1
|
Bergman ZR, Kiberenge RK, Bianco RW, Beilman GJ, Brophy CM, Hocking KM, Alvis BD, Wise ES. Norepinephrine Infusion and the Central Venous Waveform in a Porcine Model of Endotoxemic Hypotension with Resuscitation: A Large Animal Study. J INVEST SURG 2025; 38:2445603. [PMID: 39761972 PMCID: PMC11709120 DOI: 10.1080/08941939.2024.2445603] [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: 09/17/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Venous waveform analysis is an emerging technique to estimate intravascular fluid status by fast Fourier transform deconvolution. Fluid status has been shown proportional to f0, the amplitude of the fundamental frequency of the waveform's cardiac wave upon deconvolution. Using a porcine model of distributive shock and fluid resuscitation, we sought to determine the influence of norepinephrine on f0 of the central venous waveform. METHODS Eight pigs were anesthetized, catheterized and treated with norepinephrine after precipitation of endotoxemic hypotension, and subsequent fluid resuscitation to mimic sepsis physiology. Hemodynamic parameters and central venous waveforms were continually transduced throughout the protocol for post-hoc analysis. Central venous waveform f0 before, during and after norepinephrine administration were determined using Fourier analysis. RESULTS Heart rate increased, while central venous pressure, pulmonary capillary wedge pressure and stroke volume decreased throughout norepinephrine administration (p < 0.05). Mean f0 at pre-norepinephrine, and doses 0.05, 0.10, 0.15, 0.20 and 0.25 mcg/kg/min, were 2.5, 1.4, 1.7, 1.7, 1.6 and 1.4 mmHg2, respectively (repeated measures ANOVA; p < 0.001). On post-hoc comparison to pre-norepinephrine, f0 at 0.05 mcg/kg/min was decreased (p = 0.04). CONCLUSIONS As the performance of f0 was previously characterized during fluid administration, these data offer novel insight into the performance of f0 during vasopressor delivery. Central venous waveform f0 is a decreased with norepinephrine, in concordance with pulmonary capillary wedge pressure. This allows contextualization of the novel, venous-derived signal f0 during vasopressor administration, a finding that must be understood prior to clinical translation.
Collapse
Affiliation(s)
- Zachary R Bergman
- Department of Surgery, University of Minnesota Twin Cities Medical School, Minneapolis, MN, USA, 420 Delaware St SE MMC 195, Minneapolis MN 55455
| | - Roy K Kiberenge
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Richard W Bianco
- Department of Surgery, University of Minnesota Twin Cities Medical School, Minneapolis, MN, USA, 420 Delaware St SE MMC 195, Minneapolis MN 55455
| | - Gregory J Beilman
- Department of Surgery, University of Minnesota Twin Cities Medical School, Minneapolis, MN, USA, 420 Delaware St SE MMC 195, Minneapolis MN 55455
| | - Colleen M Brophy
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA, 1161 21 Ave S. D-4303 MCN, Nashville TN 37232; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kyle M Hocking
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, USA, 1161 21 Ave S. D-4303 MCN, Nashville TN 37232; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Department of Biomedical Engineering, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235-1631
| | - Bret D Alvis
- Vanderbilt University Department of Biomedical Engineering, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235-1631
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN, USA, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville TN 37232
| | - Eric S Wise
- Department of Surgery, University of Minnesota Twin Cities Medical School, Minneapolis, MN, USA, 420 Delaware St SE MMC 195, Minneapolis MN 55455
| |
Collapse
|
2
|
Curran CS, Cui X, Li Y, Gamble T, Sun J, Minkove S, Livinski AA, Eichacker PQ, Torabi-Parizi P. Meta-analysis of niacin and NAD metabolite treatment in infectious disease animal studies suggests benefit but requires confirmation in clinically relevant models. Sci Rep 2025; 15:12621. [PMID: 40221506 PMCID: PMC11993703 DOI: 10.1038/s41598-025-95735-y] [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: 10/20/2024] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Disruption of nicotinamide adenine dinucleotide (NAD) biosynthesis and function during infection may impair host defenses and aggravate inflammatory and oxidative organ injury. Increasingly, studies are investigating whether niacin or NAD metabolite treatment is beneficial in infection and sepsis animal models. We examined whether this preclinical experience supports clinical trials. A systematic review of three data bases was conducted through 2/29/2024 and a meta-analysis was performed comparing niacin or NAD metabolite treatment to control in adult animal models employing microbial challenges. Fifty-six studies met inclusion criteria, with 24 published after 2019. Most studies employed mouse (n = 40 studies) or rat (n = 12) models and administered either a bacterial toxin (n = 28) or bacterial (n = 19) challenge. Four and three studies employed viral or fungal challenges respectively. Studies investigated an NAD metabolite alone (n = 44), niacin alone (n = 9), or both (n = 3), usually administered before or within 24h after challenge (n = 50). Only three and four studies included standard antimicrobial support or started treatment > 24h after challenge respectively. In similar patterns with differing animal types (p ≥ 0.06), compared to control across those studies investigating the parameter, niacin or NAD treatment decreased the odds ratio of mortality [95% confidence interval (CI)] [0.28 (0.17, 0.49)] and in blood or tissue increased antioxidant levels [standardized mean differences (95%CI)] (SMD) [3.61 (2.20,5.02)] and decreased levels of microbes [- 2.44 (- 3.34, - 1.55)], histologic and permeability organ injury scoring [- 1.62 (- 2.27, - 0.98) and - 1.31(- 1.77, - 0.86) respectively], levels of TNFα, IL-6 and IL-1β [- 2.47 (- 3.30, - 1.64), - 3.17 (- 4.74, - 1.60) and - 8.44 (- 12.4, - 4.5) respectively] and myeloperoxidase (MPO) [- 1.60 (- 2.06, - 1.14)], although with significant, primarily quantitative heterogeneity for each (I2 ≥ 53%, p < 0.01) except MPO. Treatment increased blood or tissue NAD+ levels and decreased chemical organ injury measures and oxidation markers but differently comparing species (p ≤ 0.05). Only 2 and 9 survival studies described power analyses or animal randomization respectively and no study described treatment or non-histologic outcome measure blinding. Among survival studies, Egger's analysis (p = 0.002) suggested publication bias. While suggestive, published animal studies do not yet support clinical trials testing niacin and NAD metabolite treatment for infection and sepsis. Animal studies simulating clinical conditions and with randomized, blinded designs are needed to investigate this potentially promising therapeutic approach.
Collapse
Affiliation(s)
- Colleen S Curran
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xizhong Cui
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Yan Li
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Tom Gamble
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Junfeng Sun
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Samuel Minkove
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Alicia A Livinski
- National Institutes of Health Library, Office of Research Services, Office of the Director, National Institutes of Health, Bethesda, MD, USA
| | - Peter Q Eichacker
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Parizad Torabi-Parizi
- National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA.
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bldg 10, Room 2C138, Bethesda, USA.
| |
Collapse
|
3
|
Yang W, Zhao A, Lei W, Chen J, Yan H, Li J, Yang Y, Chen Y. Pleiotropic Role of TNIK in Sepsis-Induced Cardiomyopathy. J Cell Physiol 2025; 240:e70027. [PMID: 40176540 DOI: 10.1002/jcp.70027] [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: 09/24/2024] [Revised: 01/11/2025] [Accepted: 03/21/2025] [Indexed: 04/04/2025]
Abstract
Heart failure induced by sepsis is considered one of the foremost contributors to mortality in intensive care unit (ICU) patients. However, the molecular mechanism of myocardial damage in sepsis has not been fully elucidated at present. TNF receptor-associated factor-2 and Nck-interacting protein kinase (TNIK) are members of the germinal center kinase superfamily. TNIK exhibits a pivotal role as a conserved modulator of glucose and lipid homeostasis. Here, we aimed to investigate the potential direct roles of TNIK and whether TNIK exerts anti-septic myocardial damage by regulating the NLRP3 pathway. We initially revealed that TNIK was the crucial involvement of septic myocardial injury. Subsequently, we constructed a cecal ligation and puncture (CLP) mouse model and employed LPS-induced injury in HL-1 cardiomyocytes. Our observations revealed an upregulation of TNIK levels in both CLP-injured mice and LPS-treated HL-1 cells. However, TNIK inhibitor TNIK-IN-7 or siRNA attenuated cardiomyocyte LPS injury. Especially, TNIK siRNA can significantly downregulate TNIK as well as decrease NLRP3 and IL-1β mRNA and protein levels, though the explicit molecular mechanisms of TNIK-NLRP3 in septic myocardial require further investigation. Together, our investigation presents novel evidence suggesting TNIK as a potential therapeutic target for the prevention and therapeutic intervention in sepsis-induced cardiomyopathy.
Collapse
Affiliation(s)
- Wenwen Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Aizhen Zhao
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wangrui Lei
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Junmin Chen
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
- Department of Cardiology, Affiliated Hospital, Yan'an University, Yan'an, China
| | - Huanle Yan
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Jiawen Li
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yang Yang
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
4
|
El-Naggar AE, Helmy MM, El-Gowilly SM, El-Mas MM. The Cholinergic Amelioration of Sepsis-Induced Baroreflex Dysfunction and Brainstem Inflammation Is Negated by Central Adenosine A3 Receptors. Pharmaceuticals (Basel) 2025; 18:388. [PMID: 40143165 PMCID: PMC11946792 DOI: 10.3390/ph18030388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Sepsis has been shown to depress arterial baroreceptor function, and this effect is counterbalanced by the cholinergic anti-inflammatory pathway. Considering the importance of central adenosine receptors in baroreceptor function, this study tested whether central adenosine A3 receptors (A3ARs) modulate the cholinergic-baroreflex interaction in sepsis and whether this interaction is modulated by mitogen-activated protein kinases (MAPKs) and related proinflammatory cytokines. Methods: Sepsis was induced by cecal ligation and puncture (CLP) and rats were instrumented with femoral and intracisternal (i.c.) catheters. Baroreflex sensitivity (BRS) was measured 24 h later in conscious animals using the vasoactive method, which correlates changes in blood pressure caused by i.v. phenylephrine (PE) and sodium nitroprusside (SNP) to concomitant reciprocal changes in heart rate. Results: The reduction in reflex bradycardic (BRS-PE), but not tachycardic (BRS-SNP), responses elicited by CLP was reversed by i.v. nicotine in a dose-related manner. The BRS-PE effect of nicotine was blunted following intracisternal administration of IB-MECA (A3AR agonist, 4 µg/rat). The depressant action of IB-MECA on the BRS facilitatory action of nicotine was abrogated following central inhibition of MAPK-JNK (SP 600125), PI3K (wortmannin), and TNFα (infliximab), but not MAPK-ERK (PD 98059). Additionally, the nicotine suppression of sepsis-induced upregulation of NFκB and NOX2 expression in the nucleus tractus solitarius (NTS) was negated by A3AR activation. The molecular effect of IB-MECA on NFκB expression disappeared in the presence of SP 600125, wortmannin, or infliximab. Conclusions: The central PI3K/MAPK-JNK/TNFα pathway contributes to the restraining action of A3ARs on cholinergic amelioration of sepsis-induced central neuroinflammatory responses and impairment of the baroreceptor-mediated negative chronotropism.
Collapse
Affiliation(s)
- Amany E. El-Naggar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
| | - Mai M. Helmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
| | - Sahar M. El-Gowilly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
| | - Mahmoud M. El-Mas
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria 21511, Egypt; (A.E.E.-N.); (M.M.H.); (S.M.E.-G.)
- Department of Pharmacology and Toxicology, Faculty of Medicine, College of Medicine, Kuwait University, Jabriya 46301, Kuwait
| |
Collapse
|
5
|
Hotta A, Nishimura M, Nakada D, Uchida R, Matsuura H, Torii N, Fujita N, Hamada T, Nakamoto A, Yoshikawa N. Markedly elevated blood pressure, tachycardia, and altered consciousness in patients with bacteremia during transurethral surgeries: two case reports. JA Clin Rep 2025; 11:12. [PMID: 39960556 PMCID: PMC11832996 DOI: 10.1186/s40981-025-00774-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Transurethral surgery is often accompanied by postoperative urinary tract infection. Although early detection and adequate treatment of bacteremia are required to prevent sepsis, it is usually undetectable during surgery. We report two cases with remarkable hypertension and tachycardia during transurethral surgery in which bacteremia was diagnosed by an intraoperative blood test. CASE PRESENTATION An 80-year-old man (Case 1) underwent transurethral holmium laser prostate enucleation under spinal anesthesia, and an 88-year-old woman (Case 2) underwent transurethral resection of bladder tumor under general anesthesia. Altered consciousness (Case 1) and postoperative delirium (Case 2) were noted, in addition to remarkable intraoperative hypertension and tachycardia. We administered broad-spectrum antibiotics for possible bacteremia in both cases. The patients' hemodynamics positively recovered the following day. Intraoperative blood samples revealed gram-negative bacillus. CONCLUSIONS Hypertension, tachycardia, and altered consciousness may suggest the onset of symptomatic bacteremia during transurethral surgery, and adequate treatment is required to prevent sepsis.
Collapse
Affiliation(s)
- Arisa Hotta
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan.
| | - Momoka Nishimura
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Daisuke Nakada
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Riko Uchida
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Hiroshi Matsuura
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Naoko Torii
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Naoko Fujita
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Taku Hamada
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Ai Nakamoto
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| | - Noriko Yoshikawa
- Department of Anesthesiology, Sumitomo Hospital, 5-3-20 Nakanoshima, Kita-Ku, Osaka City, Osaka, 530-0005, Japan
| |
Collapse
|
6
|
Shen Y, Dhar R, Liu J, Hong S, Tang H, Qian X. TRPM2 deficiency contributes to M2b macrophage polarization via the PI3K/AKT/CREB pathway in murine sepsis. Innate Immun 2025; 31:17534259251343377. [PMID: 40405673 DOI: 10.1177/17534259251343377] [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: 05/24/2025] Open
Abstract
BackgroundPrevious studies suggest that transient receptor potential melastatin 2 (TRPM2) plays a protective role in sepsis by enhancing bacterial clearance. This effect is mediated through the modulation of macrophage phenotypic changes, which strengthen the immune response against infection. However, the specific role and underlying mechanism of TRPM2 in macrophage polarization during sepsis remain unclear.MethodCecal ligation and puncture (CLP) was used to establish a mouse sepsis model, and bone marrow-derived macrophages (BMDMs) and peritoneal macrophages were prepared from C57BL/6 wild-type and TRPM2 knockout (trpm2-/-) mice. IPI549 was utilized as a specific inhibitor of PI3K. Macrophage polarization, bactericidal ability, and the PI3K/protein kinase B (AKT)/cyclic adenosine monophosphate response element-binding protein signaling pathway were assessed. In addition, survival rate, bacterial burden, lung wet/dry weight ratio, lung and liver injury scores, and cytokine levels were measured in CLP-induced septic mice.ResultsIn lipopolysaccharide (LPS)-stimulated BMDMs, trpm2 deficiency increased the expression of characteristic markers associated with the M2b phenotype, reduced the bactericidal ability, and activated the PI3K/AKT/CREB signaling pathway. Consequently, both trpm2-/- BMDMs and trpm2-/- mice exhibited impaired bactericidal clearance during CLP-induced sepsis. Furthermore, IPI549 attenuated TRPM2 deletion-induced M2b polarization and restored the bactericidal function of BMDMs. Notably, IPI549 preconditioning reversed the increased susceptibility of the trpm2-/- mice to sepsis. The 7-day mortality rate was 92% in trpm2-/- mice, compared to 42% in IPI549-pretreated trpm2-/- mice. Moreover, IPI549-treated mice exhibited improved lung wet/dry ratios, reduced lung and liver injury scores, reversed M2b polarization and decreased bacterial load.ConclusionThe PI3K/AKT/CREB pathway mediates the effect of TRPM2 by inhibiting M2b macrophage polarization and promoting bacterial clearance during sepsis.
Collapse
Affiliation(s)
- Yanping Shen
- Department of Anesthesiology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Ningbo, Zhejiang, China
- Department of Anesthesiology, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Rana Dhar
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jujun Liu
- Department of Anesthesiology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shenghui Hong
- Laboratory Animal Center, Zhejiang University, Hangzhou, Zhjiang, China
| | - Huifang Tang
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaowei Qian
- Department of Anesthesiology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Ningbo, Zhejiang, China
| |
Collapse
|
7
|
Obonyo NG, Raman S, Suen JY, Peters KM, Phan MD, Passmore MR, Bouquet M, Wilson ES, Hyslop K, Palmieri C, White N, Sato K, Farah SM, Gandini L, Liu K, Fior G, Heinsar S, Ijuin S, Kyun Ro S, Abbate G, Ainola C, Sato N, Lundon B, Portatadino S, Rachakonda RH, Schneider B, Harley A, See Hoe LE, Schembri MA, Li Bassi G, Fraser JF. An ovine septic shock model of live bacterial infusion. Intensive Care Med Exp 2024; 12:94. [PMID: 39467921 PMCID: PMC11519284 DOI: 10.1186/s40635-024-00684-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Escherichia coli is the most common cause of human bloodstream infections and bacterial sepsis/septic shock. However, translation of preclinical septic shock resuscitative therapies remains limited mainly due to low-fidelity of available models in mimicking clinical illness. To overcome the translational barrier, we sought to replicate sepsis complexity by creating an acutely critically-ill preclinical bacterial septic shock model undergoing active 48-h intensive care management. AIM To develop a clinically relevant large-animal (ovine) live-bacterial infusion model for septic shock. METHODS Septic shock was induced by intravenous infusion of the live antibiotic resistant extra-intestinal pathogenic E. coli sequence type 131 strain EC958 in eight anesthetised and mechanically ventilated sheep. A bacterial dose range of 2 × 105-2 × 109 cfu/mL was used for the dose optimisation phase (n = 4) and upon dose confirmation the model was developed (n = 5). Post-shock the animals underwent an early-vasopressor and volume-restriction resuscitation strategy with active haemodynamic management and monitoring over 48 h. Serial blood samples were collected for testing of pro-inflammatory (IL-6, IL-8, VEGFA) and anti-inflammatory (IL-10) cytokines and hyaluronan assay to assess endothelial integrity. Tissue samples were collected for histopathology and transmission electron microscopy. RESULTS The 2 × 107 cfu/mL bacterial dose led to a reproducible distributive shock within a pre-determined 12-h period. Five sheep were used to demonstrate consistency of the model. Bacterial infusion led to development of septic shock in all animals. The baseline mean arterial blood pressure reduced from a median of 91 mmHg (71, 102) to 50 mmHg (48, 57) (p = 0.004) and lactate levels increased from a median of 0.5 mM (0.3, 0.8) to 2.1 mM (2.0, 2.3) (p = 0.02) post-shock. The baseline median hyaluronan levels increased significantly from 25 ng/mL (18, 86) to 168 ng/mL (86, 569), p = 0.05 but not the median vasopressor dependency index which increased within 1 h of resuscitation from zero to 0.39 mmHg-1 (0.06, 5.13), p = 0.065, and. Over the 48 h, there was a significant decrease in the systemic vascular resistance index (F = 7.46, p = 0.01) and increase in the pro-inflammatory cytokines [IL-6 (F = 8.90, p = 0.02), IL-8 (F = 5.28, p = 0.03), and VEGFA (F = 6.47, p = 0.02)]. CONCLUSIONS This critically ill large-animal model was consistent in reproducing septic shock and will be applied in investigating advanced resuscitation and therapeutic interventions.
Collapse
Affiliation(s)
- Nchafatso G Obonyo
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia.
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- KEMRI-Wellcome Trust Research Programme and Initiative to Develop African Research Leaders, Kilifi, Kenya.
- Wellcome Trust Centre for Global Health Research, Imperial College London, London, UK.
| | - Sainath Raman
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Children's Intensive Care Research Program, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Paediatric Intensive Care Unit, Queensland Children's Hospital, Brisbane, QLD, Australia
- Queensland Paediatric Sepsis Program, Children's Health and Youth Network, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Jacky Y Suen
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Kate M Peters
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Minh-Duy Phan
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Margaret R Passmore
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Mahe Bouquet
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Emily S Wilson
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Kieran Hyslop
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Chiara Palmieri
- School of Veterinary Science, Faculty of Science, University of Queensland, Gatton, QLD, Australia
| | - Nicole White
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Australian Centre for Health Services Innovation and Centre for Healthcare Transformation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kei Sato
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Samia M Farah
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Lucia Gandini
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Keibun Liu
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Gabriele Fior
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Silver Heinsar
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Department of Intensive Care, North Estonia Medical Centre, Tallinn, Estonia
- Intensive Care Unit, St. Andrew's War Memorial Hospital, Brisbane, QLD, Australia
| | - Shinichi Ijuin
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Sun Kyun Ro
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Gabriella Abbate
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Carmen Ainola
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Noriko Sato
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Brooke Lundon
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Sofia Portatadino
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Reema H Rachakonda
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Bailey Schneider
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Amanda Harley
- Children's Intensive Care Research Program, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Queensland Paediatric Sepsis Program, Children's Health and Youth Network, Children's Health Queensland, Brisbane, Queensland, Australia
- Critical Care Nursing Management Team, Queensland Children's Hospital, Brisbane, QLD, Australia
- School of Nursing, Midwifery and Social Work, University of Queensland, Brisbane, QLD, Australia
| | - Louise E See Hoe
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
| | - Mark A Schembri
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia
- Intensive Care Unit, St. Andrew's War Memorial Hospital, Brisbane, QLD, Australia
| | - John F Fraser
- Critical Care Research Group, The Prince Charles Hospital, 627 Rode Road, Level 3 Clinical Sciences Building, Chermside, Brisbane, QLD, 4032, Australia.
- Institute for Molecular Bioscience (IMB), The University of Queensland, Brisbane, QLD, Australia.
- Intensive Care Unit, St. Andrew's War Memorial Hospital, Brisbane, QLD, Australia.
| |
Collapse
|
8
|
Tayabali AF, Dirieh Y, Groulx E, Elfarawi N, Di Fruscio S, Melanson K, Moteshareie H, Al-Gafari M, Navarro M, Bernatchez S, Demissie Z, Anoop V. Survival and virulence of Acinetobacter baumannii in microbial mixtures. BMC Microbiol 2024; 24:324. [PMID: 39243004 PMCID: PMC11378493 DOI: 10.1186/s12866-024-03471-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024] Open
Abstract
Acinetobacter species such as A. venetianus and A. guillouiae have been studied for various biotechnology applications, including bioremediation of recalcitrant and harmful environmental contaminants, as well as bioengineering of enzymes and diagnostic materials. Bacteria used in biotechnology are often combined with other microorganisms in mixtures to formulate efficacious commercial products. However, if the mixture contained a closely related Acinetobacter pathogen such as A. baumannii (Ab), it remains unclear whether the survival and virulence of Ab would be masked or augmented. This uncertainty poses a challenge in ensuring the safety of such biotechnology products, since Ab is one of the most significant pathogens for both hospital and community -acquired infections. This research aimed to investigate the growth and virulence of Ab within a mixture of 11 bacterial species formulated as a mock microbial mixture (MM). Growth challenges with environmental stressors (i.e., temperature, pH, sodium, iron, and antibiotics) revealed that Ab could thrive under diverse conditions except in the presence of ciprofloxacin. When cultured alone, Ab exhibited significantly more growth in the presence of almost all the environmental stressors than when it was co-incubated with the MM. During the exposure of A549 lung epithelial cells to the MM, Ab growth was stimulated compared to that in standard mammalian culture media. Cytotoxicity caused by Ab was suppressed in the presence of the MM. Lymphocytes were significantly reduced in mice exposed to Ab with or without MM via intravenous injection. The levels of the splenic cytokines IL-1α, IL-1β, MCP-1, and MIP-1α were significantly reduced 24 h after exposure to Ab + MM. This study demonstrated that the presence of the MM marginally but significantly reduced the growth and virulence of Ab, which has implications for the safety of mixtures of microorganisms for biotechnological applications. Furthermore, these findings expand our understanding of the virulence of Ab during host-pathogen interactions.
Collapse
Affiliation(s)
- Azam F Tayabali
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| | - Yasmine Dirieh
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Emma Groulx
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Nusaybah Elfarawi
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Sabrina Di Fruscio
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Kristina Melanson
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Houman Moteshareie
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Mustafa Al-Gafari
- Biotechnology Laboratory, Environmental Health Science and Research Bureau, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Martha Navarro
- Scientific Services Division, Bureau of Chemical Safety, Food Directorate, Health Products and Food Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Stéphane Bernatchez
- Biotechnology Sections 1 and 2, New Substances Assessment and Control Bureau, Safe Environments Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Zerihun Demissie
- Biotechnology Sections 1 and 2, New Substances Assessment and Control Bureau, Safe Environments Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| | - Valar Anoop
- Biotechnology Sections 1 and 2, New Substances Assessment and Control Bureau, Safe Environments Directorate, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON, K1A 0K9, Canada
| |
Collapse
|
9
|
Kalenka A, Arens P, Müllenbach RM, Weigand MA, Brune M, Fiedler-Kalenka MO. Effects of Oxiris ® Therapy on Cytokine Elimination after a LPS Infusion-An Experimental Animal Study. Int J Mol Sci 2024; 25:9283. [PMID: 39273234 PMCID: PMC11394842 DOI: 10.3390/ijms25179283] [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: 08/05/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The clinical effectiveness of Oxiris®, particularly in reducing cytokines, remains uncertain due to the limited data provided. This study explored and analyzed the application value of Oxiris® endotoxin adsorption technology in a large animal model. Pigs received an intravenous LPS infusion. Six animals were treated 2 h after the infusion with an Oxiris® hemadsorption using a pumpless extracorporeal technique for 6 h. Five animals served as controls. Cardiocirculatory parameters, hyperspectral analysis, and a panel of cytokines were measured. The lipopolysaccharide infusion induced sepsis-like inflammation with tachycardia, elevated pulmonary pressure, elevated lactate level, as well as elevated pro-inflammatory cytokines like interferon (IFN)-γ, interleukin (IL)-1β, IL-2, IL-6, IL-8, IL-12 and tumor necrosis factor alpha (TNF-α). In addition, increases of anti-inflammatory cytokines like IL-1ra and IL-10 were found. After 3 and 6 h in both groups, pro-inflammatory cytokines were significantly reduced. No differences between the intervention and the control group could be detected after 3 and 6 h for IL-1β, IL-2, IL-6, IL-8, IL-12 and TNF-α, suggesting no effect of the Oxiris® filter on the elimination of elevated cytokines with a pumpless extracorporeal hemadsorption technique. The presented large animal model may be a promising option for studying the effects of hemadsorption techniques.
Collapse
Affiliation(s)
- Armin Kalenka
- Department of Anesthesiology, Medical Faculty, University Hospital Heidelberg, University Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
- Hospital Bergstrasse, 64646 Heppenheim, Germany
| | - Philipp Arens
- Department of Anesthesiology, Medical Faculty, University Hospital Heidelberg, University Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Ralf M Müllenbach
- Department of Anesthesiology, Critical Care Medicine, Emergency Medicine and Pain Therapy, ECMO-Center, Campus Kassel of the University of Southampton, 34125 Kassel, Germany
| | - Markus A Weigand
- Department of Anesthesiology, Medical Faculty, University Hospital Heidelberg, University Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Maik Brune
- Department of Internal Medicine I and Clinical Chemistry, Medical Faculty, University Hospital Heidelberg, University Heidelberg, 69120 Heidelberg, Germany
| | - Mascha O Fiedler-Kalenka
- Department of Anesthesiology, Medical Faculty, University Hospital Heidelberg, University Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| |
Collapse
|
10
|
Kappelmayer J, Debreceni IB, Fejes Z, Nagy B. Inflammation, Sepsis, and the Coagulation System. Hamostaseologie 2024; 44:268-276. [PMID: 38354835 DOI: 10.1055/a-2202-8544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Sepsis has been a major health problem for centuries and it is still the leading cause of hospital deaths. Several studies in the past decades have identified numerous biochemical abnormalities in severe cases, and many of these studies provide evidence of the perturbation of the hemostatic system. This can result in complications, such as disseminated intravascular coagulation that can lead to multiorgan failure. Nevertheless, large clinical studies have demonstrated that the simple approach of inhibiting the coagulation processes by any means fails to provide significant improvement in the survival of septic patients. A cause of this failure could be the fact that in sepsis the major clinical problems result not primarily from the presence of the infective agent or enhanced coagulation but from the complex dysregulated systemic host response to pathogens. If this overt reaction is not fully deciphered, appropriate interference is highly unlikely and any improvement by conventional therapeutic interventions would be limited. Cellular activation in sepsis can be targeted by novel approaches like inhibition of the heterotypic cellular interactions of blood cells by targeting surface receptors or posttranscriptional control of the hemostatic system by noncoding ribonucleic acid (RNA) molecules. Stable RNA molecules can affect the expression of several proteins. Thus, it can be anticipated that modulation of microRNA production would result in a multitude of effects that may be beneficial in septic cases. Here, we highlight some of the recent diagnostic possibilities and potential novel routes of the dysregulated host response.
Collapse
Affiliation(s)
- János Kappelmayer
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ildikó Beke Debreceni
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Fejes
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Nagy
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
11
|
Yavuz A, Küçük A, Ergörün Aİ, Dursun AD, Yiğman Z, Alkan M, Arslan M. Evaluation of the efficacy of silymarin and dexmedetomidine on kidney and lung tissue in the treatment of sepsis in rats with cecal perforation. Exp Ther Med 2024; 27:242. [PMID: 38655036 PMCID: PMC11036365 DOI: 10.3892/etm.2024.12530] [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: 06/28/2023] [Accepted: 10/27/2023] [Indexed: 04/26/2024] Open
Abstract
Sepsis is a systemic inflammatory response syndrome that develops in the host against microorganisms. This response develops away from the primary infection site and results in end-organ damage. The present study aimed to investigate the protective and therapeutic effects on lung and kidney tissue of silymarin (S) and dexmedetomidine (DEX) applied 1 h before and after sepsis induced by the cecal ligation and puncture (CLP) method in rats. A total of 62 rats was randomly divided into eight groups: i) Control (n=6); ii) cecal perforation (CLP; n=8); iii) S + CLP (n=8; S + CLP; S administered 1 h before CPL); iv) CLP + S (n=8; S administered 1 h after CLP); v) DEX + CLP (n=8; D + CLP; DEX administered 1 h before CLP); vi) CLP + D (n=8; DEX administered 1 h after CLP); vii) SD + CLP (n=8; S and DEX administered 1 h before CLP) and viii) CLP + SD (n=8; S and DEX administered 1 h after CLP). After the cecum filled with stool, it was tied with 3/0 silk under the ileocecal valve and the anterior surface of the cecum was punctured twice with an 18-gauge needle. A total of 100 mg/kg silymarin and 100 µg/kg DEX were administered intraperitoneally to the treatment groups. Lung and kidney tissue samples were collected to evaluate biochemical and histopathological parameters. In the histopathological examination, all parameters indicating kidney injury; interstitial edema, peritubular capillary dilatation, vacuolization, ablation of tubular epithelium from the basement membrane, loss of brush border in the proximal tubule epithelium, cell swelling and nuclear defragmentation; were increased in the CLP compared with the control group. Silymarin administration increased kidney damage, including ablation of tubular epithelium from the basement membrane, compared with that in the CLP group. DEX significantly reduced kidney damage compared with the CLP and silymarin groups. The co-administration of DEX + silymarin decreased kidney damage, although it was not as effective as DEX-alone. To conclude, intraperitoneal DEX ameliorated injury in CLP rats. DEX + silymarin partially ameliorated injury but silymarin administration increased damage. As a result, silymarin has a negative effects with this dosage and DEX has a protective effect. In the present study, it was determined that using the two drugs together had a greater therapeutic effect than silymarin and no differences in the effects were not observed any when the application times of the agents were changed.
Collapse
Affiliation(s)
- Aydin Yavuz
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ayşegül Küçük
- Department of Physiology, Faculty of Medicine, Kutahya Health Science University, Kutahya 43020, Turkey
| | - Aydan İremnur Ergörün
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ali Doğan Dursun
- Department of Physiology, Faculty of Medicine, Atılım University, Ankara 06830, Turkey
| | - Zeynep Yiğman
- Department of Histology and Embryology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Neuroscience and Neurotechnology Center of Excellence, Gazi University, Ankara 06510, Turkey
| | - Metin Alkan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
- Life Sciences Application and Research Center, Gazi University, Ankara 06830, Turkey
- Laboratory Animal Breeding and Experimental Research Center, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
12
|
Castro-Valenzuela BE, Franco-Molina MA, Zárate-Triviño DG, Villarreal-Treviño L, Kawas JR, García-Coronado PL, Sobrevilla-Hernández G, Rodríguez-Padilla C. Antibacterial efficacy of novel bismuth-silver nanoparticles synthesis on Staphylococcus aureus and Escherichia coli infection models. Front Microbiol 2024; 15:1376669. [PMID: 38650875 PMCID: PMC11033500 DOI: 10.3389/fmicb.2024.1376669] [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: 01/25/2024] [Accepted: 02/29/2024] [Indexed: 04/25/2024] Open
Abstract
Introduction The emergence of multi-drug-resistant bacteria is one of the main concerns in the health sector worldwide. The conventional strategies for treatment and prophylaxis against microbial infections include the use of antibiotics. However, these drugs are failing due to the increasing antimicrobial resistance. The unavailability of effective antibiotics highlights the need to discover effective alternatives to combat bacterial infections. One option is the use of metallic nanoparticles, which are toxic to some microorganisms due to their nanometric size. Methods In this study we (1) synthesize and characterize bismuth and silver nanoparticles, (2) evaluate the antibacterial activity of NPs against Staphylococcus aureus and Escherichia coli in several infection models (in vivo models: infected wound and sepsis and in vitro model: mastitis), and we (3) determine the cytotoxic effect on several cell lines representative of the skin tissue. Results and discussion We obtained bimetallic nanoparticles of bismuth and silver in a stable aqueous solution from a single reaction by chemical synthesis. These nanoparticles show antibacterial activity on S. aureus and E. coli in vitro without cytotoxic effects on fibroblast, endothelial vascular, and mammary epithelium cell lines. In an infected-wound mice model, antibacterial effect was observed, without effect on in vitro mastitis and sepsis models.
Collapse
Affiliation(s)
- Beatriz Elena Castro-Valenzuela
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Moisés Armides Franco-Molina
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Diana Ginette Zárate-Triviño
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Licet Villarreal-Treviño
- Posgrado en Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Jorge R. Kawas
- Posgrado Conjunto Agronomía-Veterinaria, Universidad Autónoma de Nuevo León, General Escobedo, Nuevo León, Mexico
| | - Paola Leonor García-Coronado
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Gustavo Sobrevilla-Hernández
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| | - Cristina Rodríguez-Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Nuevo León, Mexico
| |
Collapse
|
13
|
Wang X, Du C, Subramanian S, Turner L, Geng H, Bu HF, Tan XD. Severe gut mucosal injury induces profound systemic inflammation and spleen-associated lymphoid organ response. Front Immunol 2024; 14:1340442. [PMID: 38259439 PMCID: PMC10800855 DOI: 10.3389/fimmu.2023.1340442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 12/08/2023] [Indexed: 01/24/2024] Open
Abstract
Clinical evidence indicates a connection between gut injuries, infections, inflammation, and an increased susceptibility to systemic inflammation. Nevertheless, the animal models designed to replicate this progression are inadequate, and the fundamental mechanisms are still largely unknown. This research explores the relationship between gut injuries and systemic inflammation using a Dextran Sulfate Sodium (DSS)-induced colonic mucosal injury mouse model. Continuous treatment of adult mice with 4% DSS drinking water yielded a remarkable mortality rate by day 7, alongside intensified gut injury and detectable peripheral inflammation. Moreover, RNAscope in situ hybridization with 16S rRNA probe noted bacterial penetration into deeper colon compartments of the mice following treatment with DSS for 7 days. Histological analysis revealed inflammation in the liver and lung tissues of DSS-treated mice. In addition, we found that DSS-treated mice exhibited elevation of Alanine transaminase (ALT) and Aspartate transaminase (AST) in peripheral blood and pro-inflammatory cytokine levels in the liver. Notably, the DSS-treated mice displayed a dampened metabolic profile, reduced CD45 marker expression, and an increase in apoptosis within the lymphoid organ such as spleen. These findings suggest that high-dose DSS-induced gut injury gives rise to sepsis-like systemic inflammation characterized by multiple organ injury and profound splenocyte apoptosis and dysfunction of CD45+ cells in the spleen, indicating the role of the spleen in the pathogenesis of gut-derived systemic inflammation. Together, the severe colonic mucosal injury model facilitates research into gut damage and associated peripheral immune responses, providing a vital framework for investigating mechanisms related to clinically relevant, gut-derived systemic inflammation.
Collapse
Affiliation(s)
- Xiao Wang
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Chao Du
- Department of Pediatrics, Ann and Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Saravanan Subramanian
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Lucas Turner
- Department of Pediatrics, Ann and Robert H. Lurie Children’s Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hua Geng
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Heng-Fu Bu
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Xiao-Di Tan
- Pediatric Mucosal Inflammation and Regeneration Research Program, Center for Pediatric Translational Research and Education, Department of Pediatrics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
14
|
Kim SY, Kang J, Fawaz MV, Yu M, Xia Z, Morin EE, Mei L, Olsen K, Li XA, Schwendeman A. Phospholipids impact the protective effects of HDL-mimetic nanodiscs against lipopolysaccharide-induced inflammation. Nanomedicine (Lond) 2023; 18:2127-2142. [PMID: 38197376 PMCID: PMC10918510 DOI: 10.2217/nnm-2023-0222] [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: 08/08/2023] [Accepted: 11/01/2023] [Indexed: 01/11/2024] Open
Abstract
Aim: The impacts of synthetic high-density lipoprotein (sHDL) phospholipid components on anti-sepsis effects were investigated. Methods: sHDL composed with ApoA-I mimetic peptide (22A) and different phosphatidylcholines were prepared and characterized. Anti-inflammatory effects were investigated in vitro and in vivo on lipopolysaccharide (LPS)-induced inflammation models. Results: sHDLs composed with 1,2-dimyristoyl-sn-glycero-3-phosphocholine (22A-DMPC) most effectively neutralizes LPS, inhibits toll-like receptor 4 recruitment into lipid rafts, suppresses nuclear factor κB signaling and promotes activating transcription factor 3 activating. The lethal endotoxemia animal model showed the protective effects of 22A-DMPC. Conclusion: Phospholipid components affect the stability and fluidity of nanodiscs, impacting the anti-septic efficacy of sHDLs. 22A-DMPC presents the strongest LPS binding and anti-inflammatory effects in vitro and in vivo, suggesting a potential sepsis treatment.
Collapse
Affiliation(s)
- Sang Yeop Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jukyung Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| | - Maria V Fawaz
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| | - Ziyun Xia
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| | - Emily E Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| | - Ling Mei
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| | - Karl Olsen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
| | - Xiang-An Li
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI 48105, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
15
|
Liu H, Pan D, Li P, Wang D, Xia B, Zhang R, Lu J, Xing X, Du J, Zhang X, Jin L, Jiang L, Yao L, Li M, Wu J. Loss of ZBED6 Protects Against Sepsis-Induced Muscle Atrophy by Upregulating DOCK3-Mediated RAC1/PI3K/AKT Signaling Pathway in Pigs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302298. [PMID: 37551034 PMCID: PMC10582467 DOI: 10.1002/advs.202302298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/12/2023] [Indexed: 08/09/2023]
Abstract
Sepsis-induced muscle atrophy often increases morbidity and mortality in intensive care unit (ICU) patients, yet neither therapeutic target nor optimal animal model is available for this disease. Here, by modifying the surgical strategy of cecal ligation and puncture (CLP), a novel sepsis pig model is created that for the first time recapitulates the whole course of sepsis in humans. With this model and sepsis patients, increased levels of the transcription factor zinc finger BED-type containing 6 (ZBED6) in skeletal muscle are shown. Protection against sepsis-induced muscle wasting in ZBED6-deficient pigs is further demonstrated. Mechanistically, integrated analysis of RNA-seq and ChIP-seq reveals dedicator of cytokinesis 3 (DOCK3) as the direct target of ZBED6. In septic ZBED6-deficient pigs, DOCK3 expression is increased in skeletal muscle and myocytes, activating the RAC1/PI3K/AKT pathway and protecting against sepsis-induced muscle wasting. Conversely, opposite gene expression patterns and exacerbated muscle wasting are observed in septic ZBED6-overexpressing myotubes. Notably, sepsis patients show increased ZBED6 expression along with reduced DOCK3 and downregulated RAC1/PI3K/AKT pathway. These findings suggest that ZBED6 is a potential therapeutic target for sepsis-induced muscle atrophy, and the established sepsis pig model is a valuable tool for understanding sepsis pathogenesis and developing its therapeutics.
Collapse
Affiliation(s)
- Huan Liu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Dengke Pan
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalChengduSichuan610072China
| | - Pu Li
- Department of Critical Care Medicinethe Second Affiliated Hospital of Air Force Medical UniversityNo.569, Xinsi RoadXi'anShaanxi710038China
| | - Dandan Wang
- Laboratory of Animal (Poultry) Genetics Breeding and ReproductionMinistry of AgricultureInstitute of Animal SciencesChinese Academy of Agricultural Sciences (CAAS)Beijing100193China
| | - Bo Xia
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Ruixin Zhang
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Junfeng Lu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Xiangyang Xing
- Chengdu Clonorgan Biotechnology Co. LTDChengduSichuan610041China
| | - Jiaxiang Du
- Chengdu Clonorgan Biotechnology Co. LTDChengduSichuan610041China
| | - Xiao Zhang
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| | - Long Jin
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengduSichuan611130China
| | - Lin Jiang
- Laboratory of Animal (Poultry) Genetics Breeding and ReproductionMinistry of AgricultureInstitute of Animal SciencesChinese Academy of Agricultural Sciences (CAAS)Beijing100193China
| | - Linong Yao
- Department of Critical Care Medicinethe Second Affiliated Hospital of Air Force Medical UniversityNo.569, Xinsi RoadXi'anShaanxi710038China
| | - Mingzhou Li
- Institute of Animal Genetics and BreedingCollege of Animal Science and TechnologySichuan Agricultural UniversityChengduSichuan611130China
| | - Jiangwei Wu
- Key Laboratory of Animal GeneticsBreeding and Reproduction of Shaanxi ProvinceCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingShaanxi712100China
| |
Collapse
|
16
|
Marendy D, Gabor L, George SD, Parker A, Doyle E. Plasma lipopolysaccharide elevations in cattle associated with early-stage infection by Fasciola hepatica. Aust Vet J 2023; 101:334-338. [PMID: 37317661 DOI: 10.1111/avj.13264] [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: 09/04/2022] [Revised: 04/26/2023] [Accepted: 05/27/2023] [Indexed: 06/16/2023]
Abstract
Fasciolosis is an endemic zoonotic parasitic disease with significant impacts on human health and both animal health and production. Early post-infection impacts on the host remain unclear. The objective of this study was to determine the changes, if any, to levels of endotoxin in cattle plasma in response to early-stage infection with Fasciola hepatica. Thirty-six (36) commercial bred cattle were experimentally infected with approximately 400 viable metacercariae. Plasma lipopolysaccharide (endotoxin) levels were examined on 24 occasions from 0 h before infection to 336 h after infection using the Limulus Amoebocyte Lysate chromogenic end point assay and compared with that of six (6) uninfected control animals. Peak lipopolysaccharide levels in infected animals were reached at 52 h after infection and returned to pre-infection levels at time 144 h after infection. Infected animals had significantly elevated lipopolysaccharide levels between 24 and 120 h after infection when compared to uninfected animals. The mean change in endotoxin units (EU)/mL over time after infection was statistically significant in infected animals. Elevations of lipopolysaccharide occurred in all infected animals suggesting a possible repeatable and titratable endotoxemia conducive to therapeutic agent model development.
Collapse
Affiliation(s)
- D Marendy
- School of Environmental and Rural Science, University of New England, Armidale, New South Wales, Australia
| | - L Gabor
- Yarrandoo R&D Station, Elanco Australasia, Kemps Creek, New South Wales, Australia
| | - S D George
- Yarrandoo R&D Station, Elanco Australasia, Kemps Creek, New South Wales, Australia
| | - A Parker
- Yarrandoo R&D Station, Elanco Australasia, Kemps Creek, New South Wales, Australia
| | - E Doyle
- School of Environmental and Rural Science, University of New England, Armidale, New South Wales, Australia
| |
Collapse
|
17
|
Bahnasawy SM, Skorup P, Hanslin K, Lipcsey M, Friberg LE, Nielsen EI. Predicting cytokine kinetics during sepsis; a modelling framework from a porcine sepsis model with live Escherichia coli. Cytokine 2023; 169:156296. [PMID: 37467709 DOI: 10.1016/j.cyto.2023.156296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Describing the kinetics of cytokines involved as biomarkers of sepsis progression could help to optimise interventions in septic patients. This work aimed to quantitively characterise the cytokine kinetics upon exposure to live E. coli by developing an in silico model, and to explore predicted cytokine kinetics at different bacterial exposure scenarios. METHODS Data from published in vivo studies using a porcine sepsis model were analysed. A model describing the time courses of bacterial dynamics, endotoxin (ETX) release, and the kinetics of TNF and IL-6 was developed. The model structure was extended from a published model that quantifies the ETX-cytokines relationship. An external model evaluation was conducted by applying the model to literature data. Model simulations were performed to explore the sensitivity of the host response towards differences in the input rate of bacteria, while keeping the total bacterial burden constant. RESULTS The analysis included 645 observations from 30 animals. The blood bacterial count was well described by a one-compartment model with linear elimination. A scaling factor was estimated to quantify the ETX release by bacteria. The model successfully described the profiles of TNF, and IL-6 without a need to modify the ETX-cytokines model structure. The kinetics of TNF, and IL-6 in the external datasets were well predicted. According to the simulations, the ETX tolerance development results in that low initial input rates of bacteria trigger the lowest cytokine release. CONCLUSION The model quantitively described and predicted the cytokine kinetics triggered by E. coli exposure. The host response was found to be sensitive to the bacterial exposure rate given the same total bacterial burden.
Collapse
Affiliation(s)
| | - Paul Skorup
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Katja Hanslin
- Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Miklós Lipcsey
- Hedenstierna laboratory, Anesthesiology & Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
18
|
Jiao Y, Wai Tong CS, Rainer TH. An appraisal of studies using mouse models to assist the biomarker discovery for sepsis prognosis. Heliyon 2023; 9:e17770. [PMID: 37456011 PMCID: PMC10344760 DOI: 10.1016/j.heliyon.2023.e17770] [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: 08/24/2022] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Clinicians need reliable outcome predictors to improve the prognosis of septic patients. Mouse models are widely used in sepsis research. We aimed to review how mouse models were used to search for novel prognostic biomarkers of sepsis in order to optimize their use for future biomarker discovery. Methods We searched PubMed from 2012 to July 2022 using "((sepsis) AND (mice)) AND ((prognosis) OR (prognostic biomarker))". Results A total of 412 publications were retrieved. We selected those studies in which mouse sepsis was used to demonstrate prognostic potential of biomarker candidates and/or assist the subsequent evaluation in human sepsis for further appraisal. The most frequent models were lipopolysaccharide (LPS) injection and caecal ligation and puncture (CLP) using young male mice. Discovery technologies applied on mice include setting survival and nonsurvivable groups, detecting changes of biomarker levels and measuring physiological parameters during sepsis. None of the biomarkers achieved sufficient clinical performance for clinical use. Conclusions The number of studies and strategies using mouse models to discover prognostic biomarkers of sepsis are limited. Current mouse models need to be further optimized to better conform to human sepsis. Current biomarker platforms do not achieve predictive performance for clinical use.
Collapse
|
19
|
Cai L, Rodgers E, Schoenmann N, Raju RP. Advances in Rodent Experimental Models of Sepsis. Int J Mol Sci 2023; 24:9578. [PMID: 37298529 PMCID: PMC10253762 DOI: 10.3390/ijms24119578] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/09/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
In the development of therapeutic strategies for human diseases, preclinical experimental models have a key role. However, the preclinical immunomodulatory therapies developed using rodent sepsis were not successful in human clinical trials. Sepsis is characterized by a dysregulated inflammation and redox imbalance triggered by infection. Human sepsis is simulated in experimental models using methods that trigger inflammation or infection in the host animals, most often mice or rats. It remains unknown whether the characteristics of the host species, the methods used to induce sepsis, or the molecular processes focused upon need to be revisited in the development of treatment methods that will succeed in human clinical trials. Our goal in this review is to provide a survey of existing experimental models of sepsis, including the use of humanized mice and dirty mice, and to show how these models reflect the clinical course of sepsis. We will discuss the strengths and limitations of these models and present recent advances in this subject area. We maintain that rodent models continue to have an irreplaceable role in studies toward discovering treatment methods for human sepsis.
Collapse
Affiliation(s)
- Lun Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Elizabeth Rodgers
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Nick Schoenmann
- Department of Emergency Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Raghavan Pillai Raju
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
20
|
Song YQ, Lin WJ, Hu HJ, Wu SH, Jing L, Lu Q, Zhu W. Sodium tanshinone IIA sulfonate attenuates sepsis-associated brain injury via inhibiting NOD-like receptor 3/caspase-1/gasdermin D-mediated pyroptosis. Int Immunopharmacol 2023; 118:110111. [PMID: 37028275 DOI: 10.1016/j.intimp.2023.110111] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/19/2023] [Accepted: 03/26/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Sodium tanshinone IIA sulfonate (STS) has been reported to protect organ function in sepsis. However, the attenuation of sepsis-associated brain injury and its underlying mechanisms by STS has not been established. METHODS C57BL/6 mice were used to establish the cecal ligation perforation (CLP) model, and STS was injected intraperitoneally 30 min before the surgery. The BV2 cells were stimulated by lipopolysaccharide after being pre-treated with STS for 4 h. The STS protective effects against brain injury and in vivo anti-neuroinflammatory effects were investigated using the 48-hour survival rate and body weight changes, brain water content, histopathological staining, immunohistochemistry, ELISA, RT-qPCR, and transmission electron microscopy. The pro-inflammatory cytokines of BV2 cells were detected by ELISA and RT-qPCR. At last, the levels of NOD-like receptor 3 (NLRP3) inflammasome activation and pyroptosis in brain tissues of the CLP model and BV2 cells were detected using western blotting. RESULTS STS increased the survival rate, decreased brain water content, and improved brain pathological damage in the CLP models. STS increased the expressions of tight junction proteins ZO-1 and Claudin5 while reducing the expressions of tumor necrosis factor α (TNF-α), interleukin-1β(IL-1β), and interleukin-18 (IL-18) in the brain tissues of the CLP models. Meanwhile, STS inhibited microglial activation and M1-type polarization in vitro and in vivo. The NLRP3/caspase-1/ gasdermin D (GSDMD)-mediated pyroptosis was activated in the brain tissues of the CLP models and lipopolysaccharide (LPS)-treated BV2 cells, which was significantly inhibited by STS. CONCLUSIONS The activation of NLRP3/caspase-1/GSDMD-mediated pyroptosis and subsequent secretion of proinflammatory cytokines may be the underlying mechanisms of STS against sepsis-associated brain injury and neuroinflammatory response.
Collapse
Affiliation(s)
- Ya-Qin Song
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei-Ji Lin
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Jie Hu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shu-Hui Wu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liang Jing
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qing Lu
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Zhu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Intensive Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
21
|
Cazanga V, Palma C, Casanova T, Rojas D, Barrera K, Valenzuela C, Acevedo A, Ascui-Gac G, Pérez-Jeldres T, Pérez-Fernández R. Modulation of the Acute Inflammatory Response Induced by the Escherichia coli Lipopolysaccharide through the Interaction of Pentoxifylline and Florfenicol in a Rabbit Model. Antibiotics (Basel) 2023; 12:antibiotics12040639. [PMID: 37107001 PMCID: PMC10135147 DOI: 10.3390/antibiotics12040639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/12/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Experimental reports have demonstrated that florfenicol (FFC) exerts potent anti-inflammatory effects, improving survival in a murine endotoxemia model. Considering the anti-inflammatory and immunomodulatory properties of pentoxifylline (PTX) as an adjuvant to enhance the efficacy of antibiotics, the anti-inflammatory effects of the interaction FFC/PTX over the E. coli Lipopolysaccharide (LPS)-induced acute inflammatory response was evaluated in rabbits. METHODS Twenty-five clinically healthy New Zealand rabbits (3.8 ± 0.2 kg body weight: bw), were distributed into five experimental groups. Group 1 (control): treated with 1 mL/4 kg bw of 0.9% saline solution (SS) intravenously (IV). Group 2 (LPS): treated with an IV dose of 5 µg/kg of LPS. Group 3 (pentoxifylline (PTX) + LPS): treated with an oral dose of 30 mg/kg PTX, followed by an IV dose of 5 µg/kg of LPS 45 min after PTX. Group 4 (Florfenicol (FFC) + LPS): treated with an IM dose of 20 mg/kg of FFC, followed by an IV dose of 5 µg/kg of LPS 45 min after FFC administration. Group 5 (PTX + FFC + LPS): treated with an oral dose of 30 mg/kg of PTX, followed by an IM dose of 20 mg/kg of FFC, and, 45 min after an IV dose of 5 µg/kg of LPS was administered. The anti-inflammatory response was evaluated through changes in plasma levels of interleukins (TNF-α, IL-1β and IL-6), C-reactive protein (CRP), and body temperature. RESULTS It has been shown that each drug produced a partial inhibition over the LPS-induced increase in TNF-α, IL-1β, and CRP. When both drugs were co-administered, a synergistic inhibitory effect on the IL-1β and CRP plasma concentrations was observed, associated with a synergic antipyretic effect. However, the co-administration of PTX/FFC failed to modify the LPS-induced increase in the TNF-α plasma concentrations. CONCLUSIONS We concluded that the combination of FFC and PTX in our LPS sepsis models demonstrates immunomodulatory effects. An apparent synergistic effect was observed for the IL-1β inhibition, which peaks at three hours and then decreases. At the same time, each drug alone was superior in reducing TNF-α levels, while the combination was inferior. However, the peak of TNF-α in this sepsis model was at 12 h. Therefore, in rabbits plasma IL-1β and TNF-α could be regulated independently, thus, further research is needed to explore the effects of this combination over a more prolonged period.
Collapse
Affiliation(s)
- Victoria Cazanga
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Cristina Palma
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Tomás Casanova
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Daniela Rojas
- Veterinary Pathology Laboratory, Department of Pathology and Preventive Medicine, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Karin Barrera
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Cristhian Valenzuela
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Aracelly Acevedo
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| | - Gabriel Ascui-Gac
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, California University, San Diego, CA 92182, USA
| | - Tamara Pérez-Jeldres
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Rubén Pérez-Fernández
- Pharmacology Laboratory, Department of Clinical Sciences, Faculty of Veterinary Sciences, Universidad de Concepción, Chillan 3820572, Chile
| |
Collapse
|
22
|
Ashino T, Nakamura Y, Ohtaki H, Iwakura Y, Numazawa S. Downregulation of the gene expression of Cyp2c29 and Cyp3a11 by cecal ligation and puncture-induced sepsis is associated with interleukin-6. Int Immunopharmacol 2023; 117:110039. [PMID: 36944277 DOI: 10.1016/j.intimp.2023.110039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/07/2023] [Accepted: 03/12/2023] [Indexed: 03/22/2023]
Abstract
Sepsis is a pathological condition that affects the metabolism of administered drugs, leading to changes in the duration and intensity of their intended efficacies. Proinflammatory cytokines downregulate the expression of cytochrome P450s (P450s). The effects of P450 expression under inflammatory conditions have been studied using prophlogistic substances such as lipopolysaccharide; however, few studies have focused on clinical models of sepsis. Here, we show that cecal ligation and puncture (CLP), an approach for the study of human polymicrobial sepsis, leads to the expression of interleukin-1β (IL-1β), IL-6, and tumor necrosis factor α (TNFα) at 24 h after the CLP operation. Following CLP, IL-6-/- mice exhibited markedly lower survival than WT mice. In addition, CLP led to the significant downregulation of Cyp2c29 and Cyp3a11 gene expression in IL-1α-/-/β-/- (IL-1-/-) and TNFα-/- mice as well as in WT mice. In contrast, CLP elicited no significant effect on Cyp3a11 expression in IL-6-/- mice. Although CLP reduced the Cyp2c29 expression level in IL-6-/- mice, the expression of Cyp2c29 was lower in CLP-operated WT mice than in CLP-operated IL-6-/- mice. The reduction in the respective P450 protein levels and activities due to CLP-induced sepsis, reflected in the mRNA expression levels, was abolished by IL-6 depletion. Thus, CLP-induced sepsis downregulates P450 gene expression, particularly Cyp2c expression, and this effect is associated with IL-6 without affecting resistance to CLP-induced sepsis. These findings demonstrate the usefulness of CLP for studying the regulation of P450s and highlight IL-6 as a potential indicator of drug-metabolizing capacity under septic conditions.
Collapse
Affiliation(s)
- Takashi Ashino
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Yuki Nakamura
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Center for Pharmaceutical Education, Faculty of Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama, Kanagawa 245-0066, Japan
| | - Hirokazu Ohtaki
- Department of Functional Neurobiology, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan
| | - Satoshi Numazawa
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| |
Collapse
|
23
|
Nanospanlastics as a Novel Approach for Improving the Oral Delivery of Resveratrol in Lipopolysaccharide-Induced Endotoxicity in Mice. J Pharm Innov 2023. [DOI: 10.1007/s12247-023-09711-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
Abstract
Abstract
Purpose
Resveratrol (RSV) is a natural polyphenolic compound that has numerous biological effects. Owing to its poor bioavailability, only trace concentrations of RSV could be found at the site of action. Therefore, the present study was aimed at developing RSV-loaded nanospanlastics to improve its oral delivery and therapeutic activity.
Methods
RSV-loaded nanospanlastics were prepared using the thin film hydration technique. The developed formulations were characterized via vesicular size (VS), polydispersity index (PDI), zeta potential (ZP) measurements, fourier transform infrared (FT-IR) spectroscopy analysis and transmission electron microscopy (TEM). In vitro release profile was carried out using dialysis bag diffusion technique. In vivo study was carried out using lipopolysaccharide (LPS)-induced endotoxicity model in mice to evaluate the formulations activity.
Results
The results revealed the successful development of RSV-loaded nanospanlastics which exhibited EE% ranging from 45 to 85%, particle sizes ranging from 260.5 to 794.3 nm; negatively charged zeta potential (≤ − 20 mV) and TEM revealed their spherical shape. An in vitro release study showed biphasic pattern with sustained release of drug up to 24 h. In vivo results showed the superiority of RSV-loaded nanospanlastics over conventional niosomes in attenuating serum levels of liver and kidney functions (aspartate transaminase (AST), alanine transaminase (ALT), and creatinine) in LPS-induced endotoxic mice. Furthermore, both of them suppressed the elevated oxidative stress and inflammatory markers (malondialdehyde (MDA), nitric oxide (NO), and interleukin-1beta (IL-1β)) estimated in the liver and kidney tissues. However, the nanospanlastics showed a prevalence effect over conventional niosomes in kidney measurements and the histopathological examinations.
Conclusions
These findings reveal the potential of nanospanlastics in improving the oral delivery and therapeutic efficacy of RSV.
Collapse
|
24
|
Dumbuya JS, Li S, Liang L, Zeng Q. Paediatric sepsis-associated encephalopathy (SAE): a comprehensive review. Mol Med 2023; 29:27. [PMID: 36823611 PMCID: PMC9951490 DOI: 10.1186/s10020-023-00621-w] [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: 10/31/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Sepsis-associated encephalopathy (SAE) is one of the most common types of organ dysfunction without overt central nervous system (CNS) infection. It is associated with higher mortality, low quality of life, and long-term neurological sequelae, its mortality in patients diagnosed with sepsis, progressing to SAE, is 9% to 76%. The pathophysiology of SAE is still unknown, but its mechanisms are well elaborated, including oxidative stress, increased cytokines and proinflammatory factors levels, disturbances in the cerebral circulation, changes in blood-brain barrier permeability, injury to the brain's vascular endothelium, altered levels of neurotransmitters, changes in amino acid levels, dysfunction of cerebral microvascular cells, mitochondria dysfunction, activation of microglia and astrocytes, and neuronal death. The diagnosis of SAE involves excluding direct CNS infection or other types of encephalopathies, which might hinder its early detection and appropriate implementation of management protocols, especially in paediatric patients where only a few cases have been reported in the literature. The most commonly applied diagnostic tools include electroencephalography, neurological imaging, and biomarker detection. SAE treatment mainly focuses on managing underlying conditions and using antibiotics and supportive therapy. In contrast, sedative medication is used judiciously to treat those showing features such as agitation. The most widely used medication is dexmedetomidine which is neuroprotective by inhibiting neuronal apoptosis and reducing a sepsis-associated inflammatory response, resulting in improved short-term mortality and shorter time on a ventilator. Other agents, such as dexamethasone, melatonin, and magnesium, are also being explored in vivo and ex vivo with encouraging results. Managing modifiable factors associated with SAE is crucial in improving generalised neurological outcomes. From those mentioned above, there are still only a few experimentation models of paediatric SAE and its treatment strategies. Extrapolation of adult SAE models is challenging because of the evolving brain and technical complexity of the model being investigated. Here, we reviewed the current understanding of paediatric SAE, its pathophysiological mechanisms, diagnostic methods, therapeutic interventions, and potential emerging neuroprotective agents.
Collapse
Affiliation(s)
- John Sieh Dumbuya
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Siqi Li
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Lili Liang
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Qiyi Zeng
- Department of Paediatrics, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
25
|
Yamamoto T, Kurata M, Kaneko N, Masumoto J. Intestinal edema induced by LPS-induced endotoxemia is associated with an inflammasome adaptor ASC. PLoS One 2023; 18:e0281746. [PMID: 36800329 PMCID: PMC9937502 DOI: 10.1371/journal.pone.0281746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
The apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)/caspase-1/interleukin(IL)-1β axis, also known as the inflammasome pathway, is indispensable for IL-1β activation in response to various pathogens or own damages. Previously, we developed an NLRP3-inflammasome using a cell-free system and identified ASC targeting drugs; thus, examination of ASC-related histopathology in various diseases could help to provide indications for these drugs. Here, we generated mice deficient only in ASC-protein (ASC-deficient (AD) mice) using CRISPR/Cas9 technology, studied which tissues were most affected, and obtained histopathological images of lipopolysaccharide (LPS)-induced endotoxemia. C57BL/6 wild-type (WT) and (AD) mice were injected intraperitoneally with a lethal dose (50 μg/g) of LPS. Statistical analysis of the survival of C57BL/6 mice and AD mice was performed using the Kaplan-Meier method and the log-rank test. The histopathological findings of multiple tissues from these mice were compared. Acute inflammation (e.g., catarrhal inflammation), along with congestion was observed in the colon of WT mice but not in that of AD mice. Adhesion of neutrophils to capillaries, along with interstitial infiltration, were observed in multiple tissues from WT mice. In AD mice, neutrophil infiltration was less severe but remained evident in the stomach, small intestine, heart, liver, kidney, spleen, and brain. Notably, there was no difference between WT and AD mice with respect to alveolar neutrophil infiltration and interstitial edema. These findings suggest that even though ASC contributes to systemic inflammation, it is dependent on the tissue involved. Intestinal congestion and edema might be good candidates for anti-ASC-targeted therapy.
Collapse
Affiliation(s)
- Toshihiro Yamamoto
- Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Mie Kurata
- Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Naoe Kaneko
- Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
| | - Junya Masumoto
- Department of Pathology, Proteo-Science Center and Graduate School of Medicine, Ehime University, Toon, Ehime, Japan
- * E-mail:
| |
Collapse
|
26
|
Inhibition of SHP2 by the Small Molecule Drug SHP099 Prevents Lipopolysaccharide-Induced Acute Lung Injury in Mice. Inflammation 2023; 46:975-986. [PMID: 36732395 DOI: 10.1007/s10753-023-01784-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/05/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Excessive pulmonary inflammation in acute lung injury (ALI) causes high patient mortality. Anti-inflammatory therapy, combined with infection resistance, can help to prevent ALI and save lives. The expression of Src homology-2 domain-containing protein tyrosine phosphatase 2 (SHP2) was found to be significantly higher in macrophages and lung tissues with ALI, and SHP2-associated MAPK pathways were activated by lipopolysaccharide (LPS). The knockdown of the SHP2 gene suppressed the LPS-induced release of inflammatory factors and the phosphorylation of regulators in the NF-κB pathways in macrophages. Our findings showed crosstalk between the LPS-induced inflammatory pathway and the SHP2-associated MAPK pathways. SHP2 inhibition could be a valuable therapeutic approach for inhibiting excessive inflammation in ALI. We discovered that giving SHP099, a specific allosteric inhibitor of SHP2, to mice with ALI and sepsis relieves ALI and significantly increases animal survival. Our study highlights the important role of SHP2 in ALI development and demonstrates the potential application of SHP099 for treating ALI.
Collapse
|
27
|
Ashino T, Nakamura Y, Ohtaki H, Iwakura Y, Numazawa S. Interleukin-6 regulates the expression of hepatic canalicular efflux drug transporters after cecal ligation and puncture-induced sepsis: A comparison with lipopolysaccharide treatment. Toxicol Lett 2023; 374:40-47. [PMID: 36526125 DOI: 10.1016/j.toxlet.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/01/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Hepatic multidrug transporters expressed on the canalicular membrane play a role in the hepatobiliary excretion of xenobiotics and endogenous substrates. The aim of this study was to elucidate the role of pro-inflammatory cytokines in the regulation of hepatic drug transporter expression after cecal ligation and puncture (CLP), a valuable tool for studying polymicrobial sepsis, and to compare CLP with lipopolysaccharide (LPS) treatment. CLP reduced the expression of Mdr2/Abcb4, Mrp2/Abcc2, Bsep/Abcb11, Bcrp/Abcg2, and Mate1/Slc47a1 mRNAs in wild-type (WT) mouse livers in a time-dependent manner up to 48 h postoperation. LPS also reduced the expression of all transporters in WT mouse livers 24 h posttreatment; thereafter, expression levels tended to return to normal by 48 h posttreatment. IL-6-/- mice exhibited inhibited downregulation of drug transporters following CLP, although IL-1-/- and TNFα-/- mice exhibited the reduced expression of all transporters in a manner similar to that found in WT mice. Compared with CLP, LPS treatment reduced the expression of all transporters in all cytokine-deficient mouse livers, except for the expression of Mrp2/Abcc2 in IL-6-/- mice. Overall, these findings suggest that IL-6 is major factor in the downregulation of hepatic multidrug transporters following the onset of polymicrobial sepsis but not after LPS treatment.
Collapse
Affiliation(s)
- Takashi Ashino
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan.
| | - Yuki Nakamura
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Center for Pharmaceutical Education, Faculty of Pharmacy, Yokohama University of Pharmacy, 601 Matano, Totsuka, Yokohama, Kanagawa 245-0066, Japan
| | - Hirokazu Ohtaki
- Department of Functional Neurobiology, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, 2669 Yamazaki, Noda, Chiba 278-0022, Japan
| | - Satoshi Numazawa
- Division of Toxicology, Department of Pharmacology, Toxicology and Therapeutics, Showa University School of Pharmacy, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan; Pharmacological Research Center, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| |
Collapse
|
28
|
Central Venous Waveform Analysis and Cardiac Output in a Porcine Model of Endotoxemic Hypotension and Resuscitation. J Am Coll Surg 2023; 236:294-304. [PMID: 36648257 DOI: 10.1097/xcs.0000000000000474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cardiac output (CO) is a valuable proxy for perfusion, and governs volume responsiveness during resuscitation from distributive shock. The underappreciated venous system has nuanced physiology that confers valuable hemodynamic information. In this investigation, deconvolution of the central venous waveform by the fast Fourier transformation (FFT) algorithm is performed to assess its ability to constitute a CO surrogate in a porcine model of endotoxemia-induced distributive hypotension and resuscitation. STUDY DESIGN Ten pigs were anesthetized, catheterized, and intubated. A lipopolysaccharides infusion protocol was used to precipitate low systemic vascular resistance hypotension. Four crystalloid boluses (10 cc/kg) were then given in succession, after which heart rate, mean arterial pressure, thermodilution-derived CO, central venous pressure (CVP), and the central venous waveform were collected, the last undergoing fast Fourier transformation analysis. The amplitude of the fundamental frequency of the central venous waveform's cardiac wave (f0-CVP) was obtained. Heart rate, mean arterial pressure, CVP, f0-CVP, and CO were plotted over the course of the boluses to determine whether f0-CVP tracked with CO better than the vital signs, or than CVP itself. RESULTS Distributive hypotension to a 25% mean arterial pressure decrement was achieved, with decreased systemic vascular resistance (mean 918 ± 227 [SD] dyne/s/cm-5 vs 685 ± 180 dyne/s/cm-5; p = 0.038). Full hemodynamic parameters characterizing this model were reported. Slopes of linear regression lines of heart rate, mean arterial pressure, CVP, f0-CVP, and CO were -2.8, 1.7, 1.8, 0.40, and 0.35, respectively, demonstrating that f0-CVP values closely track with CO over the 4-bolus range. CONCLUSIONS Fast Fourier transformation analysis of the central venous waveform may allow real-time assessment of CO during resuscitation from distributive hypotension, possibly offering a venous-based approach to clinical estimation of volume responsiveness.
Collapse
|
29
|
Zhou L, Zhang D, Kong L, Xu X, Gong D. Clinical improvement of sepsis by extracorporeal centrifugal leukocyte apheresis in a porcine model. J Transl Med 2022; 20:538. [PMID: 36419190 PMCID: PMC9682844 DOI: 10.1186/s12967-022-03752-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Extracorporeal blood purification therapies targeting removal of the downstream products of the inflammatory cascade in sepsis have failed to improve mortality. As an upstream process of the inflammatory cascade, activated white blood cells should be a potential therapeutic target for sepsis, and the effect of removing such cells by extracorporeal centrifugal leukocytapheresis (LCAP) is worth considering. METHODS Fourteen peritonitis-induced septic pigs were randomly assigned to receive a sham operation (control group, n = 7) or one session of LCAP at 12 h after sepsis induction (treatment group, n = 7). Samples from peripheral blood at various time-points and from LCAP collection were tested. All pigs were euthanized at 48 h, and lung, kidney, liver and spleen tissues were obtained for histopathological examination. RESULTS Two pigs died in accidents before the induction of sepsis, and 12 pigs were finally included for the statistical analysis. A significant clinical improvement was present in the treatment group relative to the control group in terms of the mean arterial blood pressure (MAP), oxygen tension (PaO2), lactic acid level, oxygenation index (PaO2/FiO2), and carbon dioxide tension (PaCO2, P < 0.05). Flow cytometry tests showed that a mixture of B cells, dendritic cells, T helper cells, cytotoxic T cells, monocytes and neutrophils were removed from the circulation by LCAP, resulting in sepsis-induced change trends in the control cells; these change trends were all flattened in the treatment group, although nonsignificantly. CONCLUSIONS LCAP may exert a wide-spectrum and bidirectional immunomodulatory effect on sepsis, accompanied by improvements in hemodynamics and oxygenation status.
Collapse
Affiliation(s)
- Lei Zhou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, 305 Zhongshan East Road, Xuanwu District, Nanjing, 210016, China
| | - Dong Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, 305 Zhongshan East Road, Xuanwu District, Nanjing, 210016, China
| | - Ling Kong
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, 305 Zhongshan East Road, Xuanwu District, Nanjing, 210016, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, 305 Zhongshan East Road, Xuanwu District, Nanjing, 210016, China
| | - Dehua Gong
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, 305 Zhongshan East Road, Xuanwu District, Nanjing, 210016, China.
| |
Collapse
|
30
|
Danielski LG, Giustina AD, Gava FF, Barichello T, Petronilho F. The Many Faces of Astrocytes in the Septic Brain. Mol Neurobiol 2022; 59:7229-7235. [PMID: 36136265 DOI: 10.1007/s12035-022-03027-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022]
Abstract
Sepsis is a life-threatening organ dysfunction that is caused by a dysregulated host response to infection. Surviving patients have cognitive and memory damage that started during sepsis. These neurologic damages have been associated with increased BBB permeability and microglial activation. However, a few discrete studies have seen over the years pointing to the potential role of astrocytes in the pathophysiology of neurological damage after sepsis. The purpose of this article is to review information on the potential role of astrocytes during sepsis, as well as to provoke further studies in this area. These published articles show astrocytic activation after sepsis; they also evidence the release of inflammatory mediators by these cells. In this sense, the role of astrocytes should be better elucidated during sepsis progression.
Collapse
Affiliation(s)
- Lucinéia Gainski Danielski
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Graduate Program in Health Sciences, Health Sciences Unit, University of South Santa Catarina, Tubarao, SC, Brazil.,Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Amanda Della Giustina
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Fernanda Frederico Gava
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil
| | - Tatiana Barichello
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil.,Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, Translational Psychiatry Program, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, 77054, USA
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciuma, SC, Brazil.
| |
Collapse
|
31
|
Hof S, Marcus C, Kuebart A, Schulz J, Truse R, Raupach A, Bauer I, Flögel U, Picker O, Herminghaus A, Temme S. A Toolbox to Investigate the Impact of Impaired Oxygen Delivery in Experimental Disease Models. Front Med (Lausanne) 2022; 9:869372. [PMID: 35652064 PMCID: PMC9149176 DOI: 10.3389/fmed.2022.869372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/25/2022] [Indexed: 12/29/2022] Open
Abstract
Impaired oxygen utilization is the underlying pathophysiological process in different shock states. Clinically most important are septic and hemorrhagic shock, which comprise more than 75% of all clinical cases of shock. Both forms lead to severe dysfunction of the microcirculation and the mitochondria that can cause or further aggravate tissue damage and inflammation. However, the detailed mechanisms of acute and long-term effects of impaired oxygen utilization are still elusive. Importantly, a defective oxygen exploitation can impact multiple organs simultaneously and organ damage can be aggravated due to intense organ cross-talk or the presence of a systemic inflammatory response. Complexity is further increased through a large heterogeneity in the human population, differences in genetics, age and gender, comorbidities or disease history. To gain a deeper understanding of the principles, mechanisms, interconnections and consequences of impaired oxygen delivery and utilization, interdisciplinary preclinical as well as clinical research is required. In this review, we provide a "tool-box" that covers widely used animal disease models for septic and hemorrhagic shock and methods to determine the structure and function of the microcirculation as well as mitochondrial function. Furthermore, we suggest magnetic resonance imaging as a multimodal imaging platform to noninvasively assess the consequences of impaired oxygen delivery on organ function, cell metabolism, alterations in tissue textures or inflammation. Combining structural and functional analyses of oxygen delivery and utilization in animal models with additional data obtained by multiparametric MRI-based techniques can help to unravel mechanisms underlying immediate effects as well as long-term consequences of impaired oxygen delivery on multiple organs and may narrow the gap between experimental preclinical research and the human patient.
Collapse
Affiliation(s)
- Stefan Hof
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Carsten Marcus
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jan Schulz
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Richard Truse
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Annika Raupach
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Inge Bauer
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Olaf Picker
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anna Herminghaus
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Sebastian Temme
- Department of Anaesthesiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
32
|
Skorup P, Fransson A, Gustavsson J, Sjöholm J, Rundgren H, Özenci V, Wong AYW, Karlsson T, Svensén C, Günther M. Evaluation of an extracorporeal ozone-based bactericide system for the treatment of Escherichia coli sepsis. Intensive Care Med Exp 2022; 10:14. [PMID: 35467176 PMCID: PMC9038973 DOI: 10.1186/s40635-022-00443-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sepsis is associated with substantial mortality rates. Antibiotic treatment is crucial, but global antibiotic resistance is now classified as one of the top ten global public health risks facing humanity. Ozone (O3) is an inorganic molecule with no evident function in the body. We investigated the bactericide properties of ozone, using a novel system of extracorporeal ozone blood treatment. We hypothesized that ozone would decrease the concentration of viable Escherichia coli (E. coli) in human whole blood and that the system would be technically feasible and physiologically tolerable in a clinically relevant model of E. coli sepsis in swine. METHODS The E. coli strain B09-11822, a clinical isolate from a patient with septic shock was used. The in vitro study treated E. coli infected human whole blood (n = 6) with ozone. The in vivo 3.5-h sepsis model randomized swine to E. coli infusion and ozone treatment (n = 5) or E. coli infusion and no ozone treatment (n = 5). Live E. coli, 5 × 107 colony-forming units (CFU/mL) was infused in a peripheral vein. Ozone treatment was initiated with a duration of 30 min after 1.5 h. RESULTS The single pass in vitro treatment decreased E. coli by 27%, mean 1941 to 1422 CFU/mL, mean of differences - 519.0 (95% CI - 955.0 to - 82.98, P = 0.0281). pO2 increased (95% CI 31.35 to 48.80, P = 0.0007), pCO2 decreased (95% CI - 3.203 to - 1.134, P = 0.0069), oxyhemoglobin increased (95% CI 1.010 to 3.669, P = 0.0113). Methemoglobin was not affected. In the sepsis model, 9/10 swine survived. One swine randomized to ozone treatment died from septic shock before initiation of the treatment. Circulatory, respiratory, and metabolic parameters were not affected by the ozone treatment. E. coli in arterial blood, in organs and in aerobic and anaerobic blood cultures did not differ. Hemoglobin, leucocytes, and methemoglobin were not affected by the treatment. CONCLUSIONS Ozone decreased the concentration of viable E. coli in human whole blood. The system was technically feasible and physiologically tolerable in porcine sepsis/septic shock and should be considered for further studies towards clinical applications.
Collapse
Affiliation(s)
- Paul Skorup
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Anette Fransson
- Section for Experimental Traumatology, Department of Neuroscience, Karolinska Institutet, Biomedicum - 8B, 171 77, Stockholm, Sweden
| | - Jenny Gustavsson
- Section for Experimental Traumatology, Department of Neuroscience, Karolinska Institutet, Biomedicum - 8B, 171 77, Stockholm, Sweden
| | | | | | - Volkan Özenci
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.,Department of Clinical Microbiology, Karolinska University Hospital, Huddinge, Sweden
| | - Alicia Y W Wong
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Tomas Karlsson
- Department of Clinical Science at Education Södersjukhuset, Unit of Anesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| | - Christer Svensén
- Department of Clinical Science at Education Södersjukhuset, Unit of Anesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Günther
- Section for Experimental Traumatology, Department of Neuroscience, Karolinska Institutet, Biomedicum - 8B, 171 77, Stockholm, Sweden. .,Department of Clinical Science at Education Södersjukhuset, Unit of Anesthesiology and Intensive Care, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
33
|
Kim HG, Lee C, Yoon JH, Kim JH, Cho JY. BN82002 alleviated tissue damage of septic mice by reducing inflammatory response through inhibiting AKT2/NF-κB signaling pathway. Pharmacotherapy 2022; 148:112740. [DOI: 10.1016/j.biopha.2022.112740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 01/20/2023]
|
34
|
The Effects of a Meldonium Pre-Treatment on the Course of the LPS-Induced Sepsis in Rats. Int J Mol Sci 2022; 23:ijms23042395. [PMID: 35216510 PMCID: PMC8924897 DOI: 10.3390/ijms23042395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
A dysregulated and overwhelming response to an infection accompanied by the exaggerated pro-inflammatory state and metabolism disturbance leads to the fatal outcome in sepsis. Previously we showed that meldonium, an anti-ischemic drug clinically used to treat myocardial and cerebral ischemia, strongly increases mortality in faecal-induced peritonitis (FIP) in rats. We postulated that the same mechanism that is responsible for the otherwise strong anti-inflammatory effects of meldonium could be the culprit of the increased mortality. In the present study, we applied the LPS-induced model of sepsis to explore the presence of any differences from and/or similarities to the FIP model. When it comes to energy production, despite some shared similarities, it is evident that LPS and FIP models of sepsis differ greatly. A different profile of sympathoadrenal activation may account for this observation, as it was lacking in the FIP model, whereas in the LPS model it was strong enough to overcome the effects of meldonium. Therefore, choosing the appropriate model of sepsis induction is of great importance, especially if energy homeostasis is the main focus of the study. Even when differences in the experimental design of the two models are acknowledged, the role of different patterns of energy production cannot be excluded. On that account, our results draw attention to the importance of uninterrupted energy production in sepsis but also call for much-needed revisions of the current recommendations for its treatment.
Collapse
|
35
|
Wang L, Ansari S, Cai Y, McCracken B, Tiba MH, Ward K, Najarian K, Oldham K. Tracking Peripheral Artery Motion and Vascular Resistance with a Multi-Modal Wearable Sensor Under Pressure Perturbations. J Biomech Eng 2022; 144:1133339. [PMID: 35079769 DOI: 10.1115/1.4053399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Indexed: 11/08/2022]
Abstract
Peripheral artery status is a key physiological indicator of the body's cardiovascular response to both acute and chronic medical conditions. In this paper, peripheral artery behavior is tracked non-invasively by combining a photo plethysmograph (PPG) sensor and a piezoelectric (polyvinylidene difluoride, PVDF) sensor, while applying an outside pressure-varying cuff. A simple mechanical model for the local artery and tissue captures time- and pressure-dependent features present in the PPG and PVDF signals with respect to applied cuff pressure, during maneuvers applied to multiple swine subjects to perturb blood pressure and vascular resistance. These behaviors provide insight into feasibility and robustness of cardiovascular property identification by multi-modal non-invasive wearable sensing. This is found to help refine non-invasive blood pressure measurements and estimation of systemic vascular resistance (SVR) and blood pressure (BP) using selected features of sensor amplitude versus applied cuff pressure.
Collapse
Affiliation(s)
- Lu Wang
- University of Michigan, Ann Arbor, Michigan, USA
| | | | - Yingjie Cai
- University of Michigan, Ann Arbor, Michigan, USA
| | | | | | - Kevin Ward
- University of Michigan, Ann Arbor, Michigan, USA
| | | | - Kenn Oldham
- University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
36
|
Rosier F, Nuñez NF, Torres M, Loriod B, Rihet P, Pradel LC. Transcriptional Response in a Sepsis Mouse Model Reflects Transcriptional Response in Sepsis Patients. Int J Mol Sci 2022; 23:ijms23020821. [PMID: 35055007 PMCID: PMC8776114 DOI: 10.3390/ijms23020821] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 01/06/2022] [Accepted: 01/09/2022] [Indexed: 02/06/2023] Open
Abstract
Mortality due to sepsis remains unacceptably high, especially for septic shock patients. Murine models have been used to better understand pathophysiology mechanisms. However, the mouse model is still under debate. Herein we investigated the transcriptional response of mice injected with lipopolysaccharide (LPS) and compared it to either human cells stimulated in vitro with LPS or to the blood cells of septic patients. We identified a molecular signature composed of 2331 genes with an FDR median of 0%. This molecular signature is highly enriched in regulated genes in peritoneal macrophages stimulated with LPS. There is significant enrichment in several inflammatory signaling pathways, and in disease terms, such as pneumonia, sepsis, systemic inflammatory response syndrome, severe sepsis, an inflammatory disorder, immune suppression, and septic shock. A significant overlap between the genes upregulated in mouse and human cells stimulated with LPS has been demonstrated. Finally, genes upregulated in mouse cells stimulated with LPS are enriched in genes upregulated in human cells stimulated in vitro and in septic patients, who are at high risk of death. Our results support the hypothesis of common molecular and cellular mechanisms between mouse and human sepsis.
Collapse
Affiliation(s)
| | | | | | | | - Pascal Rihet
- Correspondence: (P.R.); (L.C.P.); Tel.: +33-491828723 (P.R.); +33-491828745 (L.C.P.)
| | - Lydie C. Pradel
- Correspondence: (P.R.); (L.C.P.); Tel.: +33-491828723 (P.R.); +33-491828745 (L.C.P.)
| |
Collapse
|
37
|
Li J, Li M, Li L, Ma J, Yao C, Yao S. Hydrogen sulfide attenuates ferroptosis and stimulates autophagy by blocking mTOR signaling in sepsis-induced acute lung injury. Mol Immunol 2022; 141:318-327. [PMID: 34952420 DOI: 10.1016/j.molimm.2021.12.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 10/21/2021] [Accepted: 12/01/2021] [Indexed: 12/14/2022]
Abstract
Sepsis often leads to multiple organ failure or even death and is a significant health problem that contributes to a heavy economic burden. The lung is the first organ to be affected by sepsis. Presently, there is no specific drug or method to treat sepsis and sepsis-induced acute lung injury (ALI). H2S, along with CO and NO, is a physiological gas that acts as a signaling molecule and plays an active role in fighting various lung infections. GYY4137 is a novel H2S donor that is stable in vivo and in vitro. However, particularly in the context of ferroptosis, GYY4137 affects cecal ligation and puncture (CLP)-induced ALI by a mechanism that is not understood. Ferroptosis is a new form of cell necrosis. The primary mechanism is the accumulation of cellular lipid ROS in an iron-dependent manner. The principal objective of this project was to investigate the effects of GYY4137 on ferroptosis and autophagy in a mouse model of sepsis-induced ALI. We divided the experimental mice randomly into 5 groups: (1) sham group; (2) CLP group; (3) CLP + DMSO group: (4) CLP + GYY4137 (25 mg/kg) group; and (5) CLP + GYY4137 (50 mg/kg) group. (6) CLP + Rapamycin (2.0 mg/Kg) group. (7) CLP + Chloroquine (80 mg/Kg) group. (8) the Chloroquine (80 mg/Kg) + GYY (50 mg/Kg) group. The findings showed that GYY4137 significantly protected against CLP-induced ALI by improving sepsis-induced lung histopathological changes, diminishing lung tissue damage, ameliorating oxidative stress, and attenuating the severity of lung injury in mice. In this study, we found that GYY4137 could alleviate septicemia-induced ferroptosis in ALI by increasing the expression of GPx4 and SLC7A11 in lung tissue after CLP. One unexpected finding was the extent to which the levels of ferritin and ferritin light chain increased after CLP, which may be a compensatory mechanism for storing abnormally increased iron. We also found that the expression of p-mTOR, P62, and Beclin1 was significantly increased and that LC3II/LC3I declined after LPS stimulation, but the effect was inhibited by treatment with GYY4137, indicating that GYY4137 could inhibit the activation of autophagy in sepsis-induced ALI by blocking mTOR signaling.
Collapse
Affiliation(s)
- Jianhua Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Department of Critical Care Medicine, The First Affiliated Hospital, College of Medicine, Shihezi University, Shihezi 832000, China
| | - Mengyu Li
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ling Li
- Department of Traumatology, The First Affiliated Hospital, College of Medicine, Shihezi University, Shihezi 832000, China
| | - Jiamin Ma
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chengye Yao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Shanglong Yao
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
38
|
Abstract
Endotoxaemia is an inflammatory condition which happens due to the presence of outer cell wall layer of Gram-negative bacteria in blood circulation, containing lipopolysaccharide commonly known as endotoxin. This condition causes high mortality in affected animals and sheep are highly susceptible in this regard. Several researchers have emphasised the therapeutic regimens of endotoxaemia and its sequels in sheep. Furthermore, sheep are among the most commonly used animal species in experimental studies on endotoxaemia, and for the past five decades, ovine models have been employed to evaluate different aspects of endotoxaemia. Currently, there are several studies on experimentally induced endotoxaemia in sheep, and information regarding novel therapeutic protocols in this species contributes to better understanding and treating the condition. This review aims to specifically introduce various treatment methods of endotoxaemia in sheep.
Collapse
Affiliation(s)
- A. Chalmeh
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
39
|
Vale A, Nascimento D, Pineros A, Ferreira R, Santos J, Aragon D, Cunha F, Ramalho F, Alves-Filho J, Carlotti A. Riboflavin did not provide anti-inflammatory or antioxidant effects in an experimental model of sepsis. Braz J Med Biol Res 2022; 55:e12107. [PMID: 35648977 PMCID: PMC9150426 DOI: 10.1590/1414-431x2022e12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/13/2022] [Indexed: 11/22/2022] Open
Abstract
We aimed to evaluate whether the administration of riboflavin to septic animals reduces inflammation, oxidative stress, organ dysfunction, and mortality. C57BL/6 mice, 6-8 weeks old, were allocated to the study group (polymicrobial sepsis induced by cecal ligation and puncture (CLP) + antibiotic + iv riboflavin), control (CLP + antibiotic + iv saline), or naïve (non-operated controls). Serum concentrations of alanine aminotransferase (ALT), creatine kinase-MB (CK-MB), urea, and creatinine, and markers of inflammation [interleukin (IL)-6, tumor necrosis factor (TNF)-α, keratinocyte-derived chemokine (KC), and macrophage inflammatory protein (MIP)-2)], and oxidative stress (malondialdehyde (MDA) were measured 12 h after the experiment. Animal survival rates were calculated after 7 days. Means between groups were compared using linear regression models adjusted under the Bayesian approach. No significant difference was observed between control and study groups in serum concentrations of IL-6 (95% credible interval) (-0.35 to 0.44), TNF-α (-15.7 to 99.1), KC (-0.13 to 0.05), MIP-2 (-0.84 to 0.06), MDA (-1.25 to 2.53), or ALT (-6.6 to 11.5). Serum concentrations of CK-MB (-145.1 to -30.1), urea (-114.7 to -15.1), and creatinine (-1.14 to -0.01) were higher in the study group. Survival was similar in both groups (P=0.8). Therefore, the use of riboflavin in mice undergoing sepsis induced by CLP did not reduce inflammation, oxidative stress, organ dysfunction, or mortality compared with placebo.
Collapse
|
40
|
Li F, Yan R, Wu J, Han Z, Qin M, Liu C, Lu Y. An Antioxidant Enzyme Therapeutic for Sepsis. Front Bioeng Biotechnol 2021; 9:800684. [PMID: 34888304 PMCID: PMC8650590 DOI: 10.3389/fbioe.2021.800684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/08/2021] [Indexed: 11/22/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infections that may lead to organ dysfunction with high mortality. With the rapid increase in the aging population and antimicrobial resistance, developing therapeutics for the treatment of sepsis has been an unmet medical need. Excessive production of reactive oxygen species (ROS) during inflammation is associated with the occurrence of sepsis. We report herein a treatment for sepsis based on PEGylated catalase, which can effectively break down hydrogen peroxide, a key component of ROS that is chemically stable and able to diffuse around the tissues and form downstream ROS. PEGylated catalase can effectively regulate the cytokine production by activated leukocytes, suppress the elevated level of AST, ALT, TNF-α, and IL-6 in mice with induced sepsis, and significantly improve the survival rate.
Collapse
Affiliation(s)
- Feifei Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ran Yan
- Department of Chemical and Biomolecular Engineering, The University of California, Los Angeles, CA, United States
| | - Jun Wu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zeren Han
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Meng Qin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, The University of California, Los Angeles, CA, United States
| |
Collapse
|
41
|
A host-directed macrocyclic peptide therapeutic for MDR gram negative bacterial infections. Sci Rep 2021; 11:23447. [PMID: 34873199 PMCID: PMC8648872 DOI: 10.1038/s41598-021-02619-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 01/26/2023] Open
Abstract
The emergence of infections by carbapenem resistant Enterobacteriaceae (CRE) pathogens has created an urgent public health threat, as carbapenems are among the drugs of last resort for infections caused by a growing fraction of multi-drug resistant (MDR) bacteria. There is global consensus that new preventive and therapeutic strategies are urgently needed to combat the growing problem of MDR bacterial infections. Here, we report on the efficacy of a novel macrocyclic peptide, minimized theta-defensin (MTD)-12813 in CRE sepsis. MTD12813 is a theta-defensin inspired cyclic peptide that is highly effective against CRE pathogens K. pneumoniae and E. coli in vivo. In mouse septicemia models, single dose administration of MTD12813 significantly enhanced survival by promoting rapid host-mediated bacterial clearance and by modulating pathologic cytokine responses, restoring immune homeostasis, and preventing lethal septic shock. The peptide lacks direct antibacterial activity in the presence of mouse serum or in peritoneal fluid, further evidence for its indirect antibacterial mode of action. MTD12813 is highly stable in biological matrices, resistant to bacterial proteases, and nontoxic to mice at dose levels 100 times the therapeutic dose level, properties which support further development of the peptide as a first in class anti-infective therapeutic.
Collapse
|
42
|
Xue W, Li Y, Zhang M. Pristimerin inhibits neuronal inflammation and protects cognitive function in mice with sepsis-induced brain injuries by regulating PI3K/Akt signalling. PHARMACEUTICAL BIOLOGY 2021; 59:1351-1358. [PMID: 34590530 PMCID: PMC8491740 DOI: 10.1080/13880209.2021.1981399] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 09/10/2021] [Indexed: 06/05/2023]
Abstract
CONTEXT Sepsis is a systemic inflammatory disease; pristimerin exhibits strong antibacterial, anti-inflammatory and antioxidant properties. OBJECTIVES We explored whether pristimerin protected against cognitive dysfunction and neuroinflammation in C57BL/6 J mice with sepsis-induced brain injuries. MATERIALS AND METHODS Sepsis was induced by intraperitoneal administration of 2 mg/kg lipopolysaccharide (LPS). C57BL/6 J mice were separated into four groups (n = 10 per group): positive control, negative control, pristimerin 10 mg/kg and pristimerin 100 mg/kg. Pristimerin was administered orally for 28 days prior to LPS administration and for six days thereafter. Behavioural changes were assessed one day after LPS administration using the Morris water maze and via neurological dysfunction scoring. Molecular pathogenesis was explored by measurement of malondialdehyde, superoxide dismutase, reactive oxygen species and inflammatory cytokine levels in mouse brains. Neuronal apoptosis was evaluated using the TUNEL assay. The levels of p-Akt/Akt, p-PI3K/PI3K, mTOR, Bax, Bcl-2 and caspase-3 proteins were determined via Western blotting. RESULTS Pristimerin improved cognitive function and reduces the neurological score to 1.15 ± 0.03. Pristimerin significantly reduced all cytokine levels: TNF-α by 18 ± 0.6 pg/mg, IL-1β by 43 ± 1.3 pg/mg and IL-6 by 34 ± 1.12 pg/mg. There was significant (p < 0.01) improvement in PI3K/Akt signalling and histopathological changes in the brain tissue of sepsis induced brain injured rats. CONCLUSIONS Pristimerin ameliorated neuronal injury by regulating PI3K/Akt signalling in mice with sepsis-induced brain injuries. Pristimerin may merit further development for clinical applications.
Collapse
Affiliation(s)
- Weimin Xue
- Department of Neurology, First Affiliated Hospital of Anhui University of Science and Technology, First People's Hospital of Huainan, Huainan, China
| | - Yaqiang Li
- Department of Neurology, First Affiliated Hospital of Anhui University of Science and Technology, First People's Hospital of Huainan, Huainan, China
| | - Mei Zhang
- Department of Neurology, First Affiliated Hospital of Anhui University of Science and Technology, First People's Hospital of Huainan, Huainan, China
| |
Collapse
|
43
|
Lord MS, Berret JF, Singh S, Vinu A, Karakoti AS. Redox Active Cerium Oxide Nanoparticles: Current Status and Burning Issues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102342. [PMID: 34363314 DOI: 10.1002/smll.202102342] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/09/2021] [Indexed: 06/13/2023]
Abstract
Research on cerium oxide nanoparticles (nanoceria) has captivated the scientific community due to their unique physical and chemical properties, such as redox activity and oxygen buffering capacity, which made them available for many technical applications, including biomedical applications. The redox mimetic antioxidant properties of nanoceria have been effective in the treatment of many diseases caused by reactive oxygen species (ROS) and reactive nitrogen species. The mechanism of ROS scavenging activity of nanoceria is still elusive, and its redox activity is controversial due to mixed reports in the literature showing pro-oxidant and antioxidant activity. In light of its current research interest, it is critical to understand the behavior of nanoceria in the biological environment and provide answers to some of the critical and open issues. This review critically analyzes the status of research on the application of nanoceria to treat diseases caused by ROS. It reviews the proposed mechanism of action and shows the effect of surface coatings on its redox activity. It also discusses some of the crucial issues in deciphering the mechanism and redox activity of nanoceria and suggests areas of future research.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, New South Wales, 2052, Australia
| | | | - Sanjay Singh
- National Institute of Animal Biotechnology, Hyderabad, Telangana, 500032, India
| | - Ajayan Vinu
- Global Innovative Center for Advanced Nanomaterials, College of Engineering Science and Environment, The University of Newcastle, Callaghan, New South Wales, 2308, Australia
| | - Ajay S Karakoti
- Global Innovative Center for Advanced Nanomaterials, College of Engineering Science and Environment, The University of Newcastle, Callaghan, New South Wales, 2308, Australia
| |
Collapse
|
44
|
The microbial composition of the initial insult can predict the prognosis of experimental sepsis. Sci Rep 2021; 11:22772. [PMID: 34815465 PMCID: PMC8611025 DOI: 10.1038/s41598-021-02129-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/03/2021] [Indexed: 12/21/2022] Open
Abstract
We hypothesized that the composition of sepsis-inducing bacterial flora influences the course of fecal peritonitis in rodents. Saline or fecal suspensions with a standardized dose range of bacterial colony-forming units (CFUs) were injected intraperitoneally into Sprague–Dawley rats. The qualitative composition of the initial inoculum and the ascites was analyzed separately by MALDI-TOF mass spectrometry. Invasive monitoring was conducted in separate anesthetized groups (n = 12–13/group) after 12, 24, 48 and 72 h to determine rat-specific organ failure assessment (ROFA) scores. Death and ROFA scores peaked at 24 h. At this time, 20% mortality occurred in animals receiving a monomicrobial E. coli suspension, and ROFA scores were significantly higher in the monomicrobial subgroup than in the polymicrobial one (median 6.5; 5.0–7.0 and 5.0; 4.75–5.0, respectively). ROFA scores dropped after 48 h, accompanied by a steady decrease in ascites CFUs and a shift towards intra-abdominal monomicrobial E. coli cultures. Furthermore, we found a relationship between ascites CFUs and the evolving change in ROFA scores throughout the study. Hence, quantitatively identical bacterial loads with mono- or polymicrobial dominance lead to a different degree of sepsis severity and divergent outcomes. Initial and intraperitoneal microbiological testing should be used to improve translational research success.
Collapse
|
45
|
Goggs R, Kannampuzha-Francis J, Campbell CJ, Moreau JP, Behling-Kelly E. Pharmacokinetics, pharmacodynamics and safety evaluation of 5,5'-methylenebis(2-acetoxybenzoic acid) in dogs following intravenous administration. Vet Immunol Immunopathol 2021; 242:110339. [PMID: 34715602 DOI: 10.1016/j.vetimm.2021.110339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/15/2022]
Abstract
Complement-mediated intravascular hemolysis occurs in canine immune-mediated hemolytic anemia (IMHA). Complement inhibitors might enhance treatment of this disease. Dimers of acetylsalicylic acid such as 5,5'-methylenebis(2-acetoxybenzoic acid) (DAS) have been reported to inhibit complement. This study aimed to characterize the pharmacokinetics and safety profile of a single 3 mg/kg IV dose of DAS in 6 healthy mixed-breed dogs. Serum concentrations of DAS and its primary metabolites were measured by liquid chromatography-tandem mass spectrometry at baseline and at 5, 10 and 30 min, and 1, 2, 4, 6, 8, 12, 18 and 24 h post-administration. Additional blood samples were collected 7 and 14 days after drug administration. Complete blood counts, serum chemistry panels, C-reactive protein measurements, coagulation testing and cytokine analyses were used for safety monitoring. Following IV administration of 3 mg/kg DAS, the estimated mean maximum plasma concentration was 54,709 ng/mL. Pharmacokinetic modeling suggested that DAS was eliminated with a half-life value of 8.1 h, equivalent to a clearance of 6.93 L/hr kg and a volume of distribution of 56 mL/kg. Plasma concentrations of the metabolites were measured rapidly (within 15-60 min for M1 and M2 respectively). Overall, the relative exposure to M1 and M2 suggest significant biotransformation of DAS occurred, but DAS was the most abundant circulating species. No adverse clinical reactions were noted following DAS administration and safety studies suggested DAS caused no inflammatory response or coagulation disturbance. Further clinical evaluation of DAS is warranted.
Collapse
Affiliation(s)
- Robert Goggs
- Department of Clinical Sciences, Ithaca, NY, 14853, United States.
| | - Jasmine Kannampuzha-Francis
- Department of Population Medicine, Cornell University College of Veterinary Medicine, Ithaca, NY, 14853, United States
| | | | - Jean-Pierre Moreau
- Aurin Biotech Inc., 555 Burrard St Floor 2, Vancouver, BC, V7X 1M8, Canada
| | - Erica Behling-Kelly
- Department of Population Medicine, Cornell University College of Veterinary Medicine, Ithaca, NY, 14853, United States
| |
Collapse
|
46
|
VEGF-C and podoplanin, as biomarkers of sepsis. An experimental study. REV ROMANA MED LAB 2021. [DOI: 10.2478/rrlm-2021-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Background: Sepsis is the leading cause of morbidity and mortality in intensive care units. This study explored the possible role of vascular endothelial growth factor-C (VEGF-C) and podoplanin (PDPN) in sepsis.
Methods: 22 Wistar rats were divided into three groups: two experimental (Group A and B, n=8/8) and a control (Group C, n=6). Sepsis was induced with intraperitoneal injection of ESBL (extended-spectrum beta-lactamases)-producing E-coli live bacteria for group A and with lipopolysaccharide for group B. Sterile saline solution was injected for group C. Blood samples were collected after 24 hours to determine the serum level of VEGF-C, and PDPN expression was examined in liver, kidney, and lung tissues. Bacteremia was assessed for group A.
Results: Higher serum levels of VEGF-C were found in Group A vs C (p=0.05) and group B vs. C (p=0.004), respectively.VEGF-C was also increased in animals with negative- vs. positive blood cultures from group A (p=0.04) and from group B vs. those with positive blood cultures from group A (p=0.03). High intensity of PDPN tissue expression was observed in the pulmonary alveolocytes from Group A and epithelium of the proximal renal tubules in groups B and C, compared to group A.
Conclusions: Circulating VEGF-C can be succesfuly used as a biomarker of sepsis with negative blood cultures and high risk of renal failure, whereas PDPN seems to exert a protective role against lung injuries in live bacteria-induced sepsis.
Collapse
|
47
|
Dejani NN, Elshabrawy HA, Bezerra Filho CDSM, de Sousa DP. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules 2021; 11:biom11081254. [PMID: 34439920 PMCID: PMC8394099 DOI: 10.3390/biom11081254] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/14/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023] Open
Abstract
In 2019, COVID-19 emerged as a severe respiratory disease that is caused by the novel coronavirus, Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2). The disease has been associated with high mortality rate, especially in patients with comorbidities such as diabetes, cardiovascular and kidney diseases. This could be attributed to dysregulated immune responses and severe systemic inflammation in COVID-19 patients. The use of effective antiviral drugs against SARS-CoV-2 and modulation of the immune responses could be a potential therapeutic strategy for COVID-19. Studies have shown that natural phenolic compounds have several pharmacological properties, including anticoronavirus and immunomodulatory activities. Therefore, this review discusses the dual action of these natural products from the perspective of applicability at COVID-19.
Collapse
Affiliation(s)
- Naiara Naiana Dejani
- Department of Physiology and Pathology, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
| | - Hatem A. Elshabrawy
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA;
| | - Carlos da Silva Maia Bezerra Filho
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
| | - Damião Pergentino de Sousa
- Department of Pharmaceutical Sciences, Federal University of Paraíba, João Pessoa 58051-900, Brazil;
- Postgraduate Program in Bioactive Natural and Synthetic Products, Federal University of Paraíba, João Pessoa 58051-900, Brazil
- Correspondence: ; Tel.: +55-83-3216-7347
| |
Collapse
|
48
|
Increased mortality and altered local immune response in secondary peritonitis after previous visceral operations in mice. Sci Rep 2021; 11:16175. [PMID: 34376743 PMCID: PMC8355121 DOI: 10.1038/s41598-021-95592-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/27/2021] [Indexed: 12/29/2022] Open
Abstract
Postoperative peritonitis is characterized by a more severe clinical course than other forms of secondary peritonitis. The pathophysiological mechanisms behind this phenomenon are incompletely understood. This study used an innovative model to investigate these mechanisms, combining the models of murine Colon Ascendens Stent Peritonitis (CASP) and Surgically induced Immune Dysfunction (SID). Moreover, the influence of the previously described anti-inflammatory reflex transmitted by the vagal nerve was characterized. SID alone, or 3 days before CASP were performed in female C57BL/6 N mice. Subdiaphragmatic vagotomy was performed six days before SID with following CASP. The immune status was assessed by FACS analysis and measurement of cytokines. Local intestinal inflammatory changes were characterized by immunohistochemistry. Mortality was increased in CASP animals previously subjected to SID. Subclinical bacteremia occurred after SID, and an immunosuppressive milieu occurred secondary to SID just before the induction of CASP. Previous SID modified the pattern of intestinal inflammation induced by CASP. Subdiaphragmatic vagotomy had no influence on sepsis mortality in our model of postoperative peritonitis. Our results indicate a surgery-induced inflammation of the small intestine and the peritoneal cavity with bacterial translocation, which led to immune dysfunction and consequently to a more severe peritonitis.
Collapse
|
49
|
Peptide VSAK maintains tissue glucose uptake and attenuates pro-inflammatory responses caused by LPS in an experimental model of the systemic inflammatory response syndrome: a PET study. Sci Rep 2021; 11:14752. [PMID: 34285283 PMCID: PMC8292390 DOI: 10.1038/s41598-021-94224-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 07/07/2021] [Indexed: 12/30/2022] Open
Abstract
The present investigation using Positron Emission Tomography shows how peptide VSAK can reduce the detrimental effects produced by lipopolysaccharides in Dutch dwarf rabbits, used to develop the Systemic Inflammatory Response Syndrome (SIRS). Animals concomitantly treated with lipopolysaccharides (LPS) and peptide VSAK show important protection in the loss of radiolabeled-glucose uptake observed in diverse organs when animals are exclusively treated with LPS. Treatment with peptide VSAK prevented the onset of changes in serum levels of glucose and insulin associated with the establishment of SIRS and the insulin resistance-like syndrome. Treatment with peptide VSAK also allowed an important attenuation in the circulating levels of pro-inflammatory molecules in LPS-treated animals. As a whole, our data suggest that peptide VSAK might be considered as a candidate in the development of new therapeutic possibilities focused on mitigating the harmful effects produced by lipopolysaccharides during the course of SIRS.
Collapse
|
50
|
Karabulut G, Bedirli N, Akyürek N, Bağrıaçık EÜ. Dose-related effects of dexmedetomidine on sepsis-initiated lung injury in rats. Braz J Anesthesiol 2021; 71:271-277. [PMID: 33845100 PMCID: PMC9373509 DOI: 10.1016/j.bjane.2021.02.051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 02/14/2021] [Accepted: 02/27/2021] [Indexed: 02/05/2023] Open
Abstract
Background Sepsis is one of the leading causes of death in intensive care units. Dexmedetomidine is a sedative agent with anti-inflammatory properties. This study is designed to differentiate the impact of two different doses of dexmedetomidine on lung injury induced by sepsis. Methods Adult male Wistar rats were randomly divided into four groups: sham (n = 6), control (n = 12), 5DEX (n = 12), and 10DEX (n = 12). Cecal ligation puncture (CLP) was applied for sepsis initiation. The 5DEX group received 5 μg.kg-1.h-1 and the 10DEX group received 10 μg.kg-1.h-1 dexmedetomidine intravenous infusions for a 1-hour period. Six hours after CLP, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and intercellular adhesion molecule-1 (ICAM-1) levels were analyzed in blood samples. Twenty-four hours after CLP, lung samples from the remaining rats were collected for the measurement of myeloperoxidase (MPO) activity, histological examination, and TdT- (terminal deoxynucleotidyl transferase) mediated fluorescent-dUTP labeling staining for apoptosis detection. Results Serum cytokine release, MPO activity, and apoptosis in the lung were significantly increased in the CLP group compared with the sham and dexmedetomidine groups (p < 0.05). TNF-α, ICAM-1, and MPO were significantly lower in the 10DEX group compared with both 5DEX and control groups, while IL-1β, total injury score, and apoptotic cell count had significantly lower values in both 10DEX and 5DEX groups compared with the control group (p < 0.05). Conclusion Dexmedetomidine administration played a protective role against CLP-induced lung injury. High-dose dexmedetomidine was needed for suppressing the leukocyte-mediated lung injury and apoptosis of lung tissue.
Collapse
Affiliation(s)
- Gülsüm Karabulut
- Gazi University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Ankara, Turkey.
| | - Nurdan Bedirli
- Gazi University, Faculty of Medicine, Department of Anesthesiology and Reanimation, Ankara, Turkey
| | - Nalan Akyürek
- Gazi University, Faculty of Medicine, Department of Pathology, Ankara, Turkey
| | - Emin Ümit Bağrıaçık
- Gazi University, Faculty of Medicine, Department of Immunology, Ankara, Turkey
| |
Collapse
|