may be used to target tumors. Four types of matrix metalloproteinase (MMP)-mediated tumor targeting human recombinant TNF-α (rhTNF-α) fusion protein vectors were constructed. These were subsequently introduced into Escherichia coli. rhTNF-α fusion protein with a glutathione S-transferase (GST)-tag was purified using GST resin affinity chromatography, and GST-tags were digested using factor Xa. The cytotoxic effects of the fusion protein on L929 cells were determined using MTT assays. At a concentration of 1 pM, the GST-tagged fusion protein exerted no cytotoxic effects on the cells, compared with the negative control cells (P=0.975>0.05). However, at a concentration of 1000 pM, the deblocking fusion protein exerted greater cytotoxic effects on L929 cells, compared with positive control cells (P<0.05). Treatment with the fusion protein also induced cell apoptosis in the nasopharyngeal cancer cell line, CNE-2Z, which secretes high levels of MMP-1. In conclusion, the results of the present study suggested that MMP-mediated rhTNF-α fusion protein induces CNE-2Z cells apoptosis. rhTNF-α exhibits high efficacy and tumor cell targeting capability, with low toxicity effects on healthy cells.
Collapse
Affiliation(s)
- Hui Ren
- Department of Basic Medicine, Guangdong Medical College, Zhanjiang, Guangdong 524023, P.R. China
| | - Xin Shao
- Department of Basic Medicine, Guangdong Medical College, Zhanjiang, Guangdong 524023, P.R. China
| | - Liang Zeng
- Department of Basic Medicine, Guangdong Medical College, Zhanjiang, Guangdong 524023, P.R. China
| | - Fa Wang
- Department of Basic Medicine, Guangdong Medical College, Zhanjiang, Guangdong 524023, P.R. China
| | - Di-Nan Huang
- Department of Clinical Biochemistry, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| | - Gan Hou
- Department of Clinical Biochemistry, Guangdong Medical College, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
44
|
Loosbroock C, Hunter KW. Inhibiting TNF-α signaling does not attenuate induction of endotoxin tolerance. J Inflamm Res 2014; 7:159-67. [PMID: 25506235 PMCID: PMC4259568 DOI: 10.2147/jir.s75037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) is a central mediator of inflammatory responses elicited by Toll-like receptor agonists, such as the Gram-negative bacterial outer membrane antigen lipopolysaccharide (LPS). TNF-α is responsible for altering vascular permeability and activating infiltrating inflammatory cells, such as monocytes and neutrophils. Interestingly, TNF-α has also demonstrated the ability to induce tolerance to subsequent challenges with TNF-α or LPS in monocyte and macrophage cell populations. Tolerance is characterized by the inability to mount a typical inflammatory response during subsequent challenges following the initial exposure to an inflammatory mediator such as LPS. The ability of TNF-α to induce a tolerant-like state with regard to LPS is most likely a regulatory mechanism to prevent excessive inflammation. We hypothesized that the induction of tolerance or the degree of tolerance is dependent upon the production of TNF-α during the primary response to LPS. To investigate TNF-α-dependent tolerance, human monocytic THP-1 cells were treated with TNF-α-neutralizing antibodies or antagonistic TNF-α receptor antibodies before primary LPS stimulation and then monitored for the production of TNF-α during the primary and challenge stimulation. During the primary stimulation, anti-TNF-α treatment effectively attenuated the production of TNF-α and interleukin-1β; however, this reduced production did not impact the induction of endotoxin tolerance. These results demonstrate that interfering with TNF-α signaling attenuates production of inflammatory cytokines without affecting the induction of tolerance.
Collapse
Affiliation(s)
- Christopher Loosbroock
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, USA
| | - Kenneth W Hunter
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
45
|
Singh M, Hamid AA, Maurya AK, Prakash O, Khan F, Kumar A, Aiyelaagbe OO, Negi AS, Bawankule DU. Synthesis of diosgenin analogues as potential anti-inflammatory agents. J Steroid Biochem Mol Biol 2014; 143:323-33. [PMID: 24816230 DOI: 10.1016/j.jsbmb.2014.04.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 04/02/2014] [Accepted: 04/05/2014] [Indexed: 12/22/2022]
Abstract
We herein report the synthesis of diosgenin analogues from commercially available diosgenin as the starting material. The structures of newly synthesised compounds were confirmed by (1)H NMR, (13)C NMR and mass spectrometry. All analogues were evaluated for in-vitro anti-inflammatory profile against LPS-induced inflammation in primary peritoneal macrophages isolated from mice by quantification of pro-inflammatory (TNF-α, IL-6 and IL-1β) cytokines in cell culture supernatant using the ELISA technique followed by in-vitro cytotoxicity study. Among the synthesised analogues, analogue 15 [(E) 26-(3',4',5'-trimethoxybenzylidene)-furost-5en-3β-acetate)] showed significant anti-inflammatory activity by inhibiting LPS-induced pro-inflammatory cytokines in a dose-dependent manner without any cytotoxicity. Efficacy and safety of analogue 15 were further validated in an in-vivo system using LPS-induced sepsis model and acute oral toxicity in mice. Oral administration of analogue 15 inhibited the pro-inflammatory cytokines in serum, attenuated the liver and lung injury and reduced the mortality rate in sepsis mice. Acute oral toxicity study showed that analogue 15 is non-toxic at higher dose in BALB/c mice. Molecular docking study revealed the strong binding affinity of diosgenin analogues to the active site of the pro-inflammatory proteins. These findings suggested that analogue 15 may be a useful therapeutic candidate for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Monika Singh
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - A A Hamid
- Medicinal Chemistry Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India; Department of Chemistry, University of Ilorin, Ilorin, Nigeria
| | - Anil K Maurya
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Om Prakash
- Molecular and Structural Biology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Feroz Khan
- Molecular and Structural Biology Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - Anant Kumar
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India
| | - O O Aiyelaagbe
- Organic Chemistry Unit, Department of Chemistry, University of Ibadan, Ibadan, Nigeria
| | - Arvind S Negi
- Medicinal Chemistry Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India.
| | - Dnyaneshwar U Bawankule
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow 226015, India.
| |
Collapse
|
46
|
Intestine-specific deletion of microsomal triglyceride transfer protein increases mortality in aged mice. PLoS One 2014; 9:e101828. [PMID: 25010671 PMCID: PMC4092051 DOI: 10.1371/journal.pone.0101828] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/11/2014] [Indexed: 12/16/2022] Open
Abstract
Background Mice with conditional, intestine-specific deletion of microsomal triglyceride transfer protein (Mttp-IKO) exhibit a complete block in chylomicron assembly together with lipid malabsorption. Young (8–10 week) Mttp-IKO mice have improved survival when subjected to a murine model of Pseudomonas aeruginosa-induced sepsis. However, 80% of deaths in sepsis occur in patients over age 65. The purpose of this study was to determine whether age impacts outcome in Mttp-IKO mice subjected to sepsis. Methods Aged (20–24 months) Mttp-IKO mice and WT mice underwent intratracheal injection with P. aeruginosa. Mice were either sacrificed 24 hours post-operatively for mechanistic studies or followed seven days for survival. Results In contrast to young septic Mttp-IKO mice, aged septic Mttp-IKO mice had a significantly higher mortality than aged septic WT mice (80% vs. 39%, p = 0.005). Aged septic Mttp-IKO mice exhibited increased gut epithelial apoptosis, increased jejunal Bax/Bcl-2 and Bax/Bcl-XL ratios yet simultaneously demonstrated increased crypt proliferation and villus length. Aged septic Mttp-IKO mice also manifested increased pulmonary myeloperoxidase levels, suggesting increased neutrophil infiltration, as well as decreased systemic TNFα compared to aged septic WT mice. Conclusions Blocking intestinal chylomicron secretion alters mortality following sepsis in an age-dependent manner. Increases in gut apoptosis and pulmonary neutrophil infiltration, and decreased systemic TNFα represent potential mechanisms for why intestine-specific Mttp deletion is beneficial in young septic mice but harmful in aged mice as each of these parameters are altered differently in young and aged septic WT and Mttp-IKO mice.
Collapse
|
47
|
Pedrazza L, Lunardelli A, Luft C, Cruz CU, de Mesquita FC, Bitencourt S, Nunes FB, de Oliveira JR. Mesenchymal stem cells decrease splenocytes apoptosis in a sepsis experimental model. Inflamm Res 2014; 63:719-28. [PMID: 24888322 DOI: 10.1007/s00011-014-0745-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 05/15/2014] [Accepted: 05/19/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE AND DESIGN Mesenchymal stem cells (MSCs) are potent modulators of immune responses. Sepsis is the association of a systemic inflammatory response with an infection. The aim of this study was to test the ability of MSCs derived from adipose tissue, which have immunomodulatory effects, and to inhibit the septic process in an experimental model of mice. METHODS Three experimental groups (male C57BL/6 mice) were formed for the test: control group, untreated septic group and septic group treated with MSCs (1 × 10(6) cells/animal). RESULTS In the control group, there were no deaths; in the untreated septic group, the mortality rate was 100 % within 26 h; in the septic group treated with MSCs, the mortality rate reached 40 % within 26 h. The group treated with MSCs was able to reduce the markers of tissue damage in the liver and pancreas. The treated group had a reduction of inflammatory markers. Furthermore, the MSCs-treated group was able to inhibit the increase of apoptosis in splenocytes observed in the untreated septic group. CONCLUSIONS Our data showed that MSCs ameliorated the immune response with decrease of inflammatory cytokines and increase anti-inflammatory IL-10; moreover, inhibited splenocytes apoptosis and, consequently, inhibited tissue damage during sepsis.
Collapse
Affiliation(s)
- Leonardo Pedrazza
- Laboratório de Pesquisa em Biofísica Celular e Inflamação, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga 6681, prédio 12, bloco C, sala 221, Porto Alegre, Rio Grande do Sul, CEP 90619-900, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Evaluating the efficacy and safety of two doses of the polyclonal anti-tumor necrosis factor-α fragment antibody AZD9773 in adult patients with severe sepsis and/or septic shock: randomized, double-blind, placebo-controlled phase IIb study*. Crit Care Med 2014; 42:504-11. [PMID: 24335445 DOI: 10.1097/ccm.0000000000000043] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This trial compared the efficacy/safety of two IV doses of AZD9773, a polyclonal antibody to tumor necrosis factor-α, in adult patients with severe sepsis/septic shock. DESIGN Multicenter, randomized, double-blind, placebo-controlled phase IIb trial. SETTING ICUs in seven countries (Australia, Belgium, Canada, Czech Republic, Finland, France, and Spain). PATIENTS Patients 18 years old or older with severe sepsis and/or septic shock. Patients were required to have 1) objective clinical evidence of infection; 2) at least two of four systemic inflammatory response syndrome criteria; and 3) cardiovascular and/or respiratory sepsis-related failure. INTERVENTIONS Patients were randomized 1:1:1 to a single loading infusion of AZD9773 250 U/kg followed by 50 U/kg every 12 hours (low dose, n = 100), a single loading infusion of AZD9773 500 U/kg followed by 100 U/kg every 12 hours (high dose, n = 100), or placebo (n = 100) for 5 days. Follow-up assessments were performed up to day 90. MEASUREMENTS AND MAIN RESULTS Mean number of ventilator-free days (primary endpoint) did not differ between low-dose (19.7 d) or high-dose AZD9773 (17.3 d) and placebo (18.3 d) (one-sided p = 0.18 and 0.74, respectively). Mortality rates were comparable across treatment groups; relative risk of death versus placebo at day 29 was 0.80 for low-dose AZD9773 (one-sided p = 0.25) and 1.64 for high-dose AZD9773 (p = 0.97). Most patients experienced at least one treatment-emergent adverse event (87.8% in AZD9773-treated patients, 92.9% in placebo patients) although most were mild/moderate in nature. No differences in the incidence of adverse events or laboratory or vital sign abnormalities were observed between groups. CONCLUSIONS AZD9773 rapidly and efficiently decreased plasma tumor necrosis factor-α concentration in patients with severe sepsis/septic shock, but this effect did not translate into clinical benefit.
Collapse
|
49
|
Abstract
Sepsis remains a leading cause of death in critically ill patients, despite efforts to improve patient outcome. Thus far, no magic drugs exist for severe sepsis and septic shock. Instead, early diagnosis and prompt initial management such as early goal-directed therapy are key to improve sepsis outcome. For early detection of sepsis, biological markers (biomarkers) can help clinicians to distinguish infection from host response to inflammation. Ideally, biomarkers can be used for risk stratification, diagnosis, monitoring of treatment responses, and outcome prediction. More than 170 biomarkers have been identified as useful for evaluating sepsis, including C-reactive protein, procalcitonin, various cytokines, and cell surface markers. Recently, studies have reported on the usefulness of biomarker-guided antibiotic stewardships. However, the other side of these numerous biomarkers is that no novel single laboratory marker can diagnose, predict, and track the treatment of sepsis. The purpose of this review is to summarize several key biomarkers from recent sepsis studies.
Collapse
Affiliation(s)
- Sung-Yeon Cho
- Division of Infectious Diseases, Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea. ; Vaccine Bio Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Jung-Hyun Choi
- Division of Infectious Diseases, Department of Internal Medicine, The Catholic University of Korea College of Medicine, Seoul, Korea. ; Vaccine Bio Research Institute, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
50
|
Carmona F, Manso PH, Silveira VS, Cunha FQ, de Castro M, Carlotti APCP. Inflammation, myocardial dysfunction, and mortality in children with septic shock: an observational study. Pediatr Cardiol 2014; 35:463-70. [PMID: 24091885 PMCID: PMC7100657 DOI: 10.1007/s00246-013-0801-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/14/2013] [Indexed: 12/11/2022]
Abstract
We aimed to investigate whether nuclear factor kappa-B activation, as evaluated by gene expression of its inhibitor (I-κBα) and cytokine serum levels, was associated with myocardial dysfunction and mortality in children with septic shock. Twenty children with septic shock were prospectively enrolled and grouped according to ejection fraction (EF) <45% (group 1) or EF ≥45% (group 2) on the first day after admission to the pediatric intensive care unit. No interventions were made. In the first day, patients from group 1 (n = 6) exhibited significantly greater tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-10 plasma levels. However, I-κBα gene expression was not different in both groups. Mortality and number of complications were significantly greater in group 1. Patients who died had greater plasma concentrations of TNF-α. In conclusion, TNF-α and IL-10 are involved in myocardial dysfunction accompanying septic shock in children, and TNF-α is associated with mortality.
Collapse
Affiliation(s)
- Fabio Carmona
- Ribeirao Preto Medical School, University of Sao Paulo, Avenida dos Bandeirantes 3900, Ribeirao Preto, SP, 14049-900, Brazil,
| | - Paulo H. Manso
- Ribeirao Preto Medical School, University of Sao Paulo, Avenida dos Bandeirantes 3900, Ribeirao Preto, SP 14049-900 Brazil
| | - Vanessa S. Silveira
- Ribeirao Preto Medical School, University of Sao Paulo, Avenida dos Bandeirantes 3900, Ribeirao Preto, SP 14049-900 Brazil
| | - Fernando Q. Cunha
- Ribeirao Preto Medical School, University of Sao Paulo, Avenida dos Bandeirantes 3900, Ribeirao Preto, SP 14049-900 Brazil
| | - Margaret de Castro
- Ribeirao Preto Medical School, University of Sao Paulo, Avenida dos Bandeirantes 3900, Ribeirao Preto, SP 14049-900 Brazil
| | - Ana P. C. P. Carlotti
- Ribeirao Preto Medical School, University of Sao Paulo, Avenida dos Bandeirantes 3900, Ribeirao Preto, SP 14049-900 Brazil
| |
Collapse
|