1
|
Zhang L, Zhao P, Liu Y, Shi N, Zhou Y, Peng S, Sun T, Zhang M, Wu Y, Yang X, Wen Y, Shi G, Gao X, Luo L. Detection of TNF-α using the established ab-MPs-CLIA. Talanta 2025; 285:127301. [PMID: 39637773 DOI: 10.1016/j.talanta.2024.127301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Tumor necrosis factor alpha (TNF-α) is a key cytokine in inflammation and immune responses, making its rapid and accurate detection essential for disease diagnosis and management. In this study, we developed a highly sensitive chemiluminescence immunoassay (CLIA) using antibody-coated magnetic particles (Ab-MPs-CLIA) for TNF-α detection. From nine candidate antibodies, we identified an optimal pair through epitope competition and affinity assessments, significantly improving assay performance. The Ab-MPs-CLIA achieved a detection limit of 0.25 pg/mL, 6.8 times more sensitive than Siemens commercial kits, with a broad linear range of 9.2-1077 pg/mL. The method demonstrated excellent stability, both under accelerated conditions at 37 °C for 7 days and long-term storage at 4 °C for 12 months. It showed no cross-reactivity with common interfering substances in human serum, ensuring high specificity. Notably, the entire process, from sample preparation to result, takes just 25 min, compared to 3-4 h for both ELISA and RIA, and CLIA typically offers 10-100 times higher sensitivity than these methods. These advantages make the Ab-MPs-CLIA an ideal option for clinical laboratories, providing superior sensitivity, specificity, broader dynamic range, and greater operational efficiency than existing TNF-α detection technologies.
Collapse
Affiliation(s)
- Liang Zhang
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212003, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Pinnan Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yujun Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Ning Shi
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China; Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yangyihua Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Shangde Peng
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212003, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Tieqiang Sun
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China; College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China
| | - Yahui Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China; Hunan Normal University School of Medicine, Changsha, Hunan Province, 410081, China
| | - Xuechen Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yan Wen
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212003, China.
| | - Gang Shi
- Kangpu Biotechnology (Beijing) Co., Ltd., Beijing, 100102, China.
| | - Xiang Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
2
|
Răcean MA, Săsăran MO, Mărginean CO, Cucerea M. Umbilical cord blood level of interleukins used as a predictor of early-onset neonatal sepsis: a comprehensive review. Front Cell Infect Microbiol 2025; 15:1518088. [PMID: 40171168 PMCID: PMC11959072 DOI: 10.3389/fcimb.2025.1518088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/19/2025] [Indexed: 04/03/2025] Open
Abstract
Neonatal sepsis (NS) is a major cause of morbidity and mortality in both preterm and term infants; early-onset NS (EONS) occurs in newborns within the first 72 h of life. Cytokines are messengers with low molecular weight that are produced by macrophages and lymphocytes in response to antigenic stimulations or products of inflammation. Different interleukins (IL) have higher values in EONS, when detected from peripheral venous blood. This review aims to analyze if the cytokines determined from the umbilical cord blood (UCB) of newborns may help in the rapid and accurate diagnosis of EONS in newborns originating from pregnancies with maternal-fetal infectious risk. Three databases, namely, PubMed, Scopus, and Web of Science, were searched for original research articles that assessed the relationship between interleukins and EONS. The search results retrieved a number of 18 articles that complied with the inclusion and exclusion criteria. Some studies report that neonates with EONS had higher umbilical plasma levels of cytokines such as IL-1ß, IL-6, IL-8, IL-10, IL-18, and IL-27. However, results are controversial, as many authors failed to establish the cut-off values of cytokines detected from UCB that may predict EONS. The main limitations of the current studies remain the small study samples, the heterogeneous population, and the lack of stratification of the studied population according to gestational age (GA). The cytokines that seem to be more accurate in the early diagnosis of EONS, as reported by the majority of the studies, are IL-6 and IL-8. The level of these cytokines may guide clinicians in the careful administration of antibiotics, thus aiding in the overall reduction of antimicrobial resistance.
Collapse
Affiliation(s)
- Maria Andreea Răcean
- Department of Pediatrics 4, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureş, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureş, Romania
| | - Maria Oana Săsăran
- Department of Pediatrics 3, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureş, Romania
| | - Cristina Oana Mărginean
- Department of Pediatrics 1, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureş, Romania
| | - Manuela Cucerea
- Department of Pediatrics 4, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Târgu Mureş, Romania
| |
Collapse
|
3
|
Paul R, Vyas D, Quinones Cardona V, Gilfillan M, Young M, Pough K, Carey AJ. Reduction of Overall Antibiotic Utilization Rate in a Level IV Neonatal Intensive Care Unit. Pediatrics 2025; 155:e2024066367. [PMID: 39999320 DOI: 10.1542/peds.2024-066367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/11/2024] [Indexed: 02/27/2025] Open
Abstract
OBJECTIVE Antibiotics are the most frequently prescribed pharmacologic agents in the neonatal intensive care unit (NICU). Antibiotic treatment for suspected or culture-negative sepsis surpasses that for culture-proven infection. Therefore, we sought to reduce our overall antibiotic utilization rate (AUR), defined by total antibiotic days per 1000 patient days (DOT/1000-PD), by 20% within a 4-year period (by December 2023). METHODS A multidisciplinary team was convened to develop an antibiotic stewardship quality improvement initiative in our 39-bed level IV NICU. Consensus guidelines for antibiotic duration for common indications were developed. Interventions included educational sessions, antibiotic stop dates, and antibiotic necessity documentation in the electronic health record to standardize provider justification for antibiotic prescription and duration. RESULTS A total of 552 infants were included in the analysis, 137 in the baseline and 415 in the postintervention period. Overall AUR decreased by 50% from 278 to 140 DOT/1000-PDs. AUR related to culture-negative sepsis diagnoses decreased by 64% from 22 to 8 DOT/1000-PDs. The percent of antibiotic therapy reinitiation within 2 weeks remained unchanged. CONCLUSION Implementation of NICU antibiotic consensus guidelines supported by evidence-based education on culture-negative sepsis diagnosis can effectively reduce antibiotic use in a safe manner, despite a heterogenous, high acuity, level IV NICU population. Multidisciplinary team support and standardization of antibiotic justification in the electronic health record can be coupled to reinforce compliance with established guidelines to promote long-lasting antibiotic reduction.
Collapse
Affiliation(s)
- Reema Paul
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Dipen Vyas
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
- Current affiliation: Department of Pediatrics, Division of Newborn Medicine, Children's of Mississippi, University of Mississippi Medical Center, Jackson, Mississippi
| | - Vilmaris Quinones Cardona
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Margaret Gilfillan
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Megan Young
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
- Department of Pharmacy, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Kimberly Pough
- Department of Pharmacy, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
- Current affiliation: Department of Quality and Safety Services, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Alison J Carey
- Department of Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
4
|
Qi S, Huang S, Qian R. Predictive Value of Combined Serum Nuclear Factor Erythroid 2-Related Factor 2 and Prognostic Nutritional Index for Sepsis after Percutaneous Nephrolithotomy. J Endourol 2025; 39:222-230. [PMID: 39964774 DOI: 10.1089/end.2024.0519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Objective: This prospective observational study aimed to explore the predictive value of serum nuclear factor erythroid 2-related factor 2 (Nrf2) levels and its combination with the prognostic nutritional index (PNI) for postoperative sepsis in patients undergoing percutaneous nephrolithotomy (PCNL) for renal stones. Methods: This prospective observational study included 112 patients with renal stones who underwent PCNL surgery in our hospital from January 2023 to December 2023. The diagnosis of sepsis was based on the third international consensus definitions for sepsis and septic shock (Sepsis-3). Clinical data on all patients were collected, and the PNI index was calculated. The serum levels of Nrf2, interleukin-6 (IL-6), interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and C-reactive protein (CRP) in all patients were measured using enzyme-linked immunosorbent assay. Data analysis was performed using Statistical Package for the Social Sciences (SPSS) 25.0 statistical software. Results: Compared with the nonsepsis patients, the sepsis patients had a higher proportion of positive preoperative urine cultures, longer activated partial thromboplastin time, and lower PNI scores. At 6, 12, and 24 hours after surgery, the sepsis patients had significantly lower serum Nrf2 levels and higher levels of IL-6, IL-1β, TNF-α, and CRP. Pearson's analysis showed a negative correlation between serum Nrf2 levels and IL-6 and CRP levels. The combination of PNI and serum Nrf2 levels at 24 hours after surgery had a higher predictive level, with an Area Under the Curve (AUC) of 0.844, a sensitivity of 75.3%, and a specificity of 77.8%. Finally, logistic regression analysis showed that decreased serum Nrf2 level at 24 hours after surgery was a risk factor for postoperative sepsis in renal stone patients. Conclusion: In conclusion, our study results indicate significantly lower serum Nrf2 levels in postoperative sepsis patients undergoing PCNL. Serum Nrf2 levels in combination with PNI can be used to predict the occurrence of postoperative sepsis in renal stone patients undergoing PCNL.
Collapse
Affiliation(s)
- Shengwei Qi
- Operating Room, The Third People's Hospital of Chengdu, Chengdu, P.R. China
| | - Shuangying Huang
- Operating Room, The Third People's Hospital of Chengdu, Chengdu, P.R. China
| | - Rong Qian
- Operating Room, The Third People's Hospital of Chengdu, Chengdu, P.R. China
| |
Collapse
|
5
|
Liu Z, Li F, Li N, Chen Y, Chen Z. MicroRNAs as regulators of cardiac dysfunction in sepsis: pathogenesis and diagnostic potential. Front Cardiovasc Med 2025; 12:1517323. [PMID: 40041174 PMCID: PMC11876399 DOI: 10.3389/fcvm.2025.1517323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Sepsis, a life-threatening condition arising from an uncontrolled immune response to infection, can lead to organ dysfunction, with severe inflammation potentially causing multiple organ failures. Sepsis-induced cardiac dysfunction (SIMD) is a common and severe complication of sepsis, significantly increasing patient mortality. Understanding the pathogenesis of SIMD is crucial for improving treatment, and microRNAs (miRNAs) have emerged as important regulators in this process. Methods A comprehensive literature search was conducted in PubMed, Science Direct, and Embase databases up to September 2024. The search terms included ["miRNA" or "microRNA"] and ["Cardiac" or "Heart"] and ["Sepsis" or "Septic"], with the language limited to English. After initial filtering by the database search engine, Excel software was used to further screen references. Duplicate articles, those without abstracts or full texts, and review/meta-analyses or non-English articles were excluded. Finally, 106 relevant research articles were included for data extraction and analysis. Results The pathogenesis of SIMD is complex and involves mitochondrial dysfunction, oxidative stress, cardiomyocyte apoptosis and pyroptosis, dysregulation of myocardial calcium homeostasis, myocardial inhibitory factors, autonomic nervous regulation disorders, hemodynamic changes, and myocardial structural alterations. miRNAs play diverse roles in SIMD. They are involved in regulating the above-mentioned pathological processes. Discussion Although significant progress has been made in understanding the role of miRNAs in SIMD, there are still challenges. Some studies on the pathogenesis of SIMD have limitations such as small sample sizes and failure to account for confounding factors. Research on miRNAs also faces issues like inconsistent measurement techniques and unclear miRNA-target gene relationships. Moreover, the translation of miRNA-based research into clinical applications is hindered by problems related to miRNA stability, delivery mechanisms, off-target effects, and long-term safety. In conclusion, miRNAs play a significant role in the pathogenesis of SIMD and have potential as diagnostic biomarkers. Further research is needed to overcome existing challenges and fully exploit the potential of miRNAs in the diagnosis and treatment of SIMD.
Collapse
Affiliation(s)
- Zhen Liu
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feiyang Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ningcen Li
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yong Chen
- Department of Critical Care Medicine, Tianjin Hospital of ITCWM Nankai Hospital, Tianjin, China
| | - Zelin Chen
- Research Center of Experimental Acupuncture Science, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Acupuncture-Moxibustion and Tuina, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
6
|
Wu HS, Huang HC, Chen IL. Analysis of Salivary Cytokines in Retinopathy of Prematurity. CHILDREN (BASEL, SWITZERLAND) 2025; 12:80. [PMID: 39857911 PMCID: PMC11764213 DOI: 10.3390/children12010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND/OBJECTIVES This cohort study aimed to establish a correlation between salivary cytokines and retinopathy of prematurity (ROP) in premature neonates. Additionally, we sought to identify a minimally invasive method for cytokine detection in this population. METHODS We recruited premature neonates born at less than 34 weeks gestational age (GA), with no history of maternal or neonatal infections. Salivary samples were collected on their first (D1) and seventh (D7) days of life, and cytokine levels were measured using the MILLPLEXMAP Human multiplex assay. RESULTS A total of 125 neonates were included in the study, categorized into two groups based on the severity of ROP: None to Mild and Moderate to Severe. The salivary levels of interleukin (IL)-6, IL-8, vascular endothelial growth factor (VEGF), and tumor necrosis factor (TNF)-α on D1 and D7 were significantly higher in the Moderate to Severe ROP group compared to the None to Mild ROP group (p = 0.005, 0.004, 0.026, 0.018, 0.001, 0.007, 0.025, and 0.002, respectively). After adjusting for GA, the levels of IL-6 and VEGF on D7 were significantly elevated in the Moderate to Severe ROP group compared to the None to Mild ROP group (p = 0.024 and 0.016, respectively). CONCLUSIONS This study establishes a novel, non-invasive method for the early prediction of ROP in premature neonates by correlating salivary cytokine levels in early life with the subsequent development of ROP.
Collapse
Affiliation(s)
- Hwa-Shiu Wu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan; (H.-S.W.); (H.-C.H.)
| | - Hsin-Chun Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan; (H.-S.W.); (H.-C.H.)
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Linkou 33302, Taiwan
| | - I-Lun Chen
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Kaohsiung 83301, Taiwan; (H.-S.W.); (H.-C.H.)
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Linkou 33302, Taiwan
| |
Collapse
|
7
|
Sepiashvili L, Brydon A, Koroshegyi C, Gold A, Dalvi P, Ghayoori S, Rahman M, Huang V, Maxwell C, Nguyen GC, Ito S. Reduction of tumor necrosis factor (TNF) in milk of women receiving anti-TNF antibody. Pediatr Res 2024:10.1038/s41390-024-03672-9. [PMID: 39487320 DOI: 10.1038/s41390-024-03672-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Tumor Necrosis Factor (TNF) are expressed in milk. Experimental data indicate enhanced brain growth and cognitive development in the mouse offspring given milk deficient in TNF and TNF-dependent chemokines. Although monoclonal antibodies against TNF (TNFmAb) are used during breastfeeding due to minimal milk excretion, whether it affects endogenous TNF levels in human milk is unclear. METHODS A prospective cohort study was conducted in 26 breastfeeding women with inflammatory bowel disease (IBD) and 31 non-IBD women. Milk TNF and related chemokines were measured at 5-6 weeks postpartum (early-lactation point) as a primary endpoint and further determined at 13-14 weeks postpartum. In a subset of their infants, neurocognitive development was assessed at 12 and 18 months of age using Bayley-III tool. RESULTS While milk TNF levels were not significantly different between the control and those with IBD, women with IBD receiving TNFmAb showed 70% lower median milk TNF than those without TNFmAb (P = 0.019) at early lactation period. TNF-dependent chemokines (MIP-1β and IP-10) also showed similar patterns. Neurocognitive development of the infants was not significantly different among the groups. CONCLUSIONS Women with IBD receiving TNFmAb show significantly lower milk TNF/chemokines at 5-6 weeks postpartum than those without TNFmAb. CLINICALTRIALS gov NCT03397108. IMPACT We found that milk concentrations of Tumor Necrosis Factor (TNF) and TNF-dependent chemokines (MIP-1β and IP-10) are low in women with inflammatory bowel disease who are receiving anti-TNF monoclonal antibody (TNFmAb), compared to those without concurrent anti TNF therapy. While maternal use of TNFmAb during breastfeeding is considered inconsequential to infant health due to their low levels of milk excretion, it affects profiles of endogenous cytokines in milk. Our findings provide a rationale to investigate human implications of the animal data in the literature that suggest enhancement of neurocognitive development of the offspring fed with milk deficient in TNF-dependent chemokines.
Collapse
Affiliation(s)
- Lusia Sepiashvili
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Avery Brydon
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | | | - Anna Gold
- Department of Psychology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Pooja Dalvi
- Division of Clinical Pharmacology and Toxicology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Sholeh Ghayoori
- Division of Clinical Pharmacology and Toxicology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mehzabin Rahman
- Division of Clinical Pharmacology and Toxicology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Vivian Huang
- Division of Gastroenterology, Mount Sinai Hospital, Toronto, ON, Canada
- Division of Gastroenterology and Hepatology, University of Toronto, Toronto, ON, Canada
| | - Cynthia Maxwell
- Department of Gynecology, Women's College Hospital, Toronto, ON, Canada
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Toronto, ON, Canada
| | - Geoffrey C Nguyen
- Division of Gastroenterology, Mount Sinai Hospital, Toronto, ON, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Shinya Ito
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.
- Division of Clinical Pharmacology and Toxicology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada.
- Translational Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
8
|
Neykhonji M, Asgharzadeh F, Farazestanian M, Al-Asady AM, Kaffashbashi M, Parizadeh SA, Attarian M, Nazari SE, Rahmani F, Eskandari M, Avan A, Hasanzadeh M, Ryzhikov M, Khazaei M, Hassanian SM. Oenothera biennis improves pregnancy outcomes by suppressing inflammation and fibrosis in an intra-uterine adhesion rat model. Sci Rep 2024; 14:22376. [PMID: 39333188 PMCID: PMC11437134 DOI: 10.1038/s41598-024-69488-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/05/2024] [Indexed: 09/29/2024] Open
Abstract
Intrauterine adhesion (IUA), also referred to as Asherman's syndrome, is characterized by fibrosis, inflammation, and can cause amenorrhea and infertility due to abnormal endometrial healing. Histological and Molecular methods were used to evaluate the efficacy of EPO, which is traditionally known for its anti-inflammatory and fibrinolytic properties, in preventing the formation of IUA. Oral administration of EPO reduced the formation of adhesion bands and promoted endometrial regeneration. EPO administration decreased extracellular matrix accumulation, evidenced by the down-regulation of tissue COL1A1 and COL3A1 expression. The anti-inflammatory effect of EPO was confirmed by a reduction in oxidants and down-regulation of pro-inflammatory cytokines including TNF-α, IL-6, IFN-γ, and IL-1β. Furthermore, EPO improved embryonic development parameters, including size and weight of embryo, as well as increased embryo count and live embryo percentage in the rat IUA model. EPO also positively enhanced implantation markers, particularly enlargement and mass gain in the placenta of the treated group, consequently improving pregnancy outcomes such as the number of babies, percent of live babies, baby weight and gestation time. Histopathological investigation provides evidence that oral administration of EPO showed no toxicity on the main three organs including liver, kidney and heart. These results showed that EPO can be considered as a safe and natural product with potent anti-inflammatory and fibrinolytic properties without any observed side effects for the treatment of IUA.
Collapse
Affiliation(s)
- Marzieh Neykhonji
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marjaneh Farazestanian
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdulridha Mohammed Al-Asady
- Department of Medical Sciences, Faculty of Nursing, University of Warith Al-Anbiyaa, Karbala, Iraq
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Karbala, Iraq
| | - Maziar Kaffashbashi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mahsa Attarian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Elnaz Nazari
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Kashmar School of Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Eskandari
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Human Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Hasanzadeh
- Department of Obstetrics and Gynecology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- School of Medicine, Saint Louis University, Saint Louis, MO, USA
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
9
|
Loaiza R, Fattahi F, Kalbitz M, Grailer JJ, Russell MW, Jalife J, Valdivia HH, Zetoune FS, Ward PA. The Impact of Extracellular Histones and Absence of Toll-like Receptors on Cardiac Functional and Electrical Disturbances in Mouse Hearts. Int J Mol Sci 2024; 25:8653. [PMID: 39201339 PMCID: PMC11354419 DOI: 10.3390/ijms25168653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
In polymicrobial sepsis, the extracellular histones, mainly released from activated neutrophils, significantly contribute to cardiac dysfunction (septic cardiomyopathy), as demonstrated in our previous studies using Echo-Doppler measurements. This study aims to elucidate the roles of extracellular histones and their interactions with Toll-like receptors (TLRs) in cardiac dysfunction. Through ex vivo assessments of ECG, left ventricle (LV) function parameters, and in vivo Echo-Doppler studies in mice perfused with extracellular histones, we aim to provide comprehensive insights into the mechanisms underlying sepsis-induced cardiac dysfunction. Langendorff-perfused hearts from both wild-type and TLR2, TLR3, or TLR4 knockout (KO) mice were examined. Paced mouse hearts were perfused with histones to assess contractility and relaxation. Echo-Doppler studies evaluated cardiac dysfunction after intravenous histone injection. Histone perfusion caused defects in contractility and relaxation, with TLR2 and TLR3 KO mice being partially protected. Specifically, TLR2 KO mice exhibited the greatest reduction in Echo-Doppler abnormalities, while TLR4 KO exacerbated cardiac dysfunction. Among individual histones, H1 induced the most pronounced abnormalities in cardiac function, apoptosis of cardiomyocytes, and LDH release. Our data highlight significant interactions between histones and TLRs, providing insights into histones especially H1 as potential therapeutic targets for septic cardiomyopathy. Further studies are needed to explore specific histone-TLR interactions and their mechanisms.
Collapse
Affiliation(s)
- Randall Loaiza
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA; (R.L.); (J.J.); (H.H.V.)
- CENIBiot Laboratory, The National Center of High Technology (CeNAT-CONARE), San José 10109, Costa Rica
| | - Fatemeh Fattahi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
| | - Miriam Kalbitz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
- Department of Orthopaedic Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital of Ulm, 89081 Ulm, Germany
- Military Medical City Hospital, Doha 486441, Qatar
| | - Jamison J. Grailer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
- Integrated Biology R&D, Bioassay Development, Promega Corporation, Madison, WI 53711, USA
| | - Mark W. Russell
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109, USA;
| | - Jose Jalife
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA; (R.L.); (J.J.); (H.H.V.)
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - Hector H. Valdivia
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, USA; (R.L.); (J.J.); (H.H.V.)
- Department of Medicine, Cardiovascular Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Firas S. Zetoune
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
| | - Peter A. Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (M.K.); (J.J.G.); (F.S.Z.)
| |
Collapse
|
10
|
Sahoo DK, Wong D, Patani A, Paital B, Yadav VK, Patel A, Jergens AE. Exploring the role of antioxidants in sepsis-associated oxidative stress: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1348713. [PMID: 38510969 PMCID: PMC10952105 DOI: 10.3389/fcimb.2024.1348713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Sepsis is a potentially fatal condition characterized by organ dysfunction caused by an imbalanced immune response to infection. Although an increased inflammatory response significantly contributes to the pathogenesis of sepsis, several molecular mechanisms underlying the progression of sepsis are associated with increased cellular reactive oxygen species (ROS) generation and exhausted antioxidant pathways. This review article provides a comprehensive overview of the involvement of ROS in the pathophysiology of sepsis and the potential application of antioxidants with antimicrobial properties as an adjunct to primary therapies (fluid and antibiotic therapies) against sepsis. This article delves into the advantages and disadvantages associated with the utilization of antioxidants in the therapeutic approach to sepsis, which has been explored in a variety of animal models and clinical trials. While the application of antioxidants has been suggested as a potential therapy to suppress the immune response in cases where an intensified inflammatory reaction occurs, the use of multiple antioxidant agents can be beneficial as they can act additively or synergistically on different pathways, thereby enhancing the antioxidant defense. Furthermore, the utilization of immunoadjuvant therapy, specifically in septic patients displaying immunosuppressive tendencies, represents a promising advancement in sepsis therapy.
Collapse
Affiliation(s)
- Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - David Wong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anil Patani
- Department of Biotechnology, Smt. S. S. Patel Nootan Science and Commerce College, Sankalchand Patel University, Gujarat, India
| | - Biswaranjan Paital
- Redox Regulation Laboratory, Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Virendra Kumar Yadav
- Department of Life Sciences, Hemchandracharya North Gujarat University, Gujarat, India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Gujarat, India
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
11
|
Chao Y, Huang W, Xu Z, Li P, Gu S. Effect of RUNX1/FOXP3 axis on apoptosis of T and B lymphocytes and immunosuppression in sepsis. Open Med (Wars) 2023; 18:20230728. [PMID: 37636994 PMCID: PMC10448307 DOI: 10.1515/med-2023-0728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 04/20/2023] [Accepted: 05/08/2023] [Indexed: 08/29/2023] Open
Abstract
Lymphocyte apoptosis is a latent factor for immunosuppression in sepsis. Forkhead box protein P3 (FOXP3) can interact with RUNX family transcription factor 1 (RUNX1) in regulatory T cells. Our research was to probe whether RUNX1/FOXP3 axis affects immunosuppression in the process of sepsis by modulating T and B lymphocyte apoptosis. We constructed sepsis model in mice and mouse CD4+ T and CD19+ B lymphocytes. RUNX1 and FOXP3 expressions and apoptosis in cells were assessed by western blot, quantitative real-time PCR, and flow cytometer. Inflammation of serum and pathological damage was assessed by ELISA and H&E staining. Relationship between RUNX1 and FOXP3 was assessed by co-immunoprecipitation. The findings showed that RUNX1 ameliorated the survival rate, pathological damage, and decreased inflammation-related factors, and inhibited apoptosis of CD4+ T and CD19+ B cells in cecal ligation and puncture mice. Furthermore, RUNX1 up-regulated the viability and down-regulated apoptotic rate with the changed expressions of apoptosis-related molecules in lipopolysaccharide (LPS)-mediated CD4+ T and CD19+ B cells. Additionally, FOXP3 interacted with RUNX1, and its silencing decreased RUNX1 expression and reversed the inhibitory effect of RUNX1 on apoptosis of LPS-mediated CD4+ T and CD19+ B cells. In summary, the RUNX1/FOXP3 axis alleviated immunosuppression in sepsis progression by weakening T and B lymphocyte apoptosis.
Collapse
Affiliation(s)
- Yangfa Chao
- Department of Surgical Area 4, Shenzhen Bao’an Traditional Chinese Medicine Hospital Group, Shenzhen, Guangdong Province, 518000, China
| | - Wenting Huang
- Department of Acupuncture, Luohu District Chronic Disease Prevention and Treatment Hospital, Shenzhen, China
| | - Zhiheng Xu
- The Second Department of Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Ping Li
- Department of Surgical Area 4, Shenzhen Bao’an Traditional Chinese Medicine Hospital Group, Shenzhen, Guangdong Province, 518000, China
| | - Shaodong Gu
- Department of Surgical Area 4, Shenzhen Bao’an Traditional Chinese Medicine Hospital Group, No. 25
Yu’an 2nd Road, Bao’an District, Shenzhen, Guangdong Province, 518000, China
| |
Collapse
|
12
|
Green EA, Garrick SP, Peterson B, Berger PJ, Galinsky R, Hunt RW, Cho SX, Bourke JE, Nold MF, Nold-Petry CA. The Role of the Interleukin-1 Family in Complications of Prematurity. Int J Mol Sci 2023; 24:2795. [PMID: 36769133 PMCID: PMC9918069 DOI: 10.3390/ijms24032795] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/22/2023] [Indexed: 02/05/2023] Open
Abstract
Preterm birth is a major contributor to neonatal morbidity and mortality. Complications of prematurity such as bronchopulmonary dysplasia (BPD, affecting the lung), pulmonary hypertension associated with BPD (BPD-PH, heart), white matter injury (WMI, brain), retinopathy of prematurity (ROP, eyes), necrotizing enterocolitis (NEC, gut) and sepsis are among the major causes of long-term morbidity in infants born prematurely. Though the origins are multifactorial, inflammation and in particular the imbalance of pro- and anti-inflammatory mediators is now recognized as a key driver of the pathophysiology underlying these illnesses. Here, we review the involvement of the interleukin (IL)-1 family in perinatal inflammation and its clinical implications, with a focus on the potential of these cytokines as therapeutic targets for the development of safe and effective treatments for early life inflammatory diseases.
Collapse
Affiliation(s)
- Elys A. Green
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Steven P. Garrick
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Briana Peterson
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Philip J. Berger
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Robert Galinsky
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia
| | - Rod W. Hunt
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Steven X. Cho
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| | - Jane E. Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3168, Australia
| | - Marcel F. Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
- Monash Newborn, Monash Children’s Hospital, Melbourne, VIC 3168, Australia
| | - Claudia A. Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC 3168, Australia
| |
Collapse
|
13
|
Zhou J, Zhang Y, Zhuang Q. IL2RB affects Th1/Th2 and Th17 responses of peripheral blood mononuclear cells from septic patients. Allergol Immunopathol (Madr) 2023; 51:1-7. [PMID: 37169553 DOI: 10.15586/aei.v51i3.757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 08/30/2022] [Indexed: 05/13/2023]
Abstract
BACKGROUND Immune dysfunction is a common and serious complication of sepsis. This study finds key genes linked to immunity in sepsis. METHODS The "Limma package" was used to analyze GSE154918 datasets for differentially expressed genes. The differentially expressed genes were then enriched for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and interleukin 2 receptor subunit Beta (IL2RB) protein coding gene was chosen for investigation. IL2RB expression in peripheral blood mononuclear cells (PBMC) was assessed by polymerase chain reaction. White blood cells of septic patients and healthy controls were collected from hospitals and linked with acute physiology and chronic health evaluation (APACHE) II, sequential organ failure assessment (SOFA), C-reactive protein (CRP), and procalcitonin (PCT) of septic patients using Pearson's correlation analysis. PBMC cells were transfected with IL2RB, and the effect of transfection was observed on cellular interferon gamma (IFN-γ), interleukin (IL)-12, IL-4, IL-10, and IL-17A. RESULTS A total of 686 differential genes, comprising 446 upregulated and 240 down regulated genes, were identified. The enrichment of KEGG pathway revealed that the majority of differential genes were enriched in the T helper (Th1)/Th2 cell and Th17 cell differentiation pathways. In patients with sepsis, correlation analysis revealed a negative correlation between IL2RB and APACHE II score, SOFA score, CRP, and PCT. IFN-γ and IL-12 levels were elevated in PBMC of septic patients after IL2RB transfection, but IL-4, IL-10, and IL-17A levels were lowered. CONCLUSION Sepsis-induced immunological dysfunction is improved by IL2RB, which also balances Th1/Th2 responses and prevents Th17 activation. © 2023 Codon Publications. Published by Codon Publications.
Collapse
Affiliation(s)
- Jiaqian Zhou
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Zhang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
| | - Qing Zhuang
- Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
| |
Collapse
|
14
|
The design of cell-selective tryptophan and arginine-rich antimicrobial peptides by introducing hydrophilic uncharged residues. Acta Biomater 2022; 153:557-572. [PMID: 36115654 DOI: 10.1016/j.actbio.2022.09.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 08/18/2022] [Accepted: 09/09/2022] [Indexed: 11/20/2022]
Abstract
Antimicrobial peptides (AMPs) are considered to be powerful weapons in the fight against traditional antibiotic resistance due to their unique membrane-disruptive mechanism. The combination of traditional and classical hydrophobic tryptophan (W) residues and hydrophilic charged arginine (R) residues is considered as the first choice for the minimalist design of AMPs due to its potent performance in antibacterial activity. However, some W- and R-rich AMPs that are not rationally designed and contain excessive repeats of W and R residues may cause severe cytotoxicity and hemolysis. To address this issue, we designed the (WRX)n (where X = hydrophilic uncharged amino residues; n = number of repeat units) series engineered peptides with high cell selectivity by introducing hydrophilic uncharged threonine (T), serine (S), glutamine (Q) or asparagine (N) residues into the minimalist design of W- and R-rich AMPs. The results showed that the introduction of these hydrophilic uncharged amino residues, especially T residues, significantly improved the cell selectivity of the W- and R-rich engineered peptides. Among (WRX)n series engineered peptides, T6 presents a mixture structure of β-turn and α-helix. It has broad spectrum and potent antibacterial activity (no activity against probiotics), good biocompatibility, high selectivity index, strong tolerance (physiological salts, serum acid, alkali, and heat conditions), rapid and efficient time-kill kinetics, and no tendency of resistance. Studies on antibacterial mechanism show that T6 exert antibacterial activity mainly by disrupting bacterial cell membrane and inducing the accumulation of reactive oxygen species in bacterial cells. Furthermore, T6 exhibited potent antibacterial and anti-inflammatory capabilities in vivo in a mouse peritonitis-sepsis model infected with Escherichia coli. In conclusion, our study confirms an effective strategy for the minimalist design of highly cell selective W- and R-rich AMPs by introducing hydrophilic uncharged T residues, which may trigger widespread attention to hydrophilic uncharged amino acid residues, including T residues, and provide new insights into the design of peptide-based antibacterial biomaterials. STATEMENT OF SIGNIFICANCE: We have introduced hydrophilic uncharged T, S, Q or N residues into the minimalist design of W- and R-rich engineered peptides and found that the introduction of these hydrophilic uncharged amino residues, especially the T residues, can significantly improve the cell selectivity of W- and R-rich engineered peptides. The target compound T6 showed potent antibacterial activity, high cell selectivity, strong tolerance, good in vivo efficacy and killed bacteria through multiple mechanisms mainly membrane-disruptive. These findings may spark widespread interest in hydrophilic uncharged amino acid residues, and provide new insights into the design of peptide-based antimicrobial biomaterials.
Collapse
|
15
|
Ning M, Liu Y, Wang D, Wei J, Hu G, Xing P. Knockdown of TRIM27 alleviated sepsis-induced inflammation, apoptosis, and oxidative stress via suppressing ubiquitination of PPARγ and reducing NOX4 expression. Inflamm Res 2022; 71:1315-1325. [PMID: 35962797 PMCID: PMC9375190 DOI: 10.1007/s00011-022-01625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/03/2022] [Accepted: 08/05/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Sepsis is a global fatal disease and leads to severe lung injury due to dysfunction of inflammation response. TRIM27 is closely related to the diseased with dysfunction of inflammation response. The aim of this study was to clarify the role and mechanism of TRIM27 in sepsis-induced lung injury. METHODS The lipopolysaccharide (LPS)-induced septic mouse model was successfully established. The lung injury was evaluated by lung wet/dry (W/D) ratio and hematoxylin-eosin (H&E) staining. The cell apoptosis was evaluated by TUNEL assay. The inflammatory cytokines were measured by quantitative real time-PCR (qRT-PCR) assay and commercial enzyme-linked immunosorbent assay (ELISA). The oxidative stress was assessed by the contents of superoxide dismutase (SOD) and malondialdehyde (MDA), and the expression of dihydroethidium (DHE). RESULTS In this study, we demonstrated that TRIM27 was up-regulated in LPS-induced septic mice. In loss-of-function experiments, knockdown of TRIM27 alleviated sepsis-induced lung injury, inflammation, apoptosis, and oxidative stress. More importantly, knockdown of TRIM27 was observed to reduce p-p65/NOX4 expression via suppressing ubiquitination of PPARγ. In rescue experiments, overexpression of NOX4 abolished the effect of sh-TRIM27 on alleviating sepsis-induced inflammation, apoptosis, and oxidative stress. CONCLUSION These findings highlighted that knockdown of TRIM27 alleviated sepsis-induced inflammation, oxidative stress and apoptosis via suppressing ubiquitination of PPARγ and reducing NOX4 expression, which supports the potential utility of TRIM27 as a therapeutic target in septic lung injury.
Collapse
Affiliation(s)
- Meng Ning
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Yingwu Liu
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Donglian Wang
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China
| | - Jin Wei
- Department of Heart Center, Tianjin Third Central Hospital, Tianjin, 300170, China.,Department of Heart Center, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, 300170, China.,Department of Heart Center, Artificial Cell Engineering Technology Research Center, Tianjin, 300170, China
| | - Guoyong Hu
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China
| | - Pengcheng Xing
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 222, West Three Road Around Lake, Nanhui New Town, Pudong New Area, Shanghai, China.
| |
Collapse
|
16
|
Chen S, Kuang M, Qu Y, Huang S, Gong B, Lin S, Wang H, Wang G, Tao H, Yu J, Yang Z, Jiang M, Xie Q. Expression of Serum Cytokines Profile in Neonatal Sepsis. Infect Drug Resist 2022; 15:3437-3445. [PMID: 35794925 PMCID: PMC9252297 DOI: 10.2147/idr.s368772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/23/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Sepsis remains a major cause of neonatal death. To better characterize the inflammatory response during neonatal sepsis, we compared the differences in serum cytokines and chemokines between full-term neonates with sepsis and without infection. Methods We enrolled 40 full-term neonates with sepsis and 26 full-term neonates without infection as controls between October 2016 and June 2018. Forty cytokines /chemokines in serum were analyzed using the Luminex Bead Immunoassay System. Results Our results showed that serum IL-6, IL-8, TNF-α, IL-1β, MIF, CXCL13, CXCL1, CXCL2, CXCL5, CXCL6, CXCL16, CCL27, CCL2, CCL8, CCL3, CCL20, CCL23, and CX3CL1 levels were significantly increased in neonates with sepsis compared to those in the control group (all p<0.05). The levels of serum CCL20, and IL-17 were higher in late-onset sepsis (LOS) than those in early-onset sepsis (EOS) (all p<0.05). Conversely, serum CXCL16 was lower in LOS than that in EOS (p<0.05). Conclusion Our findings revealed that excessive pro-inflammatory cytokines might be involved in neonatal sepsis. In addition, chemokines significantly increased the recruitment of immune cells after infection to participate in the anti-infection defense of neonates, but this could lead to damage.
Collapse
Affiliation(s)
- Suipeng Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Mengjiao Kuang
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Ying Qu
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,Department of Clinical Laboratory, Wenzhou People's Hospital, The Third Affiliated Hospital of Shanghai University, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Shirui Huang
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Binbin Gong
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Suzhen Lin
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Huiyan Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Guiye Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Hongqun Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Jian Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Zuqin Yang
- Newborn Department of Pediatrics, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Minghua Jiang
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Qipeng Xie
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China
| |
Collapse
|
17
|
Frimpong A, Owusu EDA, Amponsah JA, Obeng-Aboagye E, van der Puije W, Frempong AF, Kusi KA, Ofori MF. Cytokines as Potential Biomarkers for Differential Diagnosis of Sepsis and Other Non-Septic Disease Conditions. Front Cell Infect Microbiol 2022; 12:901433. [PMID: 35811678 PMCID: PMC9260692 DOI: 10.3389/fcimb.2022.901433] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 01/08/2023] Open
Abstract
Sepsis defined as a dysregulated immune response is a major cause of morbidity in children. In sub-Saharan Africa, the clinical features of sepsis overlap with other frequent infections such as malaria, thus sepsis is usually misdiagnosed in the absence of confirmatory tests. Therefore, it becomes necessary to identify biomarkers that can be used to distinguish sepsis from other infectious diseases. We measured and compared the plasma levels of 18 cytokines (Th1 [GM-CSF, IFN-γ, TNF-α, IL-1β, 1L-2, IL-6, IL-8, IL-12/IL-23p40, IL-15], Th2[IL-4, IL-5, IL-13), Th17 [IL17A], Regulatory cytokine (IL-10) and 7 chemokines (MCP-1/CCL2, MIP-1α/CCL3, MIP-1β/CCL4, RANTES/CCL5, Eotaxin/CCL11, MIG/CXCL9 and IP-10/CXCL10 using the Human Cytokine Magnetic 25-Plex Panel in plasma samples obtained from children with sepsis, clinical malaria and other febrile conditions. Children with sepsis had significantly higher levels of IL-1β, IL-12 and IL-17A compared to febrile controls but lower levels of MIP1-β/CCL4, RANTES/CCL5 and IP10/CXCL10 when compared to children with malaria and febrile controls. Even though levels of most inflammatory responses were higher in malaria compared to sepsis, children with sepsis had a higher pro-inflammatory to anti-inflammatory ratio which seemed to be mediated by mostly monocytes. A principal component analysis and a receiver operator characteristic curve analysis, identified seven potential biomarkers; IL-1β, IL-7, IL-12, IL-1RA, RANTES/CCL5, MIP1β/CCL4 and IP10/CXCL10 that could discriminate children with sepsis from clinical malaria and other febrile conditions. The data suggests that sepsis is associated with a higher pro-inflammatory environment. These pro-inflammatory cytokines/chemokines could further be evaluated for their diagnostic potential to differentiate sepsis from malaria and other febrile conditions in areas burdened with infectious diseases.
Collapse
Affiliation(s)
- Augustina Frimpong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- *Correspondence: Augustina Frimpong, ; Michael Fokuo Ofori,
| | - Ewurama D. A. Owusu
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Jones Amo Amponsah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Elizabeth Obeng-Aboagye
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - William van der Puije
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Abena Fremaah Frempong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- *Correspondence: Augustina Frimpong, ; Michael Fokuo Ofori,
| |
Collapse
|
18
|
Regulation of S100As Expression by Inflammatory Cytokines in Chronic Lymphocytic Leukemia. Int J Mol Sci 2022; 23:ijms23136952. [PMID: 35805957 PMCID: PMC9267105 DOI: 10.3390/ijms23136952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The calcium-binding proteins S100A4, S100A8, and S100A9 are upregulated in chronic lymphocytic leukemia (CLL), while the S100A9 promotes NF-κB activity during disease progression. The S100-protein family has been involved in several malignancies as mediators of inflammation and proliferation. The hypothesis of our study is that S100A proteins are mediators in signaling pathways associated with inflammation-induced proliferation, such as NF-κB, PI3K/AKT, and JAK/STAT. The mononuclear cells (MNCs) of CLL were treated with proinflammatory IL-6, anti-inflammatory IL-10 cytokines, inhibitors of JAK1/2, NF-κB, and PI3K signaling pathways, to evaluate S100A4, S100A8, S100A9, and S100A12 expression as well as NF-κB activation by qRT-PCR, immunocytochemistry, and immunoblotting. The quantity of S100A4, S100A8, and S100A9 positive cells (p < 0.05) and their protein expression (p < 0.01) were significantly decreased in MNCs of CLL patients compared to healthy controls. The S100A levels were generally increased in CD19+ cells compared to MNCs of CLL. The S100A4 gene expression was significantly stimulated (p < 0.05) by the inhibition of the PI3K/AKT signaling pathway in MNCs. IL-6 stimulated S100A4 and S100A8 protein expression, prevented by the NF-κB and JAK1/2 inhibitors. In contrast, IL-10 reduced S100A8, S100A9, and S100A12 protein expressions in MNCs of CLL. Moreover, IL-10 inhibited activation of NF-κB signaling (4-fold, p < 0.05). In conclusion, inflammation stimulated the S100A protein expression mediated via the proliferation-related signaling and balanced by the cytokines in CLL.
Collapse
|
19
|
Hames RG, Jasiunaite Z, Ercoli G, Wanford JJ, Carreno D, Straatman K, Martinez-Pomares L, Yesilkaya H, Glenn S, Moxon ER, Andrew PW, Kyriacou CP, Oggioni MR. Diurnal Differences in Intracellular Replication Within Splenic Macrophages Correlates With the Outcome of Pneumococcal Infection. Front Immunol 2022; 13:907461. [PMID: 35720383 PMCID: PMC9201068 DOI: 10.3389/fimmu.2022.907461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 12/04/2022] Open
Abstract
Circadian rhythms affect the progression and severity of bacterial infections including those caused by Streptococcus pneumoniae, but the mechanisms responsible for this phenomenon remain largely elusive. Following advances in our understanding of the role of replication of S. pneumoniae within splenic macrophages, we sought to investigate whether events within the spleen correlate with differential outcomes of invasive pneumococcal infection. Utilising murine invasive pneumococcal disease (IPD) models, here we report that infection during the murine active phase (zeitgeber time 15; 15h after start of light cycle, 3h after start of dark cycle) resulted in significantly faster onset of septicaemia compared to rest phase (zeitgeber time 3; 3h after start of light cycle) infection. This correlated with significantly higher pneumococcal burden within the spleen of active phase-infected mice at early time points compared to rest phase-infected mice. Whole-section confocal microscopy analysis of these spleens revealed that the number of pneumococci is significantly higher exclusively within marginal zone metallophilic macrophages (MMMs) known to allow intracellular pneumococcal replication as a prerequisite step to the onset of septicaemia. Pneumococcal clusters within MMMs were more abundant and increased in size over time in active phase-infected mice compared to those in rest phase-infected mice which decreased in size and were present in a lower percentage of MMMs. This phenomenon preceded significantly higher levels of bacteraemia alongside serum IL-6 and TNF-α concentrations in active phase-infected mice following re-seeding of pneumococci into the blood. These data greatly advance our fundamental knowledge of pneumococcal infection by linking susceptibility to invasive pneumococcal infection to variation in the propensity of MMMs to allow persistence and replication of phagocytosed bacteria. These findings also outline a somewhat rare scenario whereby the active phase of an organism’s circadian cycle plays a seemingly counterproductive role in the control of invasive infection.
Collapse
Affiliation(s)
- Ryan G Hames
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Zydrune Jasiunaite
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Giuseppe Ercoli
- Centre for Inflammation and Tissue Repair, UCL Respiratory, Division of Medicine, University College Medical School, London, United Kingdom
| | - Joseph J Wanford
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - David Carreno
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Kornelis Straatman
- Advanced Imaging Facility, University of Leicester, Leicester, United Kingdom
| | | | - Hasan Yesilkaya
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Sarah Glenn
- Preclinical Research Facility, University of Leicester, Leicester, United Kingdom
| | - E Richard Moxon
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Peter W Andrew
- Department of Respiratory Sciences, University of Leicester, Leicester, United Kingdom
| | - Charalambos P Kyriacou
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Marco R Oggioni
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
20
|
miR-197 Participates in Lipopolysaccharide-Induced Cardiomyocyte Injury by Modulating SIRT1. Cardiol Res Pract 2022; 2022:7687154. [PMID: 35223094 PMCID: PMC8872679 DOI: 10.1155/2022/7687154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/11/2022] [Indexed: 11/25/2022] Open
Abstract
Sepsis is a systemic inflammation and is capable of inducing myocarditis, which is a major leading cause of death in patients. Studies have found that miR-197 is correlated with the prognosis of patients with inflammatory heart disease, but its effect on sepsis-induced cardiomyocyte injury remains unclear. We treated H9c2 cells with lipopolysaccharide (LPS), then detected the cell viability via the cell counting kit-8 (CCK-8) assay and quantified miR-197 expression via quantitative real-time polymerase chain reaction (qRT-PCR). Then, we investigated the role of miR-197 in LPS-induced H9c2 cells by CCK-8 assay, flow cytometry, lactate dehydrogenase (LDH) measurement, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, and western blot. Subsequently, silent information regulator 1 (SIRT1) was downregulated in H9c2 cells to explore its interaction with miR-197 under LPS induction. LPS induced miR-197 overexpression in H9c2 cells. LPS restrained viability, the expressions of B-cell lymphoma-2 (Bcl-2) and SIRT1, but promoted apoptosis, LDH release, and levels of interleukin-6 (IL-6), interleukin-1β (IL-1β), acetyl (AC)-p53, BCL2-associated X (Bax), and cleaved caspase-3 in H9c2 cells. miR-197 inhibition reversed the effects of LPS on H9c2 cells. The protective role of miR-197 downregulation in LPS-induced H9c2 cells was reversed by SIRT1 silencing. miR-197 contributed to LPS-induced cardiomyocyte injury by modulating SIRT1, which might be used as a molecular marker in the management of sepsis.
Collapse
|
21
|
Nguyen TH, Turek I, Meehan-Andrews T, Zacharias A, Irving HR. A systematic review and meta-analyses of interleukin-1 receptor associated kinase 3 (IRAK3) action on inflammation in in vivo models for the study of sepsis. PLoS One 2022; 17:e0263968. [PMID: 35167625 PMCID: PMC8846508 DOI: 10.1371/journal.pone.0263968] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 02/01/2022] [Indexed: 12/27/2022] Open
Abstract
Background Interleukin-1 receptor associated kinase 3 (IRAK3) is a critical modulator of inflammation and is associated with endotoxin tolerance and sepsis. Although IRAK3 is known as a negative regulator of inflammation, several studies have reported opposing functions, and the temporal actions of IRAK3 on inflammation remain unclear. A systematic review and meta-analyses were performed to investigate IRAK3 expression and its effects on inflammatory markers (TNF-α and IL-6) after one- or two-challenge interventions, which mimic the hyperinflammatory and immunosuppression phases of sepsis, respectively, using human or animal in vivo models. Methods This systematic review and meta-analyses has been registered in the Open Science Framework (OSF) (Registration DOI: 10.17605/OSF.IO/V39UR). A systematic search was performed to identify in vivo studies reporting outcome measures of expression of IRAK3 and inflammatory markers. Meta-analyses were performed where sufficient data was available. Results The search identified 7778 studies for screening. After screening titles, abstracts and full texts, a total of 49 studies were included in the systematic review. The review identified significant increase of IRAK3 mRNA and protein expression at different times in humans compared to rodents following one-challenge, whereas the increases of IL-6 and TNF-α protein expression in humans were similar to rodent in vivo models. Meta-analyses confirmed the inhibitory effect of IRAK3 on TNF-α mRNA and protein expression after two challenges. Conclusions A negative correlation between IRAK3 and TNF-α expression in rodents following two challenges demonstrates the association of IRAK3 in the immunosuppression phase of sepsis. Species differences in underlying biology affect the translatability of immune responses of animal models to human, as shown by the dissimilarity in patterns of IRAK3 mRNA and protein expression between humans and rodents following one challenge that are further influenced by variations in experimental procedures.
Collapse
Affiliation(s)
- Trang H. Nguyen
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
- * E-mail: (HRI); (THN)
| | - Ilona Turek
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Terri Meehan-Andrews
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Anita Zacharias
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Helen R. Irving
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
- * E-mail: (HRI); (THN)
| |
Collapse
|
22
|
Niknam M, Liaghat T, Zarghami M, Akrami M, Shahnematollahi SM, Ahmadipour A, Moazzen F, Soltanabadi S. Ghrelin and ghrelin/total cholesterol ratio as independent predictors for coronary artery disease: a systematic review and meta-analysis. J Investig Med 2022; 70:759-765. [PMID: 35042826 DOI: 10.1136/jim-2021-002100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 11/04/2022]
Abstract
The present meta-analysis aimed to summarize the available data regarding the circulating levels of ghrelin in patients with cardiovascular diseases (CVDs). A comprehensive search was performed in electronic databases including PubMed, Scopus, EMBASE, and Web of Science up to January 20, 2021. Since the circulating levels of ghrelin were measured in different units across the included studies, they were expressed as the standardized mean difference (SMD) and 95% CI (summary effect size). A random-effects model comprising the DerSimonian and Laird method was used to pool SMDs. Sixteen articles (20 studies) comprised of 1087 cases and 437 controls were included. The pooled results showed that there were no significant differences between cases and controls in terms of ghrelin levels (SMD=-0.61, 95% CI -1.38 to 0.16; p=0.120; I2=96.9%, p<0.001). The ghrelin concentrations in the CAD stratum were significantly lower than in controls, whereas they increased in other disease strata. New combined biomarkers demonstrated a significant decrease in the SMD of the ghrelin/total cholesterol (TC) ratio (-1.02; 95% CI -1.74 to -0.29, p=0.000; I2=94.5%). However, no significant differences were found in the SMD of the ghrelin/high-density lipoprotein cholesterol ratio, ghrelin/low-density lipoprotein cholesterol ratio, and ghrelin/triglyceride (TG) ratio in cases with CVDs compared with the control group. Ghrelin was associated with CAD; therefore, it may be considered a biomarker for distinguishing between patients with and without CAD. Furthermore, the ghrelin/TC ratio could be proposed as a diagnostic marker for CVD.
Collapse
Affiliation(s)
- Maryam Niknam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Taraneh Liaghat
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Zarghami
- Cardiology Department, Fasa University of Medical Science, Fasa, Iran
| | - Mehdi Akrami
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Ahmad Ahmadipour
- Student Research Committee, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fatemeh Moazzen
- Department of Hematology, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Sahar Soltanabadi
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
23
|
Wu D, Wang L, Hong D, Zheng C, Zeng Y, Ma H, Lin J, Chen J, Zheng R. Interleukin 35 contributes to immunosuppression by regulating inflammatory cytokines and T cell populations in the acute phase of sepsis. Clin Immunol 2022; 235:108915. [PMID: 34995813 DOI: 10.1016/j.clim.2021.108915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/17/2021] [Accepted: 12/22/2021] [Indexed: 12/30/2022]
Abstract
Cytokines interact closely with each other and play a crucial role in the progression of sepsis. We focused on the associations of a cytokine network with IL-35 in sepsis. First, the retrospective study included 42 patients with sepsis and 23 healthy controls. Blood samples were collected from patients on days 1, 2, 4. Levels of IL-35, IL-1β, IL-4, IL-6, IL-10, IL-17A, TNF-α and IFN-γ were measured. They all increased to various extend on days 1, 2, 4, and strongly associated with markers of disease severity. Network analysis revealed a network formed by IL-35, with IL-6, IL-10, IL-17A, TNF-α and IFN-γ throughout the acute phase of sepsis(days 1, 2 and4). Then, the CLP-induced septic rats were used. The recombinant human IL-35(rIL-35) upregulated the levels of IL-10, but downregulated IL-4, IL-6, IL-17A, TNF-α and IFN-γ, while it had no significant effect on IL-1β, and upregulated the percentages of CD4+CD25+Tregs, and iTR35, but downregulated Teff cells in the peripheral blood. The rIL-35 reduced inflammation damage and improved prognosis of the septic rats. IL-35 forms a network with other cytokines and plays a major role in the immunopathogenesis of sepsis.
Collapse
Affiliation(s)
- Dansen Wu
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China.
| | - Liming Wang
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Donghuang Hong
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Caifa Zheng
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Yongping Zeng
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Huolan Ma
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Jing Lin
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Jialong Chen
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Ronghui Zheng
- Department of Critical Care Medicine, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fujian Medical University, Fuzhou 350001, Fujian, China
| |
Collapse
|
24
|
Zhou Y, Qing M, Xu M. Circ-BNIP3L knockdown alleviates LPS-induced renal tubular epithelial cell injury during sepsis-associated acute kidney injury by miR-370-3p/MYD88 axis. J Bioenerg Biomembr 2021; 53:665-677. [PMID: 34731384 DOI: 10.1007/s10863-021-09925-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a frequent complication of the critically ill patient with high morbidity and mortality. Thus, the goal of this study was to investigate the role of circular RNA BCL2 Interacting Protein 3 Like (circ-BNIP3L) in the pathophysiological mechanism of SA-AKI. The SA-AKI cell model was established by using lipopolysaccharide (LPS)-induced HK-2 cells in vitro. Cell survival was analyzed using cell counting kit-8 (CCK-8) assay, EdU (5-ethynyl-2'-deoxyuridine) assay, flow cytometry and Western blot, respectively. Levels of tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were detected using ELISA analysis. The superoxide dismutase (SOD) activity and malondialdehyde (MDA) level were examined using commercial kits. Levels of genes and proteins were detected by qRT-PCR and Western blot. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were used to identify the target relationship between miR-370-3p and circ-BNIP3L or MYD88 (myeloid differentiation primary response 88). Circ-BNIP3L was highly expressed in SA-AKI patients and LPS-induced HK-2 cells. Silencing of circ-BNIP3L attenuated LPS-induced growth inhibition, inflammation, and oxidative stress in HK-2 cells. Mechanistically, circ-BNIP3L competitively bound to miR-370-3p to up-regulate the expression of its target MYD88. Moreover, miR-370-3p inhibition reversed the beneficial effects of circ-BNIP3L knockdown on LPS-stimulated HK-2 cells. Meanwhile, miR-370-3p overexpression abolished LPS-induced injury in HK-2 cells, which was counteracted by MYD88 up-regulation. Circ-BNIP3L knockdown alleviated LPS-induced renal tubular epithelial cell injury by miR-370-3p/MYD88 axis, opening up a completely new avenue for the treatment of sepsis-associated AKI.
Collapse
Affiliation(s)
- Yanyan Zhou
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, No.139 Renmin Road, 410000, Changsha City, Hunan Province, China
| | - Meiying Qing
- Department of Urinary Surgery, The Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China
| | - Min Xu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, No.139 Renmin Road, 410000, Changsha City, Hunan Province, China.
| |
Collapse
|
25
|
Ashley BK, Hassan U. Point-of-critical-care diagnostics for sepsis enabled by multiplexed micro and nanosensing technologies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1701. [PMID: 33650293 PMCID: PMC8447248 DOI: 10.1002/wnan.1701] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 11/12/2022]
Abstract
Sepsis is responsible for the highest economic and mortality burden in critical care settings around the world, prompting the World Health Organization in 2018 to designate it as a global health priority. Despite its high universal prevalence and mortality rate, a disproportionately low amount of sponsored research funding is directed toward diagnosis and treatment of sepsis, when early treatment has been shown to significantly improve survival. Additionally, current technologies and methods are inadequate to provide an accurate and timely diagnosis of septic patients in multiple clinical environments. For improved patient outcomes, a comprehensive immunological evaluation is critical which is comprised of both traditional testing and quantifying recently proposed biomarkers for sepsis. There is an urgent need to develop novel point-of-care, low-cost systems which can accurately stratify patients. These point-of-critical-care sensors should adopt a multiplexed approach utilizing multimodal sensing for heterogenous biomarker detection. For effective multiplexing, the sensors must satisfy criteria including rapid sample to result delivery, low sample volumes for clinical sample sparring, and reduced costs per test. A compendium of currently developed multiplexed micro and nano (M/N)-based diagnostic technologies for potential applications toward sepsis are presented. We have also explored the various biomarkers targeted for sepsis including immune cell morphology changes, circulating proteins, small molecules, and presence of infectious pathogens. An overview of different M/N detection mechanisms are also provided, along with recent advances in related nanotechnologies which have shown improved patient outcomes and perspectives on what future successful technologies may encompass. This article is categorized under: Diagnostic Tools > Biosensing.
Collapse
Affiliation(s)
- Brandon K. Ashley
- Department of Biomedical Engineering, Rutgers, State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Umer Hassan
- Department of Biomedical Engineering, Rutgers, State University of New Jersey, Piscataway, NJ, 08854, USA
- Department of Electrical Engineering, Rutgers, State University of New Jersey, Piscataway, NJ, 08854, USA
- Global Health Institute, Rutgers, State University of New Jersey. Piscataway, NJ, 08854, USA
| |
Collapse
|
26
|
Bengnér J, Quttineh M, Gäddlin PO, Salomonsson K, Faresjö M. Serum amyloid A - A prime candidate for identification of neonatal sepsis. Clin Immunol 2021; 229:108787. [PMID: 34175457 DOI: 10.1016/j.clim.2021.108787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/20/2021] [Accepted: 06/20/2021] [Indexed: 10/21/2022]
Abstract
Neonatal sepsis is common, lethal, and hard to diagnose. In combination with clinical findings and blood culture, biomarkers are crucial to make the correct diagnose. A Swedish national inquiry indicated that neonatologists were not quite satisfied with the available biomarkers. We assessed the kinetics of 15 biomarkers simultaneously: ferritin, fibrinogen, granulocyte colony-stimulating factor (G-CSF), interferon (IFN)-γ, interleukin (IL)-1β, -6, -8, -10, macrophage inflammatory protein (MIP)-1β, procalcitonin, resistin, serum amyloid A (SAA), tumor necrosis factor (TNF)-α, tissue plasminogen activator-3 and visfatin. The goal was to observe how quickly they rise in response to infection, and for how long they remain elevated. From a neonatal intensive care unit, newborns ≥28 weeks gestational age were recruited. Sixty-eight newborns were recruited to the study group (SG), and fifty-one to the control group (CG). The study group subjects were divided into three subgroups depending on clinical findings: confirmed sepsis (CSG), suspected sepsis (SSG) and no sepsis. CSG and SSG were also merged into an entire sepsis group (ESG) for sub-analysis. Blood samples were collected at three time-points; 0 h, 12-24 h and 48-72 h, in order to mimic a "clinical setting". At 0 h, visfatin was elevated in SSG compared to CG; G-CSF, IFN-γ, IL-1β, -8 and - 10 were elevated in SSG and ESG compared to CG, whereas IL-6 and SAA were elevated in all groups compared to CG. At 12-24 h, IL-8 was elevated in ESG compared to CG, visfatin was elevated in ESG and SSG compared to CG, and SAA was elevated in all three groups compared to CG. At 48-72 h, fibrinogen was elevated in ESG compared to CG, IFN-γ and IL-1β were elevated in SSG and ESG compared to CG, whereas IL-8 and SAA were elevated in all three groups compared to CG. A function of time-formula is introduced as a tool for theoretical prediction of biomarker levels at any time-point. We conclude that SAA has the most favorable kinetics regarding diagnosing neonatal sepsis, of the biomarkers studied. It is also readily available methodologically, making it a prime candidate for clinical use.
Collapse
Affiliation(s)
- Johannes Bengnér
- Paediatric Clinic, Ryhov County Hospital, Region Jönköping County, Jönköping, Sweden
| | - Maysae Quttineh
- Department of Laboratory Medicine, Region Jönköping County, Jönköping, Sweden
| | - Per-Olof Gäddlin
- Paediatric Clinic, Ryhov County Hospital, Region Jönköping County, Jönköping, Sweden
| | - Kent Salomonsson
- Virtual Engineering Research Environment, School of Engineering Science, University of Skövde, Skövde, Sweden
| | - Maria Faresjö
- Biomedical Platform, Department of Natural Science and Biomedicine, School of Health and Welfare, Jönköping University, Jönköping, Sweden; Department of Biology and Biological Engineering, Chalmers University of Technology, Göteborg, Sweden.
| |
Collapse
|
27
|
Su Y, Song X, Teng J, Zhou X, Dong Z, Li P, Sun Y. Mesenchymal stem cells-derived extracellular vesicles carrying microRNA-17 inhibits macrophage apoptosis in lipopolysaccharide-induced sepsis. Int Immunopharmacol 2021; 95:107408. [PMID: 33915488 DOI: 10.1016/j.intimp.2021.107408] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Sepsis, as a disease affecting the microcirculation and tissue perfusion, results in tissue hypoxia and multiple organ dysfunctions. Bone mesenchymal stem cell (BMSC)-derived extracellular vesicles (EVs) have been demonstrated to transfer trivial molecules (proteins/peptides, mRNA, microRNA and lipids) to alleviate sepsis. We sought to define the function of microRNA (miR)-17 carried in BMSC-EVs in sepsis. METHODS The purity of the extracted BMSCs was identified and confirmed by detection of the surface markers by flow cytometry, followed by osteoblastic, adipogenic, and chondrocyte differentiation experiments. Subsequently, EVs were collected from the medium of BMSCs. The uptake of PKH-67-labeled BMSC-EVs or EVs carrying cy3-miR-17 by RAW264.7 cells was observed under laser confocal microscopy. Furthermore, a series of gain- and loss-of-function approaches were conducted to test the effects of LPS, miR-17 and BRD4 on the inflammatory factors (IL-1β, IL-6 and TNF-α), number of M1 macrophages and M2 macrophages, inflammatory-related signal pathway factors (EZH2, c-MYC and TRAIL), macrophage proliferation, and apoptosis in sepsis. The survival rates were measured in vivo. RESULTS BMSC-EVs was internalized by the RAW264.7 cells. BDR4 was verified as a target of miR-17, while the expression pattern of miR-17 was upregulated in BMSC-EVs. MiR-17 carried by BMSC-EVs inhibited LPS-induced inflammation and apoptosis of RAW264.7 cells, but improved the viability of RAW264.7 cells. Next, in vitro experiments supported that miR-17 inhibited LPS-induced inflammation in RAW264.7 cells through BRD4/EZH2/TRAIL axis. BRD4 overexpression reversed the effects of miR-17. Moreover, the therapeutic function of BMSC-EVs carried miR-17 was verified by in vivo experiments. CONCLUSIONS MiR-17 derived from BMSCs-EVs regulates BRD4-mediated EZH2/TRAIL axis to essentially inhibit LPS-induced macrophages inflammation.
Collapse
Affiliation(s)
- Yuan Su
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Xiaoxia Song
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Jinlong Teng
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Xinbei Zhou
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Zehua Dong
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Ping Li
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China
| | - Yunbo Sun
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, PR China.
| |
Collapse
|
28
|
Nguyen TH, Turek I, Meehan-Andrews T, Zacharias A, Irving H. Analysis of interleukin-1 receptor associated kinase-3 (IRAK3) function in modulating expression of inflammatory markers in cell culture models: A systematic review and meta-analysis. PLoS One 2020; 15:e0244570. [PMID: 33382782 PMCID: PMC7774834 DOI: 10.1371/journal.pone.0244570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 12/13/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND IRAK3 is a critical modulator of inflammation in innate immunity. IRAK3 is associated with many inflammatory diseases, including sepsis, and is required in endotoxin tolerance to maintain homeostasis of inflammation. The impact of IRAK3 on inflammatory markers such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in cell culture models remains controversial. OBJECTIVE To analyse temporal effects of IRAK3 on inflammatory markers after one- or two-challenge interventions in cell culture models. METHODS A systematic search was performed to identify in vitro cell studies reporting outcome measures of expression of IRAK3 and inflammatory markers. Meta-analyses were performed where sufficient data were available. Comparisons of outcome measures were performed between different cell lines and human and mouse primary cells. RESULTS The literature search identified 7766 studies for screening. After screening titles, abstracts and full-texts, a total of 89 studies were included in the systematic review. CONCLUSIONS The review identifies significant effects of IRAK3 on decreasing NF-κB DNA binding activity in cell lines, TNF-α protein level at intermediate time intervals (4h-15h) in cell lines or at long term intervals (16h-48h) in mouse primary cells following one-challenge. The patterns of TNF-α protein expression in human cell lines and human primary cells in response to one-challenge are more similar than in mouse primary cells. Meta-analyses confirm a negative correlation between IRAK3 and inflammatory cytokine (IL-6 and TNF-α) expression after two-challenges.
Collapse
Affiliation(s)
- Trang Hong Nguyen
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Ilona Turek
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Terri Meehan-Andrews
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Anita Zacharias
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Helen Irving
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| |
Collapse
|
29
|
Davis MD, Winters BR, Madden MC, Pleil JD, Sessler CN, Wallace MAG, Ward-Caviness CK, Montpetit AJ. Exhaled breath condensate biomarkers in critically ill, mechanically ventilated patients. J Breath Res 2020; 15:016011. [DOI: 10.1088/1752-7163/abc235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Yang KD, He Y, Xiao S, Ai Q, Yu JL. Identification of progranulin as a novel diagnostic biomarker for early-onset sepsis in neonates. Eur J Clin Microbiol Infect Dis 2020; 39:2405-2414. [PMID: 32720089 DOI: 10.1007/s10096-020-03981-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/03/2020] [Indexed: 10/23/2022]
Abstract
Neonatal early-onset sepsis (EOS) is associated with high morbidity and mortality. Accurate early diagnosis is crucial for prompt treatment and a better clinical outcome. We aimed to identify new biomarkers for the diagnosis of EOS. A total of 152 neonates with a risk of EOS were divided into an EOS group and a non-EOS group according to the conventional diagnostic criteria. Blood samples were collected within 0-24, 24-48, and 48-72 h after birth. Serum levels of progranulin (PGRN), interleukin (IL)-33, IL-17a, IL-23, IL-6, tumor necrosis factors α (TNF-α), interferon γ (IFN-γ), granulocyte-macrophage colony-stimulating factor (GM-CSF), procalcitonin (PCT), and C-reactive protein (CRP) were determined. PGRN levels were significantly elevated in the EOS neonates compared with the levels in the non-EOS neonates (1.53 vs. 0.77 ng/ml (median), P < 0.001), with an area under the receiver operating characteristic (ROC) curve (AUC) of 0.76 (P < 0.001). Compared with PGRN, IL-33, IL-17a, IL-23, IL-6, PCT, and CRP showed significant (AUC > 0.70) but slightly less predictive power for EOS within the same time range. Stepwise multivariate regression analysis identified PGRN, IL-33, and PCT as independent predictors of EOS. In addition, the combined measurements of PGRN, IL-33, and PCT showed significantly higher predictive power for EOS than any of the three markers alone. PGRN showed greater efficacy for predicting EOS than the traditional markers PCT and CRP as well as other potential markers tested in this study. PGRN may serve as an effective biomarker for the early diagnosis of EOS.
Collapse
Affiliation(s)
- Kai-Di Yang
- Department of Neonatology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road, Yuzhong District, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Yu He
- Department of Neonatology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road, Yuzhong District, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Sa Xiao
- Department of Neonatology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road, Yuzhong District, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Qing Ai
- Department of Neonatology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road, Yuzhong District, Chongqing, 400014, China.,Chongqing Key Laboratory of Pediatrics, Chongqing, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
| | - Jia-Lin Yu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, 136 Zhongshan Road, Yuzhong District, Chongqing, 400014, China. .,Department of Pediatrics, Shenzhen University General Hospital, Shenzhen, Guangdong, China. .,Chongqing Key Laboratory of Pediatrics, Chongqing, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China. .,China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China.
| |
Collapse
|
31
|
Liu X, Zhu N, Zhang B, Xu SB. Long Noncoding RNA TCONS_00016406 Attenuates Lipopolysaccharide-Induced Acute Kidney Injury by Regulating the miR-687/PTEN Pathway. Front Physiol 2020; 11:622. [PMID: 32655407 PMCID: PMC7325890 DOI: 10.3389/fphys.2020.00622] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 01/03/2023] Open
Abstract
Acute kidney injury (AKI) is a common and serious complication of sepsis accompanied by kidney dysfunction resulting from various etiologies and pathophysiological processes. Unfortunately, there is currently no ideal therapeutic strategy for AKI. Numerous studies have confirmed that long noncoding RNAs (lncRNAs) play important regulatory roles in the pathogenesis of sepsis-associated AKI. In this study, lncRNA TCONS_00016406 (termed lncRNA 6406), a novel lncRNA identified by using TargetScan, was significantly downregulated in the kidney tissues of mice with sepsis-associated AKI. This study aimed to explore the role of lncRNA 6406 in lipopolysaccharide (LPS)-induced AKI and its potential molecular mechanism. The models of sepsis-induced AKI (called LPS-induced AKI models) in mice and cell lines were established with male C57BL/6 mice and renal tubular epithelial (PTEC) cells, respectively. Twenty-four hours after LPS administration, kidneys and cell samples were collected after various treatments to examine the alterations in the lncRNA 6406 levels and to evaluate the effects on LPS-induced inflammation, oxidative stress, and apoptosis through real-time PCR (RT-PCR) analysis, western blotting, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining. The results revealed that lncRNA 6406 could significantly attenuate LPS-induced AKI, as shown by the alleviation of inflammation, the suppression of oxidative stress and the inhibition of apoptosis. Mechanistically, a luciferase reporter assay and additional research showed that lncRNA 6406 functioned as a ceRNA to sponge miRNA-687, thereby modulating LPS-stimulated AKI by targeting the miR-687/PTEN axis; thus, this study presents a novel therapeutic strategy or sepsis-associated AKI.
Collapse
Affiliation(s)
- Xuelan Liu
- Department of Emergency, Ningbo Medical Center Li Huili Hospital, Ningbo, China
| | - Na Zhu
- Department of Emergency, Ningbo Medical Center Li Huili Hospital, Ningbo, China
| | - Bo Zhang
- Department of Emergency, Ningbo Medical Center Li Huili Hospital, Ningbo, China
| | - Shao Bo Xu
- Department of Emergency, Ningbo Medical Center Li Huili Hospital, Ningbo, China
| |
Collapse
|
32
|
Ng S, Strunk T, Lee AH, Gill EE, Falsafi R, Woodman T, Hibbert J, Hancock REW, Currie A. Whole blood transcriptional responses of very preterm infants during late-onset sepsis. PLoS One 2020; 15:e0233841. [PMID: 32479514 PMCID: PMC7263612 DOI: 10.1371/journal.pone.0233841] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Host immune responses during late-onset sepsis (LOS) in very preterm infants are poorly characterised due to a complex and dynamic pathophysiology and challenges in working with small available blood volumes. We present here an unbiased transcriptomic analysis of whole peripheral blood from very preterm infants at the time of LOS. METHODS RNA-Seq was performed on peripheral blood samples (6-29 days postnatal age) taken at the time of suspected LOS from very preterm infants <30 weeks gestational age. Infants were classified based on blood culture positivity and elevated C-reactive protein concentrations as having confirmed LOS (n = 5), possible LOS (n = 4) or no LOS (n = 9). Bioinformatics and statistical analyses performed included pathway over-representation and protein-protein interaction network analyses. Plasma cytokine immunoassays were performed to validate differentially expressed cytokine pathways. RESULTS The blood leukocyte transcriptional responses of infants with confirmed LOS differed significantly from infants without LOS (1,317 differentially expressed genes). However, infants with possible LOS could not be distinguished from infants with no LOS or confirmed LOS. Transcriptional alterations associated with LOS included genes involved in pathogen recognition (mainly TLR pathways), cytokine signalling (both pro-inflammatory and inhibitory responses), immune and haematological regulation (including cell death pathways), and metabolism (altered cholesterol biosynthesis). At the transcriptional-level cytokine responses during LOS were characterised by over-representation of IFN-α/β, IFN-γ, IL-1 and IL-6 signalling pathways and up-regulation of genes for inflammatory responses. Infants with confirmed LOS had significantly higher levels of IL-1α and IL-6 in their plasma. CONCLUSIONS Blood responses in very preterm infants with LOS are characterised by altered host immune responses that appear to reflect unbalanced immuno-metabolic homeostasis.
Collapse
Affiliation(s)
- Sherrianne Ng
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, WA, Australia
- Division of the Institute of Reproductive and Developmental Biology, Imperial College Parturition Research Group, Imperial College London, London, United Kingdom
- March of Dimes European Prematurity Research Centre, Imperial College London, London, United Kingdom
| | - Tobias Strunk
- Department of Health, Neonatal Directorate, King Edward Memorial Hospital, Child and Adolescent Health Service, Perth, WA, Australia
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Amy H. Lee
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erin E. Gill
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Reza Falsafi
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tabitha Woodman
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, WA, Australia
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Julie Hibbert
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
| | - Robert E. W. Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew Currie
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, WA, Australia
- Neonatal Infection & Immunity Team, Wesfarmers Centre of Vaccine & Infectious Diseases, Telethon Kids Institute, Perth, WA, Australia
- * E-mail:
| |
Collapse
|
33
|
Interleukin-31 interaction with inflammasome: A promising diagnostic and prognostic panel for early sepsis identification in critically ill patients. Cytokine 2020; 131:155102. [PMID: 32305780 DOI: 10.1016/j.cyto.2020.155102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/30/2020] [Accepted: 04/11/2020] [Indexed: 11/20/2022]
Abstract
PURPOSE This case-control study aimed to evaluate the ability to use a panel of IL-31, IL-1ß and NLRP3 to differentiate sepsis from systemic inflammatory response syndrome (SIRS) and to predict septic shock. METHODS Serum levels of IL-31, IL-1ß and NLRP3 were measured by ELISA in 149 participants; 38 with sepsis, 51 with SIRS, 30 with septic shock and 30 healthy controls. RESULTS Lower levels of IL-31 were found in sepsis (10.21 ± 4.34 pg/ml) compared to SIRS (16.74 ± 3.18 pg/ml) and to controls with the lowest levels detected in septic shock (6.26 ± 2.72 pg/ml). IL-1ß and NLRP3 levels were higher in sepsis (54.99 ± 14.11 pg/ml and 9.93 ± 2.38 ng/ml) compared to SIRS (27.8 ± 6.94 pg/ml and 4.86 ± 1.33 ng/ml) with the highest levels seen in septic shock (125.1 ± 32.79 pg/ml and 19.43 ± 6.48 ng/ml) respectively. IL-31 discriminated sepsis in patients showing SIRS with 80% sensitivity and 70% specificity and, identified septic shock with 78.6% sensitivity and 60.3% specificity. IL-1ß identified sepsis from SIRS with 93.3% and 83.3% specificity. NLRP3 discriminated sepsis from SIRS with 94.5% sensitivity and 93.3% specificity. And, with sensitivity 99.1% and 90.1% and specificity 98.9% and 80% IL-1ß and NLRP3 could respectively define septic shock. A panel of combined markers provided 100% sensitivity and specificity. The three biomarkers proved to be independent prognostic biomarkers. At 95% CI, IL-31 hazard ratio (HR) was 0.716, p = 0.001; IL-1β HR was 1.023, p ≤ 0.001; and NLRP3 HR was 1.114, p ≤ 0.001. Additionally, IL-1ß proved to be an independent predictor of septic shock (β = 0.355; p = 0.035). CONCLUSION The cross-relation between IL-31, IL-1ß and NLRP3 in sepsis can provide a promising diagnostic and prognostic panel.
Collapse
|
34
|
Omer M, Melo AM, Kelly L, Mac Dermott EJ, Leahy TR, Killeen O, Saugstad OD, Savani RC, Molloy EJ. Emerging Role of the NLRP3 Inflammasome and Interleukin-1β in Neonates. Neonatology 2020; 117:545-554. [PMID: 33075792 DOI: 10.1159/000507584] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/28/2020] [Indexed: 11/19/2022]
Abstract
Infection and persistent inflammation have a prominent role in the pathogenesis of brain injury and cerebral palsy, as well as other conditions associated with prematurity such as bronchopulmonary dysplasia. The NLRP3 inflammasome-interleukin (IL)-1β pathway has been extensively studied in adults and pre-clinical models, improving our understanding of innate immunity and offering an attractive therapeutic target that is already contributing to clinical management in many auto-inflammatory disorders. IL-1 blockade has transformed the course and outcome of conditions such as chronic infantile neurological, cutaneous, articular (CINCA/NOMID) syndrome. Inflammasome activation and upregulation has recently been implicated in neonatal brain and lung inflammatory disease and may be a novel therapeutic target.
Collapse
Affiliation(s)
- Murwan Omer
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland
| | - Ashanty Maggvie Melo
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland
| | - Lynne Kelly
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland
| | - Emma Jane Mac Dermott
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Timothy Ronan Leahy
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Orla Killeen
- Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland
| | - Ola Didrik Saugstad
- Department of Pediatric Research, University of Oslo, Oslo, Norway.,Ann and Robert H. Lurie Children's Hospital of Chicago, Department of Neonatology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Rashmin C Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eleanor J Molloy
- Discipline of Paediatrics, Trinity College Dublin, The University of Dublin, Dublin, Ireland, .,Children's Hospital Ireland (CHI) at Tallaght, Dublin, Ireland, .,Trinity Translational Medicine Institute, St. James Hospital, Dublin, Ireland, .,Department of Paediatrics, Coombe Women's and Infant's University Hospital, Dublin, Ireland, .,Department of Immunology, Rheumatology, and Neonatology, CHI at Crumlin, Dublin, Ireland,
| |
Collapse
|
35
|
Varljen T, Sekulovic G, Rakic O, Maksimovic N, Jekic B, Novakovic I, Damnjanovic T. Genetic variant rs16944 in IL1B gene is a risk factor for early-onset sepsis susceptibility and outcome in preterm infants. Inflamm Res 2019; 69:155-157. [PMID: 31863173 DOI: 10.1007/s00011-019-01301-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/25/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Interleukin-1-B (IL1B) is a proinflammatory cytokine that plays an important role in sepsis. The aim of this study was to evaluate the relationships between IL1B-511G/A polymorphism and susceptibility and outcome of early-onset sepsis (EOS) in preterm infants. METHODS DNA was extracted from the buccal swabs of 471 (285 with EOS and 186 control) preterm infants. Genotypes of rs16944 polymorphism were determined with real-time PCR method. RESULTS We found statistically significant higher frequency of IL1B-511AA genotype in EOS group than in control group (p = 0.012). Also, IL1B-511AA genotype is statistically significantly more frequent in patients with lethal EOS outcome (p = 0.011). CONCLUSION Genotype IL1B-511AA was associated with susceptibility to EOS and it is a significant predictor of lethal outcome in preterm infants with EOS.
Collapse
Affiliation(s)
- Tatjana Varljen
- Institute of Legal Medicine, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | | | - Nela Maksimovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Biljana Jekic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Ivana Novakovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Tatjana Damnjanovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
36
|
Kany S, Vollrath JT, Relja B. Cytokines in Inflammatory Disease. Int J Mol Sci 2019; 20:ijms20236008. [PMID: 31795299 PMCID: PMC6929211 DOI: 10.3390/ijms20236008] [Citation(s) in RCA: 1085] [Impact Index Per Article: 180.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/24/2019] [Accepted: 11/25/2019] [Indexed: 12/31/2022] Open
Abstract
This review aims to briefly discuss a short list of a broad variety of inflammatory cytokines. Numerous studies have implicated that inflammatory cytokines exert important effects with regard to various inflammatory diseases, yet the reports on their specific roles are not always consistent. They can be used as biomarkers to indicate or monitor disease or its progress, and also may serve as clinically applicable parameters for therapies. Yet, their precise role is not always clearly defined. Thus, in this review, we focus on the existing literature dealing with the biology of cytokines interleukin (IL)-6, IL-1, IL-33, tumor necrosis factor-alpha (TNF-α), IL-10, and IL-8. We will briefly focus on the correlations and role of these inflammatory mediators in the genesis of inflammatory impacts (e.g., shock, trauma, immune dysregulation, osteoporosis, and/or critical illness).
Collapse
Affiliation(s)
- Shinwan Kany
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany;
- Department of Cardiology with Emphasis on Electrophysiology, University Heart Centre, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Jan Tilmann Vollrath
- Department of Trauma, Hand and Reconstructive Surgery, Goethe University, 60590 Frankfurt, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6721395
| |
Collapse
|
37
|
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: 453] [Impact Index Per Article: 75.5] [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
|
38
|
C23, an oligopeptide derived from cold-inducible RNA-binding protein, suppresses inflammation and reduces lung injury in neonatal sepsis. J Pediatr Surg 2019; 54:2053-2060. [PMID: 30691879 PMCID: PMC6609502 DOI: 10.1016/j.jpedsurg.2018.12.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 12/04/2018] [Accepted: 12/23/2018] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Neonatal sepsis remains a leading cause of infant mortality. Cold-inducible RNA binding protein (CIRP) is an inflammatory mediator that induces TNF-α production in macrophages. C23 is a CIRP-derived peptide that blocks CIRP from binding its receptor. We therefore hypothesized that treatment with C23 reduces systemic inflammation and protects the lungs in neonatal sepsis. METHODS Sepsis was induced in C56BL/6 mouse pups (5-7 days) by intraperitoneal injection of adult cecal slurry (0.525 mg/g body weight, LD100). One hour later pups received retroorbital injection of C23 (8 mg/kg) or vehicle (normal saline). Ten hours after sepsis induction, blood and tissues were collected for analysis. RESULTS C23 treatment resulted in a 58% and 69% reduction in serum levels of proinflammatory cytokines IL-6 and IL-1β, respectively, and a 40% and 45% reduction of AST and LDH, as compared to vehicle-treated septic pups. In the lungs, C23 treatment reduced expression of cytokines IL-6 and IL-1β by 78% and 74%. In addition, the mRNA level of neutrophil chemoattractants KC and MIP-2 was reduced by 84% and 74%, respectively. These results corresponded to a reduction in histologic lung injury score. Vehicle-treated pups scored 0.49 ± 0.19, while C23 treatment reduced scores to 0.29 ± 0.12 (p < 0.05; Max = 1). Apoptosis in the lungs, measured by TUNEL assay, was also decreased by 53% with C23 treatment (p < 0.05). CONCLUSIONS Inhibition of CIRP with C23 treatment is protective in septic neonatal mice as demonstrated by reduced inflammatory markers systemically and in the lung. Therefore, C23 has promising therapeutic potential in treatment of neonatal sepsis. LEVEL OF EVIDENCE Level I.
Collapse
|
39
|
Shen Y, Yu J, Jing Y, Zhang J. MiR-106a aggravates sepsis-induced acute kidney injury by targeting THBS2 in mice model. Acta Cir Bras 2019; 34:e201900602. [PMID: 31432993 PMCID: PMC6705346 DOI: 10.1590/s0102-865020190060000002] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 05/02/2019] [Indexed: 11/21/2022] Open
Abstract
PURPOSE To investigate the role and related mechanisms of miR-106a in sepsis-induced AKI. METHODS Serum from sepsis and healthy patients was collected, sepsis mouse model was established by cecal ligation and puncture (CLP). TCMK-1 cells were treated with lipopolysaccharide (LPS) and transfected with THBS2-small interfering RNA (siTHBS2), miR-106a inhibitor, miR-106a mimics and their negative controls (NCs). The expression of miR-106a, thrombospondin 2 (THBS2), Bax, cleaved caspase-3 and Bcl-2, cell viability, relative caspase-3 activity and TNF-α, IL-1β, IL-6 content were respectively detected by quantitative real-time polymerase chain reaction (qRT-PCR), western blotting, Cell Counting Kit-8 (CCK-8) and enzyme linked immunosorbent assay (ELISA). The relationship between miR-106a and THBS2 was confirmed by dual luciferase reporter assay. RESULTS MiR-106a was up-regulated in serum of sepsis patients, CLP-induced mice models and LPS-induced TCMK-1 cells. LPS reduced cell viability and Bcl-2 expression, and increased caspase-3 activity, Bax expression, the content of TNF-α, IL-1β, IL-6. THBS2 was a target of miR-106a. The decreases of caspase-3 activity, TNF-α, IL-1β, IL-6, Bax expression and the increases of cell viability, Bcl-2 expression caused by miR-106a knockdown were reversed when THBS2 silencing in LPS-stimulated TCMK-1 cells. CONCLUSION MiR-106a aggravated LPS-induced inflammation and apoptosis of TCMK-1 cells via regulating THBS2 expression.
Collapse
Affiliation(s)
- Yezhou Shen
- Bachelor, Intensive Care Unit, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China. Conception and design of the study, acquisition of data, technical procedures, manuscript preparation and writing
| | - Jiaoyang Yu
- Master, Intensive Care Unit, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China. Technical procedures, acquisition of data
| | - Yunyan Jing
- Master, Intensive Care Unit, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China. Technical procedures, acquisition of data
| | - Jian Zhang
- Bachelor, Intensive Care Unit, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China. Statistical analysis, interpretation of data
| |
Collapse
|
40
|
Varljen T, Rakic O, Sekulovic G, Jekic B, Maksimovic N, Janevski MR, Novakovic I, Damnjanovic T. Association between Tumor Necrosis Factor-α Promoter -308 G/A Polymorphism and Early Onset Sepsis in Preterm Infants. TOHOKU J EXP MED 2019; 247:259-264. [PMID: 31006736 DOI: 10.1620/tjem.247.259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Early-onset neonatal sepsis (EOS) is diagnosed during the first 7 days of neonatal life and is the major cause of morbidity and mortality among preterm infants. Genetic predisposition may have an impact on EOS susceptibility and outcome. The aim of our study was to explore the association between TNF-α -308 G/A or IL-6 -174 G/C gene polymorphism and the susceptibility and outcome of EOS in preterm infants. The study included 471 preterm infants: 282 with EOS (151 with culture proven sepsis and 131 with clinical sepsis) and 189 without infection (control group). TNF-α -308 G/A and IL-6 -174 G/C were genotyped using Real-time RCR method. We observed significantly higher frequency of A allele of TNF-α -308 G/A polymorphism in blood culture proven EOS (p = 0.017) or clinical EOS (p = 0.025) compared with the control group. Logistic regression confirmed significant association between TNF-α -308 GA+AA genotypes and development of culture proven EOS (B = -0.718, p = 0.013) or clinical EOS (B = -0.602, p = 0.027). No significant differences in IL6 -174G/C alleles or genotypes distribution have been observed between culture proven EOS group, clinical EOS group and the control group. An association between TNF-α -308 G/A or IL-6 -174 G/C genotypes and EOS lethal outcome was not observed (p = 0.652 and p = 0.384, respectively). According to our analysis of large cohort of preterm infants with clearly defined EOS groups, the TNF-α -308 A allele may be a risk factor for the EOS occurrence.
Collapse
Affiliation(s)
- Tatjana Varljen
- Institute of Legal Medicine, Faculty of Medicine, University of Belgrade
| | | | | | - Biljana Jekic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade
| | - Nela Maksimovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade
| | | | - Ivana Novakovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade
| | | |
Collapse
|
41
|
Wu TT, Chen CC, Lin JT, Young GH, Wang HC, Chen HM. The anti-inflammatory function of adenine occurs through AMPK activation and its downstream transcriptional regulation in THP-1 cells. Biosci Biotechnol Biochem 2019; 83:2220-2229. [PMID: 31392929 DOI: 10.1080/09168451.2019.1650632] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Pathogenic bacteria induced sepsis is a risk factor for hospital mortality. Monocyte-derived inflammatory cytokines participate in the sepsis progression. The anti-inflammatory effect of adenine has been previously reported by our laboratory and others. However, the mechanism of action has different opinions and remains unclear in monocyte. Here, adenine was found to significantly inhibit the secretion of lipopolysaccharide-induced inflammatory cytokines such as TNF-α, IL-1β and IL-6 in THP-1 cells. The bioinformatic analysis results showed that the anti-inflammatory function is possibly due to the inhibition of NF-κB signaling. And this result is confirmed by using immunocytochemistry. Moreover, this effect can be suppressed by the AMPK inhibitor. Results also showed that adenine can activate AMPK and its multiple downstream targets. Data from mass spectrometry showed that adenine promotes significant elevation of intracellular AMP. Our data indicate that the anti-inflammatory mechanism of adenine may involve adenine phosphoribosyltransferase-catalyzed intracellular AMP elevation, which stimulates AMPK activation.
Collapse
Affiliation(s)
- Tzong-Ta Wu
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chin-Chen Chen
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Jiun-Tsai Lin
- Energenesis Biomedical Co. Ltd, New Taipei City, Taiwan
| | | | | | - Han-Min Chen
- Graduate Institute of Applied Science and Engineering, Fu-Jen Catholic University, New Taipei City, Taiwan.,Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
42
|
Dong L, Li H, Zhang S, Yang G. miR‑148 family members are putative biomarkers for sepsis. Mol Med Rep 2019; 19:5133-5141. [PMID: 31059023 PMCID: PMC6522910 DOI: 10.3892/mmr.2019.10174] [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: 02/06/2018] [Accepted: 10/01/2018] [Indexed: 12/19/2022] Open
Abstract
Sepsis is a type of systemic inflammatory response caused by infection. The present study aimed to identify novel targets for the treatment of sepsis. We conducted bioinformatic analysis of the microarray Gene Expression Omnibus dataset GSE12624, which includes data on 34 patients with sepsis and 36 healthy individuals without sepsis. Differentially expressed genes (DEGs) in sepsis patients were identified using Bayesian methods included in the limma package in R. Correlations among the expression values of DEGs were analyzed using the weighted gene co-expression network analysis (WGCNA) to construct a co-expression network. Subsequently, the generated co-expression network was visualized using Cytoscape 3.3 software. Additionally, a protein-protein interaction (PPI) network was constructed based on all the DEGs using STRING. Finally, the integrated regulatory network was constructed based on DEGs, microRNAs (miRNAs) and transcription factors (TFs). A total of 407 DEGs were identified in the sepsis samples, including 227 upregulated DEGs and 180 downregulated DEGs. WGCNA grouped the DEGs into 13 co-expressed modules. Additionally, MAP3K8 and RPS6KA5 in the MEyellow module were enriched in the MAPK and TNF signaling pathways. In addition, the PPI network comprised 48 nodes and 112 edges, which included the pairs MAP3K8-RPS6KA5, MAP3K8-IL10, RPS6KA5-EXOSC4 and EXOSC4-EXOSC5. Lastly, the TF-miRNA-target DEG regulatory network was constructed based on eight TFs (NF-κB), seven miRNAs (miR152, miR-148A/B), and 52 TF-miRNA-target gene triplets (17 upregulated genes, including MAP3K8, and 10 downregulated genes, including RPS6KA5). Our analysis showed that the members of the miR-148 family (miR-148A/B and miR-152) are candidate biomarkers for sepsis.
Collapse
Affiliation(s)
- Lei Dong
- Intensive Care Unit, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Hongwei Li
- Intensive Care Unit, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Shunli Zhang
- Department of Pediatrics, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| | - Guanzheng Yang
- Department of Respiratory Medicine, Jining No. 1 People's Hospital, Jining, Shandong 272011, P.R. China
| |
Collapse
|
43
|
Abstract
Sepsis, which kills over 200,000 patients and costs over $20 billion in the United States alone, presents a constant but preventable challenge in the healthcare system. Among the more challenging problems that it presents is misdiagnosis due to conflation with other inflammatory processes, as its mechanisms are identical to those of other inflammatory states. Unfortunately, current biomarker tests can only assess the severity and mortality risk of each case, whereas no single test exists that can predict sepsis prior to the onset of symptoms for the purpose of pre-emptive care and monitoring. We propose that a single test utilizing three, rather than two, biomarkers that appear most quickly in the blood and are the most specific for sepsis rather than trauma, may improve diagnostic accuracy and lead to lessened patient morbidity and mortality. Such a test would vastly improve patient outcomes and quality of life, prevent complications for sepsis survivors, and prevent hospital readmissions, saving the American healthcare system money. This review summarizes the current use of sepsis biomarkers to prognosticate morbidity and mortality, and rejects the current single-biomarker and even combination biomarker tests as non-specific and inaccurate for current patient needs/pro-inflammatory cytokines, general markers of inflammation, and proteins specific to myeloid cells (and therefore to infection) are discussed. Ultimately, the review suggests a three-biomarker test of procalcitonin (PCT), interleukin-6 (IL-6), and soluble triggering receptor expressed on myeloid cells-1 (sTREM-1) to diagnose sepsis before the onset of symptoms.
Collapse
|
44
|
Cai X, Chen Y, Xie X, Yao D, Ding C, Chen M. Astaxanthin prevents against lipopolysaccharide-induced acute lung injury and sepsis via inhibiting activation of MAPK/NF-κB. Am J Transl Res 2019; 11:1884-1894. [PMID: 30972212 PMCID: PMC6456544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 01/11/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Endotoxin-induced acute inflammatory diseases such as sepsis, mediated by excessive production of various pro-inflammatory cytokines remain the leading cause of mortality in critically ill patients. Lipopolysaccharide (LPS), the characteristic endotoxin found in the outer membrane of Gram-negative bacteria, can induce the innate immunity system and through Mitogen activated protein kinase (MAPK) and Nuclear Factor-κB (NF-κB), increase the production of inflammatory mediators. Astaxanthin (ASX), a xanthophyll carotenoid, exerts beneficial effects against oxidation, inflammation, and cancer. But poor evidence has been reported that whether it has protective effects on LPS-induced injury. This study aims to investigate the effects of ASX on LPS-induced sepsis and acute lung injury and to demonstrate its mechanisms. METHODS Mouse prime macrophage (MPM) challenged with LPS were used for in vitro pharmacological activity and mechanistic studies. Inflammatory facors (tumor necrosis factor-alpha and interleukin-6 levels) in MPM were determined. The mouse models of LPS-induced sepsis and acute lung injury administrated with or without the compound were used for in vivo studies. RESULTS Pre-treatment of MPM with ASX inhibited MAPK/NF-κB signaling pathway, and attenuated LPS-increased inflammatory factors in vitro. In animal models of LPS-induced sepsis and acute lung injury, administration of ASX significantly improved survival and protected lung injury. Subsequently, ASX was shown to suppress LPS-induced inflammatory factors increase, MAPK phosphorylation, and NF-κB activation in vivo. CONCLUSIONS ASX exerts impressively protective effects on LPS-induced injury in vitro and in vivo. Taken together, it might be used as a potential candidate for clinical sepsis.
Collapse
Affiliation(s)
- Xueding Cai
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, P. R. China
| | - Yanfan Chen
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, P. R. China
| | - Xiaona Xie
- Pulmonary Division, Zhejiang University of Traditional Chinese Medicine Affiliated with Wenzhou HospitalWenzhou 325000, Zhejiang, P. R. China
| | - Dan Yao
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, P. R. China
| | - Cheng Ding
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, P. R. China
| | - Mayun Chen
- Division of Pulmonary Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, Zhejiang, P. R. China
| |
Collapse
|
45
|
Aguayo V, Valdés Fernandez BN, Rodríguez-Valentín M, Ruiz-Jiménez C, Ramos-Benítez MJ, Méndez LB, Espino AM. Fasciola hepatica GST downregulates NF-κB pathway effectors and inflammatory cytokines while promoting survival in a mouse septic shock model. Sci Rep 2019; 9:2275. [PMID: 30783117 PMCID: PMC6381083 DOI: 10.1038/s41598-018-37652-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022] Open
Abstract
Parasitic helminths and helminth-derived molecules have demonstrated to possess powerful anti-inflammatory properties and confirmed therapeutic effects on inflammatory diseases. The helminth Fasciola hepatica has been reported to suppress specific Th1 specific immune responses induced by concurrent bacterial infections, thus demonstrating its anti-inflammatory ability in vivo. In this study, we demonstrate that native F. hepatica glutathione S-transferase (nFhGST), a major parasite excretory-secretory antigen, majorly comprised of Mu-class GST isoforms, significantly suppresses the LPS-induced TNFα and IL1β of mouse bone-marrow derived macrophages in vitro and the pro-inflammatory cytokine/chemokine storm within C57BL/6 mice exposed to lethal doses of LPS increasing their survival rate by more than 85%. Using THP1-Blue CD14 cells, a human monocyte cell line, we also demonstrate that nFhGST suppresses NF-κB activation in response to multiple TLR-ligands, including whole bacteria clinical isolates and this suppression was found to be dose-dependent and independent of the timing of exposure. Moreover, the suppressive effect of nFhGST on NF-κB activation was shown to be independent of enzyme activity or secondary structure of protein. As part of its anti-inflammatory effect nFhGST target multiple proteins of the canonic and non-canonic NF-κB signaling pathway as well as also JAK/STAT pathway. Overall, our results demonstrate the potent anti-inflammatory properties of nFhGST and its therapeutic potential as an anti-inflammatory agent.
Collapse
Affiliation(s)
- Vasti Aguayo
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | | | | | - Caleb Ruiz-Jiménez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Marcos J Ramos-Benítez
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico
| | - Loyda B Méndez
- School of Science & Technology Universidad del Este, Carolina, Puerto Rico
| | - Ana M Espino
- University of Puerto Rico, Medical Sciences Campus, Department of Microbiology, San Juan, Puerto Rico.
| |
Collapse
|
46
|
Xu F, Sang W, Li L, He X, Wang F, Wen T, Zeng N. Protective effects of ethyl acetate extracts of Rimulus Cinnamon on systemic inflammation and lung injury in endotoxin-poisoned mice. Drug Chem Toxicol 2018; 42:309-316. [PMID: 30257565 DOI: 10.1080/01480545.2018.1509987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rimulus cinnamon is the dried twig of Cinnamomum cassia Presl. It is widely used in China for the treatment of inflammatory processes, amenorrhea, and other diseases. We aimed to study the protective effects of ethyl acetate extracts of R. cinnamon (EAE) on systemic inflammation and lung injury in endotoxin-poisoned mice. EAE was administered 5 d prior to lipopolysaccharide (LPS) challenge with 15 mg/kg LPS. The administration of EAE increased the levels of interferon-γ (IFN-γ) and decreased the levels of interleukin-18 (IL-18) and tumor necrosis factor-α (TNF-α) in the serum. Additionally, EAE relieved the pathological changes in the tissues of the lungs and spleen, and significantly reduced the number of neutrophils in the lung tissues. In addition, treatment with EAE decreased the mRNA expression of the NLR family, pyrin domain-containing protein 3 (NLRP3), caspase-1, and interleukin-1β (IL-1β) in the lungs, as well as the expression of NLRP3, caspase-1 (p20), and pro-IL-1β proteins. These results demonstrated the promising anti-inflammatory effects of EAE in endotoxin-poisoned mice. Furthermore, EAE could alleviate the lung injury of endotoxin-poisoned mice by antagonizing the activation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Feng Xu
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| | - Wentao Sang
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| | - Ling Li
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| | - Xinyu He
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| | - Feng Wang
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| | - Taoqun Wen
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| | - Nan Zeng
- a Department of Pharmacology, College of Pharmacy , Chengdu University of TCM , Chengdu , Sichuan , P.R. China
| |
Collapse
|
47
|
Xu J, Lu C, Liu Z, Zhang P, Guo H, Wang T. Schizandrin B protects LPS-induced sepsis via TLR4/NF-κB/MyD88 signaling pathway. Am J Transl Res 2018; 10:1155-1163. [PMID: 29736208 PMCID: PMC5934574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 03/12/2018] [Indexed: 06/08/2023]
Abstract
Schizandrin B (Sch B) is the main component isolated from Schizandra fruit (Schisandra chinensis). While Sch B is established as having antioxidant, anti-proliferation and anti-inflammatory properties, but its activity in sepsis remains unclear. In the present study, we investigated the anti-inflammatory effects of Sch B in sepsis. Our experimental results demonstrated that Sch B inhibited production of IL-1β, TNF-α, IL-6 and HMGB1 by LPS-activated RAW264.7 cells. Moreover, Sch B suppressed expression of iNOS, reduced production of PGE2, blocked expression of MyD88 and TLR4, suppressed the activity of NF-κB and decreased phosphorylation of MAPKs in LPS-activated RAW264.7 cells. Administration of Sch B also reduced production of IL-1β and TNF-α, attenuated infiltration of inflammatory cells and tissue damage in lung, liver and kidney, and enhanced survival rate of LPS-challenged mice. Taken together, our data suggest that Sch B has anti-inflammatory properties against LPS-induced inflammation and sepsis. Sch B could protect against LPS-induced sepsis via the TLR4/NF-κB/MyD88 signaling pathway, and potentially be a novel anti-inflammatory and immunosuppressive drug candidate for treating sepsis.
Collapse
Affiliation(s)
- Jianjun Xu
- Burn Wound Center,The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Caijiao Lu
- Burn Wound Center,The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Zhengjun Liu
- Burn Wound Center,The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Peng Zhang
- Burn Wound Center,The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Hailei Guo
- Burn Wound Center,The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| | - Tingting Wang
- Department of Infectious Internal Medicine, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325000, P. R. China
| |
Collapse
|
48
|
Zuo L, Zhou L, Xu T, Li Z, Liu L, Shi Y, Kang J, Gao G, Du S, Sun Z, Zhang X. Antiseptic Activity of Ethnomedicinal Xuebijing Revealed by the Metabolomics Analysis Using UHPLC-Q-Orbitrap HRMS. Front Pharmacol 2018; 9:300. [PMID: 29651245 PMCID: PMC5884946 DOI: 10.3389/fphar.2018.00300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 03/15/2018] [Indexed: 01/06/2023] Open
Abstract
Xuebijing (XBJ) injection is an ethnomedicinal formula that has been widely used in the therapy of sepsis in China. However, the underlying theraputic mechanisms remain uninvestigated. In this research, a metabolomic method based on UHPLC-Q-Orbitrap HRMS was applied to make a holistic evaluation of XBJ on septic rats which were induced by the classical cecal ligation and puncture (CLP) operation. The plasma metabolic changes were profiled and evaluated by multivariate analytical (MVA) methods. In the results, a total of 41 differential metabolites were identified between CLP-operated group and sham-operated group, which were mainly involved in amino acid metabolism and lipid metabolism. After pathway analysis, it was finally discovered that the majority of the influenced metabolic pathways caused by sepsis mainly involved in energy metabolism, oxidative stress, and inflammation metabolism. When intervened by XBJ injection, 32 of the 41 disordered metabolites had been adjusted in reverse, which suggested that XBJ could mediate the abnormal metabolic pathways synergistically. In conclusion, the present study systematically investigated the efficacy and its underlying therapeutic mechanisms of XBJ on sepsis, while offering a new insight for the subsequent relevant exploration of other Chinese medicine at the same time.
Collapse
Affiliation(s)
- Lihua Zuo
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Lin Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Tanye Xu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zhuolun Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Liwei Liu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Yingying Shi
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Jian Kang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Guanmin Gao
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuzhang Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Zhi Sun
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
49
|
Leal YA, Álvarez-Nemegyei J, Lavadores-May AI, Girón-Carrillo JL, Cedillo-Rivera R, Velazquez JR. Cytokine profile as diagnostic and prognostic factor in neonatal sepsis. J Matern Fetal Neonatal Med 2018; 32:2830-2836. [DOI: 10.1080/14767058.2018.1449828] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Yelda A. Leal
- Cancer Population Record of Merida, Medical High Speciality Unit, Mexican Institute of Social Security (IMSS), Merida, Yucatan, Mexico
| | | | - Ana I. Lavadores-May
- Department of Clinical Neonatology, Regional Hospital 1, IMSS. Merida, Yucatan, Mexico
| | | | - Roberto Cedillo-Rivera
- Clinic and Epidemiology Interinstitutional Research Unit, Medicine Faculty, Autonomous University of Yucatan, Merida, Yucatan, Mexico
| | - Juan R. Velazquez
- Allergy and Immunogenetics Department, National Institute of Respiratory Diseases (INER), Mexico City, Mexico
| |
Collapse
|
50
|
Multiplex Cytokine Profiling Identifies Interleukin-27 as a Novel Biomarker For Neonatal Early Onset Sepsis. Shock 2018; 47:140-147. [PMID: 27648693 DOI: 10.1097/shk.0000000000000753] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Early onset sepsis (EOS) remains a major cause of mortality and morbidity in neonates, and traditional clinical markers effective for adults are less effective in these patients. This study aimed to assess the value of individual plasma biomarkers as well as biomarker combinations for predicting EOS in neonates. METHODS This prospective study included 151 neonates with suspected EOS. Plasma levels of interleukin (IL)-27, IL-6, IL-8, tumor necrosis factor (TNF)-α, heat shock protein (HSP) 70, macrophage inflammatory protein (MIP)-1α, MIP-1β, granzyme B, and matrix metalloproteinase (MMP)-8 were measured through multiplex cytokine profiling and assessed along with C-reactive protein (CRP) and procalcitonin (PCT). Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive ability of biomarkers individually and in combination. Logistic regression model was constructed to identify independent predictors of EOS. RESULTS The proven sepsis and probable sepsis groups were combined to form the infected group (n = 68), and the possible sepsis and low-risk sepsis groups were combined to form the uninfected group (n = 83). The ROC area under the curve was 0.747 for IL-27 (P <0.01). In addition, IL-6, TNF-α, HSP 70, MMP-8, PCT, and CRP were significantly predictive of EOS, whereas IL-8, granzyme B, MIP-1α, and MIP-1β were not. Both IL-27 and PCT were identified as independent predictors of EOS in the multivariate model, and the combined use of these markers showed significantly increased predictive ability for EOS. CONCLUSION Our results indicate that elevated IL-27 strongly correlates with EOS and may provide additional diagnostic value along with PCT.
Collapse
|