1
|
Li Y, Li X, Wu W, Liu P, Liu J, Jiang H, Deng L, Ni C, Wu X, Zhao Y, Ren J. Insights into the roles of macrophages in Klebsiella pneumoniae infections: a comprehensive review. Cell Mol Biol Lett 2025; 30:34. [PMID: 40140770 PMCID: PMC11948646 DOI: 10.1186/s11658-025-00717-7] [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: 08/20/2024] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Klebsiella pneumoniae (KP) infections represent a significant global health challenge, characterized by severe inflammatory sequelae and escalating antimicrobial resistance. This comprehensive review elucidates the complex interplay between macrophages and KP, encompassing pathogen recognition mechanisms, macrophage activation states, cellular death pathways, and emerging immunotherapeutic strategies. We critically analyze current literature on macrophage pattern recognition receptor engagement with KP-associated molecular patterns. The review examines the spectrum of macrophage responses to KP infection, including classical M1 polarization and the newly described M(Kp) phenotype, alongside metabolic reprogramming events such as glycolytic enhancement and immune responsive gene 1 (IRG1)-itaconate upregulation. We systematically evaluate macrophage fate decisions in response to KP, including autophagy, apoptosis, pyroptosis, and necroptosis. Furthermore, we provide a critical assessment of potential future therapeutic modalities. Given the limitations of current treatment paradigms, elucidating macrophage-KP interactions is imperative. Insights gained from this analysis may inform the development of novel immunomodulatory approaches to augment conventional antimicrobial therapies, potentially transforming the clinical management of KP infections.
Collapse
Affiliation(s)
- Yangguang Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuanheng Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenqi Wu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Peizhao Liu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Juanhan Liu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haiyang Jiang
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Liting Deng
- School of Medicine, Southeast University, Nanjing, 210000, China
| | - Chujun Ni
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Xiuwen Wu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Yun Zhao
- Department of General Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210009, China.
- Clinical Translational Research Center for Surgical Infection and Immunity of Nanjing Medical University, Nanjing, China.
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Wiger CW, Ranheim T, Arnesen H, Vaage J, Pischke SE, Yndestad A, Stensløkken K, Torp M. TLR4 Inhibition Attenuated LPS-Induced Proinflammatory Signaling and Cytokine Release in Mouse Hearts and Cardiomyocytes. Immun Inflamm Dis 2025; 13:e70133. [PMID: 39853914 PMCID: PMC11760985 DOI: 10.1002/iid3.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Sepsis is associated with myocardial injury and early mortality. The innate immune receptor Toll-like receptor 4 (TLR4) can recognize pathogen-associated-molecular-patterns (PAMPs) and damage-associated molecular patterns (DAMPs); the latter are released during tissue injury. We hypothesized that TLR4 inhibition reduces proinflammatory signaling and cytokine release in: (1) LPS or Escherichia coli-treated isolated mouse heart; (2) LPS-treated mouse primary adult cardiomyocytes; and (3) the isolated heart during ischemia-reperfusion. METHODS Isolated C57BL/6N male mouse hearts were perfused for 120 min, with either LPS, E. coli, with and without CLI-095 (TLR4 inhibitor). Primary adult mouse cardiomyocytes were treated with LPS or LPS + CLI-095. Isolated hearts, exposed to 35 min of global ischemia, were treated with either vehicle or CLI-095 during reperfusion. Infarct size was quantified by triphenyltetrazolium staining. Cytokine expression was analyzed with ELISA, western blot analysis, and qPCR. RESULTS In isolated hearts, E. coli increased the expression of proinflammatory cytokines (IL-6 and CXCL2), which was not attenuated with TLR4 inhibition. TLR4 inhibition reduced expression (p = 0.004) and release of IL-6 (p < 0.0001) in LPS-exposed isolated hearts. LPS activated the nuclear-factor κ-light-chain-enhancer of activated B cells signaling pathway (NF-κB) in primary adult cardiomyocytes. Moreover, TLR4 inhibition reduced LPS-induced mRNA expression and release of IL-6 in primary adult cardiomyocytes. Isolated hearts treated with CLI-095 during reperfusion after ischemia (induced DAMPs release) showed reduced infarct size (39 ± 17% to 26 ± 8%, p = 0.034) and decreased IL-6 release (p = 0.006). CONCLUSION Inhibition of TLR4 reduced proinflammatory signaling and cytokine release in LPS-treated and ischemia-reperfused isolated mouse hearts and in primary adult murine cardiomyocytes.
Collapse
Affiliation(s)
- Christine W. Wiger
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Henriette Arnesen
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Jarle Vaage
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
- Department of Research and Innovation, Division of Emergencies and Critical CareOslo University HospitalOsloNorway
| | | | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Kåre‐Olav Stensløkken
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - May‐Kristin Torp
- Division of Physiology, Department of Molecular MedicineInstitute of Basic Medical SciencesUniversity of OsloOsloNorway
- Østfold Hospital TrustGrålumNorway
| |
Collapse
|
3
|
Chen Y, Huang D, Xie A, Shan Y, Zhao S, Gao C, Chen J, Shi H, Fang W, Peng J. Capn3b-deficient zebrafish model reveals a key role of autoimmune response in LGMDR1. J Genet Genomics 2024; 51:1375-1388. [PMID: 39349278 DOI: 10.1016/j.jgg.2024.09.011] [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: 05/27/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/02/2024]
Abstract
Mutations in calcium-dependent papain-like protease CALPAIN3 (CAPN3) cause Limb-Girdle Muscular Dystrophy Recessive Type 1 (LGMDR1), the most common limb-girdle muscular dystrophy in humans. In addition to progressive muscle weakness, persistent inflammatory infiltration is also a feature of LGMDR1. Despite the underlying mechanism remaining poorly understood, we consider that it may relate to the newly defined role of CAPN3/Capn3b in the nucleolus. Here, we report that the loss of function of zebrafish capn3b, the counterpart of human CAPN3, induces an autoimmune response akin to that in LGMDR1 patients. capn3b mutant larvae are more susceptible to Listeria monocytogenes injection, characterized by recruiting more macrophages. Under germ-free conditions, transcriptome analysis of the capn3b mutant muscle reveals a significant upregulation of the chemokine-production-related genes. Coincidently, more neutrophils are recruited to the injury site imposed by either muscle stabbing or tail fin amputation. Nucleolar proteomic analysis and enzymatic assays reveal NKAP, an activating factor of the NF-κB pathway, to be a target of CAPN3. We conclude that the accumulation of Nkap and other factors in the capn3b mutant may be involved in the over-activation of innate immunity. Our studies indicate that the zebrafish capn3b mutant is a powerful model for studying the immunity-related progression of human LGMDR1.
Collapse
Affiliation(s)
- Yayue Chen
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Delai Huang
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Aixuan Xie
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Shan
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shuyi Zhao
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ce Gao
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jun Chen
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hui Shi
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Weihuan Fang
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Jinrong Peng
- MOE Key Laboratory for Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
4
|
Kircheis R, Planz O. Special Issue "The Role of Toll-Like Receptors (TLRs) in Infection and Inflammation 2.0". Int J Mol Sci 2024; 25:9709. [PMID: 39273656 PMCID: PMC11396464 DOI: 10.3390/ijms25179709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Toll-like receptors (TLRs) are key players in the innate immune system, in host' first-line defense against pathogens [...].
Collapse
Affiliation(s)
| | - Oliver Planz
- Institute of Cell Biology and Immunology, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
5
|
Ye H, Zou X, Fang X. Advancing cell-based therapy in sepsis: An anesthesia outlook. Chin Med J (Engl) 2024; 137:1522-1534. [PMID: 38708689 PMCID: PMC11230747 DOI: 10.1097/cm9.0000000000003097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Indexed: 05/07/2024] Open
Abstract
ABSTRACT Sepsis poses a health challenge globally owing to markedly high rates of morbidity and mortality. Despite employing bundle therapy over two decades, approaches including transient organ supportive therapy and clinical trials focusing on signaling pathways have failed in effectively reversing multiple organ failure in patients with sepsis. Prompt and appropriate perioperative management for surgical patients with concurrent sepsis is urgent. Consequently, innovative therapies focusing on remedying organ injuries are necessitated. Cell therapy has emerged as a promising therapeutic avenue for repairing local damage to vital organs and restoring homeostasis during perioperative treatment for sepsis. Given the pivotal role of immune cell responses in the pathogenesis of sepsis, stem cell-based interventions that primarily modulate immune responses by interacting with multiple immune cells have progressed into clinical trials. The strides made in single-cell sequencing and gene-editing technologies have advanced the understanding of disease-specific immune responses in sepsis. Chimeric antigen receptor (CAR)-immune cell therapy offers an intriguing option for the treatment of sepsis. This review provides a concise overview of immune cell therapy, its current status, and the strides made in the context of sepsis research, discussing potential strategies for the management of patients with sepsis during perioperative stages.
Collapse
Affiliation(s)
- Hui Ye
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xiaoyu Zou
- The Children's Hospital, National Clinical Research Center for Child Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 312000, China
| | - Xiangming Fang
- Department of Anesthesiology and Intensive Care, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
6
|
He M, Ye J, Zheng W, Qiao P, Gu H, Qin W, He X. The impact of gender differences on the clinical characteristics of critically ill patients with venous thromboembolism: A retrospective, observational study. Medicine (Baltimore) 2024; 103:e38423. [PMID: 38875425 PMCID: PMC11175867 DOI: 10.1097/md.0000000000038423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 02/29/2024] [Accepted: 05/10/2024] [Indexed: 06/16/2024] Open
Abstract
The aim of this study is to describe the general information, disease and treatment of venous thromboembolism (VTE) in critically ill patients, to explore the characteristics of severe patients with deep venous VTE and provide clinical reference data for the prevention and treatment of VTE in critically ill patients. This study carried out a retrospective study on the medical records of patients who were treated in the intensive care unit of Affiliated Hospital of Jining Medical College from 2020 to 2022. The general data, general conditions, drug use, past history, VTE prevention measures, post-VTE conditions, and Padua risk score of 297 patients with VTE during the period of hospitalization in intensive care unit (ICU) were classified and statistically analyzed. A total of 297 including 171 male and 126 male patient medical records were included in the analysis, we compared multiple clinical indicators between male and female patients. Compared to women, male patients have a higher acute physiology and chronic health evaluation II score(APACHE-II score) (P = .027), a higher state of consciousness (P = .003), a higher rate of smoking and drinking history (P < .001), a lower rate of heart failure (P = .003) and chronic depression (P = .013), and a higher rate of recommended operations for male patients after VTE (P = .031). The prothrombin time (PT) (P = .041) and activated partial thromboplastin time (APTT) (P = .040) of male patients were significantly higher than those of female patients, while triglyceride (P = .009) and total cholesterol (TC) (P = .001) were significantly lower than those of female patients. The difference in D-dimer (P = .739) was not significant. This study shows that male and female patients with VTE in the ICU have significant differences in general characteristics, general clinical conditions, history of past illness, thrombosis character, and examination items, this may be the reason for the different rates of VTE between male and female patients in the ICU.
Collapse
Affiliation(s)
- Meng He
- Department of Prehospital Emergency, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| | - Jiuhang Ye
- Department of Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| | - Weiwei Zheng
- Department of Critical Care Medicine, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| | - Peng Qiao
- Department of Emergency Intensive Care Unit, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| | - Haiyan Gu
- Emergency Internal Medicine Ward, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| | - Wenwen Qin
- Emergency Internal Medicine Ward, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| | - Xuehong He
- Department of Nursing, Affiliated Hospital of Jining Medical University, Jining, Shandong, P. R. China
| |
Collapse
|
7
|
Huang S, Perry A, Sanchez Parra C, Gonzalez Torriente A, Ghumman H, Charkowick S, Colon J, Heide M, Jaglal M, Mhaskar R, Rico JF. Incidence of Thrombosis in COVID-19 Patients Compared to Non-COVID-19 Sepsis Patients in the Intensive Care Unit. J Clin Med 2024; 13:2974. [PMID: 38792515 PMCID: PMC11121895 DOI: 10.3390/jcm13102974] [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: 11/12/2023] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Background/Objectives: The hypercoagulable state associated with COVID-19 infection is associated with adverse outcomes and mortality. Studies have also demonstrated high rates of venous thromboembolism (VTE) events among patients with sepsis. We aimed to evaluate how the increase in thrombotic events in critically ill patients with COVID-19 infection compares to that of critically ill patients with non-COVID-19 sepsis. Methods: A chart review was performed of patients 18 years or older admitted to the intensive care unit (ICU) at Tampa General Hospital between 1 January 2020 and 31 December 2020 diagnosed with COVID-19 or sepsis secondary to other pathogens. Non-COVID-19 sepsis patients and COVID-19 patients were propensity-matched 3:1 on the Charlson Comorbidity Index. Multivariate analyses adjusting for confounding were conducted to report odds ratio (OR) and 95% confidence intervals (95% CIs) of predictors for thrombotic events and overall mortality. Results: After propensity score matching, 492 sepsis patients and 164 COVID-19 patients were included in the analysis. COVID-19 patients were significantly older (p = 0.021) and showed higher BMI (p < 0.001) than sepsis patients. COVID-19 patients did not show significantly higher odds of thrombosis after adjustment for confounders (OR 0.85, 95% CI 0.42-1.72), but had significantly lower odds of mortality than sepsis patients (OR 0.33, 95% CI 0.16-0.66). Conclusions: Our results suggest that further study is required to lower the rate of VTE in COVID-19 and non-COVID-19 sepsis patients admitted to the ICU; it is also reasonable to consider similar thromboembolism practices between these two patient groups.
Collapse
Affiliation(s)
- Sherri Huang
- Department of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL 33606, USA
| | - Ashley Perry
- Department of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL 33606, USA
| | - Carlos Sanchez Parra
- Department of Internal Medicine and Pediatrics, University of South Florida, Tampa, FL 33606, USA
- Division of Pediatric Cardiology, Texas Children’s Hospital, Houston, TX 77030, USA
| | | | - Haider Ghumman
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Shaun Charkowick
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Joshua Colon
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - McKenzi Heide
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Michael Jaglal
- Department of Hematology, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Hematology and Oncology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Rahul Mhaskar
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Juan Felipe Rico
- Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, FL 33606, USA
| |
Collapse
|
8
|
Kok CR, Bram Z, Thissen JB, Horseman TS, Fong KSK, Reichert-Scrivner SA, Paguirigan C, O'Connor K, Thompson K, Scheiber AE, Mabery S, Ngauy V, Uyehara CF, Be NA. The military gear microbiome: risk factors surrounding the warfighter. Appl Environ Microbiol 2024; 90:e0117623. [PMID: 38170999 PMCID: PMC10807412 DOI: 10.1128/aem.01176-23] [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: 07/11/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024] Open
Abstract
Combat extremity wounds are highly susceptible to contamination from surrounding environmental material. This bioburden could be partially transferred from materials in immediate proximity to the wound, including fragments of the uniform and gear. However, the assessment of the microbial bioburden present on military gear during operational conditions of deployment or training is relatively unexplored. Opportunistic pathogens that can survive on gear represent risk factors for infection following injury, especially following combat blasts, where fibers and other materials are embedded in wounded tissue. We utilized 16S rRNA sequencing to assess the microbiome composition of different military gear types (boot, trouser, coat, and canteen) from two operational environments (training in Hawai'i and deployed in Indonesia) across time (days 0 and 14). We found that microbiome diversity, stability, and composition were dependent on gear type, training location, and sampling timepoint. At day 14, species diversity was significantly higher in Hawai'i samples compared to Indonesia samples for boot, coat, and trouser swabs. In addition, we observed the presence of potential microbial risk factors, as opportunistic pathogenic species, such as Acinetobacter, Pseudomonas, and Staphylococcus, were found to be present in all sample types and in both study sites. These study outcomes will be used to guide the design of antimicrobial materials and uniforms and for infection control efforts following combat blasts and other injuries, thereby improving treatment guidance during military training and deployment.IMPORTANCECombat extremity wounds are vulnerable to contamination from environments of proximity to the warfighter, leading to potential detrimental outcomes such as infection and delayed wound healing. Therefore, microbial surveillance of such environments is necessary to aid the advancement of military safety and preparedness through clinical diagnostics, treatment protocols, and uniform material design.
Collapse
Affiliation(s)
- Car Reen Kok
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, USA
| | | | - James B. Thissen
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Timothy S. Horseman
- Tripler Army Medical Center, Honolulu, Hawaii, USA
- School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | | | | | | | | | | | | | - Shalini Mabery
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, USA
| | - Viseth Ngauy
- Tripler Army Medical Center, Honolulu, Hawaii, USA
| | | | - Nicholas A. Be
- Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, California, USA
| |
Collapse
|
9
|
Ionescu (Miron) AI, Atasiei DI, Ionescu RT, Ultimescu F, Barnonschi AA, Anghel AV, Anghel CA, Antone-Iordache IL, Mitre R, Bobolocu AM, Zamfir A, Lișcu HD, Coniac S, Șandru F. Prediction of Subclinical and Clinical Multiple Organ Failure Dysfunction in Breast Cancer Patients-A Review Using AI Tools. Cancers (Basel) 2024; 16:381. [PMID: 38254870 PMCID: PMC11154446 DOI: 10.3390/cancers16020381] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
This review explores the interconnection between precursor lesions of breast cancer (typical ductal hyperplasia, atypical ductal/lobular hyperplasia) and the subclinical of multiple organ failure syndrome, both representing early stages marked by alterations preceding clinical symptoms, undetectable through conventional diagnostic methods. Addressing the question "Why patients with breast cancer exhibit a tendency to deteriorate", this study investigates the biological progression from a subclinical multiple organ failure syndrome, characterized by insidious but indisputable lesions, to an acute (clinical) state resembling a cascade akin to a waterfall or domino effect, often culminating in the patient's demise. A comprehensive literature search was conducted using PubMed, Google Scholar, and Scopus databases in October 2023, employing keywords such as "MODS", "SIRS", "sepsis", "pathophysiology of MODS", "MODS in cancer patients", "multiple organ failure", "risk factors", "cancer", "ICU", "quality of life", and "breast cancer". Supplementary references were extracted from the retrieved articles. This study emphasizes the importance of early identification and prevention of the multiple organ failure cascade at the inception of the malignant state, aiming to enhance the quality of life and extend survival. This pursuit contributes to a deeper understanding of risk factors and viable therapeutic options. Despite the existence of the subclinical multiple organ failure syndrome, current diagnostic methodologies remain inadequate, prompting consideration of AI as an increasingly crucial tool for early identification in the diagnostic process.
Collapse
Affiliation(s)
- Andreea-Iuliana Ionescu (Miron)
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
- Department of Medical Oncology, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Dimitrie-Ionut Atasiei
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Radu-Tudor Ionescu
- Department of Computer Science, University of Bucharest, 010041 Bucharest, Romania;
| | - Flavia Ultimescu
- Department of Pathology, Institute of Oncology “Prof. Dr. Alexandru Trestioreanu”, 022328 Bucharest, Romania;
- Department of Pathological Anatomy, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andrei-Alexandru Barnonschi
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Alexandra-Valentina Anghel
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Cătălin-Alexandru Anghel
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Ionuț-Lucian Antone-Iordache
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Ruxandra Mitre
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
- Department of Medical Oncology, Colțea Clinical Hospital, 030167 Bucharest, Romania;
| | - Alexandra Maria Bobolocu
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Andreea Zamfir
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
| | - Horia-Dan Lișcu
- Department of Oncological Radiotherapy and Medical Imaging, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania; (A.-I.I.); (A.-A.B.); (A.-V.A.); (C.-A.A.); (I.-L.A.-I.); (R.M.); (A.M.B.); (A.Z.); (H.-D.L.)
- Department of Radiotherapy, Colțea Clinical Hospital, 030167 Bucharest, Romania
| | - Simona Coniac
- Department of Medical Oncology, Colțea Clinical Hospital, 030167 Bucharest, Romania;
- Department of Endocrinology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Florica Șandru
- Department of Dermatovenerology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
- Department of Dermatology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| |
Collapse
|
10
|
Cicchinelli S, Pignataro G, Gemma S, Piccioni A, Picozzi D, Ojetti V, Franceschi F, Candelli M. PAMPs and DAMPs in Sepsis: A Review of Their Molecular Features and Potential Clinical Implications. Int J Mol Sci 2024; 25:962. [PMID: 38256033 PMCID: PMC10815927 DOI: 10.3390/ijms25020962] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Sepsis is a serious organ dysfunction caused by a dysregulated immune host reaction to a pathogen. The innate immunity is programmed to react immediately to conserved molecules, released by the pathogens (PAMPs), and the host (DAMPs). We aimed to review the molecular mechanisms of the early phases of sepsis, focusing on PAMPs, DAMPs, and their related pathways, to identify potential biomarkers. We included studies published in English and searched on PubMed® and Cochrane®. After a detailed discussion on the actual knowledge of PAMPs/DAMPs, we analyzed their role in the different organs affected by sepsis, trying to elucidate the molecular basis of some of the most-used prognostic scores for sepsis. Furthermore, we described a chronological trend for the release of PAMPs/DAMPs that may be useful to identify different subsets of septic patients, who may benefit from targeted therapies. These findings are preliminary since these pathways seem to be strongly influenced by the peculiar characteristics of different pathogens and host features. Due to these reasons, while initial findings are promising, additional studies are necessary to clarify the potential involvement of these molecular patterns in the natural evolution of sepsis and to facilitate their transition into the clinical setting.
Collapse
Affiliation(s)
- Sara Cicchinelli
- Department of Emergency, S.S. Filippo e Nicola Hospital, 67051 Avezzano, Italy;
| | - Giulia Pignataro
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Stefania Gemma
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Andrea Piccioni
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Domitilla Picozzi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Veronica Ojetti
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Francesco Franceschi
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli—IRRCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.P.); (S.G.); (A.P.); (D.P.); (V.O.); (F.F.)
| |
Collapse
|
11
|
Kim JH, Han J, Afridi R, Kim JH, Rahman MH, Park DH, Lee WS, Song GJ, Suk K. A multiplexed siRNA screen identifies key kinase signaling networks of brain glia. Life Sci Alliance 2023; 6:e202201605. [PMID: 36878638 PMCID: PMC9990460 DOI: 10.26508/lsa.202201605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The dynamic behaviors of brain glial cells in various neuroinflammatory conditions and neurological disorders have been reported; however, little is known about the underlying intracellular signaling pathways. Here, we developed a multiplexed kinome-wide siRNA screen to identify the kinases regulating several inflammatory phenotypes of mouse glial cells in culture, including inflammatory activation, migration, and phagocytosis of glia. Subsequent proof-of-concept experiments involving genetic and pharmacological inhibitions indicated the importance of T-cell receptor signaling components in microglial activation and a metabolic shift from glycolysis to oxidative phosphorylation in astrocyte migration. This time- and cost-effective multiplexed kinome siRNA screen efficiently provides exploitable drug targets and novel insight into the mechanisms underlying the phenotypic regulation of glial cells and neuroinflammation. Moreover, the kinases identified in this screen may be relevant in other inflammatory diseases and cancer, wherein kinases play a critical role in disease signaling pathways.
Collapse
Affiliation(s)
- Jong-Heon Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Han
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ruqayya Afridi
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Hong Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Won Suk Lee
- Neuracle Science Co., Ltd. Seoul, Republic Korea
| | - Gyun Jee Song
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung-si, Republic Korea; Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic Korea
| | - Kyoungho Suk
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
12
|
Maisat W, Yuki K. Narrative review of systemic inflammatory response mechanisms in cardiac surgery and immunomodulatory role of anesthetic agents. Ann Card Anaesth 2023; 26:133-142. [PMID: 37706376 PMCID: PMC10284469 DOI: 10.4103/aca.aca_147_22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/05/2022] [Accepted: 12/18/2022] [Indexed: 09/15/2023] Open
Abstract
Although surgical techniques and perioperative care have made significant advances, perioperative mortality in cardiac surgery remains relatively high. Single- or multiple-organ failure remains the leading cause of postoperative mortality. Systemic inflammatory response syndrome (SIRS) is a common trigger for organ injury or dysfunction in surgical patients. Cardiac surgery involves major surgical dissection, the use of cardiopulmonary bypass (CPB), and frequent blood transfusions. Ischemia-reperfusion injury and contact activation from CPB are among the major triggers for SIRS. Blood transfusion can also induce proinflammatory responses. Here, we review the immunological mechanisms of organ injury and the role of anesthetic regimens in cardiac surgery.
Collapse
Affiliation(s)
- Wiriya Maisat
- Division of Cardiac Anesthesia, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, USA
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Immunology, Harvard Medical School, Boston, USA
- Department of Anesthesiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Koichi Yuki
- Division of Cardiac Anesthesia, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, USA
- Department of Anaesthesia, Harvard Medical School, Boston, USA
- Department of Immunology, Harvard Medical School, Boston, USA
| |
Collapse
|
13
|
Ondee T, Pongpirul K, Udompornpitak K, Sukkummee W, Lertmongkolaksorn T, Senaprom S, Leelahavanichkul A. High Fructose Causes More Prominent Liver Steatohepatitis with Leaky Gut Similar to High Glucose Administration in Mice and Attenuation by Lactiplantibacillus plantarum dfa1. Nutrients 2023; 15:1462. [PMID: 36986190 PMCID: PMC10056651 DOI: 10.3390/nu15061462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
High-sugar diet-induced prediabetes and obesity are a global current problem that can be the result of glucose or fructose. However, a head-to-head comparison between both sugars on health impact is still lacking, and Lactiplantibacillus plantarum dfa1 has never been tested, and has recently been isolated from healthy volunteers. The mice were administered with the high glucose or fructose preparation in standard mouse chaw with or without L. plantarum dfa1 gavage, on alternate days, and in vitro experiments were performed using enterocyte cell lines (Caco2) and hepatocytes (HepG2). After 12 weeks of experiments, both glucose and fructose induced a similar severity of obesity (weight gain, lipid profiles, and fat deposition at several sites) and prediabetes condition (fasting glucose, insulin, oral glucose tolerance test, and Homeostatic Model Assessment for Insulin Resistance (HOMA score)). However, fructose administration induced more severe liver damage (serum alanine transaminase, liver weight, histology score, fat components, and oxidative stress) than the glucose group, while glucose caused more prominent intestinal permeability damage (FITC-dextran assay) and serum cytokines (TNF-α, IL-6, and IL-10) compared to the fructose group. Interestingly, all of these parameters were attenuated by L. plantarum dfa1 administration. Because there was a subtle change in the analysis of the fecal microbiome of mice with glucose or fructose administration compared to control mice, the probiotics altered only some microbiome parameters (Chao1 and Lactobacilli abundance). For in vitro experiments, glucose induced more damage to high-dose lipopolysaccharide (LPS) (1 µg/mL) to enterocytes (Caco2 cell) than fructose, as indicated by transepithelial electrical resistance (TEER), supernatant cytokines (TNF-α and IL-8), and glycolysis capacity (by extracellular flux analysis). Meanwhile, both glucose and fructose similarly facilitated LPS injury in hepatocytes (HepG2 cell) as evaluated by supernatant cytokines (TNF-α, IL-6, and IL-10) and extracellular flux analysis. In conclusion, glucose possibly induced a more severe intestinal injury (perhaps due to LPS-glucose synergy) and fructose caused a more prominent liver injury (possibly due to liver fructose metabolism), despite a similar effect on obesity and prediabetes. Prevention of obesity and prediabetes with probiotics was encouraged.
Collapse
Affiliation(s)
- Thunnicha Ondee
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Krit Pongpirul
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- School of Global Health, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Clinical Research Center, Bumrungrad International Hospital, Bangkok 10110, Thailand
- Department of Infection Biology & Microbiomes, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3GB, UK
| | - Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Warumphon Sukkummee
- Center of Excellence in Clinical Pharmacokinetics and Pharmacogenomics, Department of Pharmacology, Faculty of Medicine Chulalongkorn University, Bangkok 10330, Thailand
| | - Thanapat Lertmongkolaksorn
- Research Management and Development Division, Office of the President, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Sayamon Senaprom
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology Research Unit (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
14
|
Skulachev VP, Vyssokikh MY, Chernyak BV, Averina OA, Andreev-Andrievskiy AA, Zinovkin RA, Lyamzaev KG, Marey MV, Egorov MV, Frolova OJ, Zorov DB, Skulachev MV, Sadovnichii VA. Mitochondrion-targeted antioxidant SkQ1 prevents rapid animal death caused by highly diverse shocks. Sci Rep 2023; 13:4326. [PMID: 36922552 PMCID: PMC10017827 DOI: 10.1038/s41598-023-31281-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
The response to stress involves the activation of pathways leading either to protection from the stress origin, eventually resulting in development of stress resistance, or activation of the rapid death of the organism. Here we hypothesize that mitochondrial reactive oxygen species (mtROS) play a key role in stress-induced programmed death of the organism, which we called "phenoptosis" in 1997. We demonstrate that the synthetic mitochondria-targeted antioxidant SkQ1 (which specifically abolishes mtROS) prevents rapid death of mice caused by four mechanistically very different shocks: (a) bacterial lipopolysaccharide (LPS) shock, (b) shock in response to intravenous mitochondrial injection, (c) cold shock, and (d) toxic shock caused by the penetrating cation C12TPP. Importantly, under all these stresses mortality was associated with a strong elevation of the levels of pro-inflammatory cytokines and administration of SkQ1 was able to switch off the cytokine storms. Since the main effect of SkQ1 is the neutralization of mtROS, this study provides evidence for the role of mtROS in the activation of innate immune responses mediating stress-induced death of the organism. We propose that SkQ1 may be used clinically to support patients in critical conditions, such as septic shock, extensive trauma, cooling, and severe infection by bacteria or viruses.
Collapse
Affiliation(s)
- V P Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.
| | - M Yu Vyssokikh
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.
| | - B V Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991. .,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991.
| | - O A Averina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - A A Andreev-Andrievskiy
- Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991.,Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - R A Zinovkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - K G Lyamzaev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - M V Marey
- Research Center for Obstetrics, Gynecology and Perinatology, Moscow, Russia, 117198
| | - M V Egorov
- Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - O J Frolova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991.,Institute of Mitoengineering, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - D B Zorov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - M V Skulachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia, 119991
| | - V A Sadovnichii
- Faculty of Mechanics and Mathematics, Lomonosov Moscow State University, Moscow, Russia, 119991
| |
Collapse
|
15
|
Abstract
ABSTRACT Sepsis and trauma remain the leading causes of morbidity and mortality. Our understanding of the molecular pathogenesis in the development of multiple organ dysfunction in sepsis and trauma has evolved as more focus is on secondary injury from innate immunity, inflammation, and the potential role of endogenous danger molecules. Studies of the past several decades have generated evidence for extracellular RNAs (exRNAs) as biologically active mediators in health and disease. Here, we review studies on plasma exRNA profiling in mice and humans with sepsis and trauma, the role and mode of action by exRNAs, such as ex-micro(mi)RNAs, in host innate immune response, and their potential implications in various organ injury during sepsis and trauma.
Collapse
Affiliation(s)
- Williams Brittney
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rosemary Kozar
- Shock Trauma Center and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Chao Wei
- Translational Research Program, Department of Anesthesiology, and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
16
|
Miyashita D, Inoue R, Tsuno T, Okuyama T, Kyohara M, Nakahashi-Oda C, Nishiyama K, Fukushima S, Inada Y, Togashi Y, Shibuya A, Terauchi Y, Shirakawa J. Protective effects of S100A8 on sepsis mortality: Links to sepsis risk in obesity and diabetes. iScience 2022; 25:105662. [PMID: 36505926 PMCID: PMC9732389 DOI: 10.1016/j.isci.2022.105662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/23/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Obesity and diabetes are independent risk factors for death during sepsis. S100A8, an alarmin, is related to inflammation, obesity, and diabetes. Here, we examine the role of S100A8 in sepsis of obesity and diabetes models. Injection of S100A8 prolongs the survival of septic mice induced by lethal endotoxemia, Escherichia coli injection, or cecal ligation and puncture. S100A8 decrease the LPS-induced expression of proinflammatory cytokines in peritoneal macrophages by inhibiting TLR4-mediated signals in an autocrine manner. db/db, ob/ob, and western diet-fed mice demonstrate reduced upregulation of S100A8 induced by LPS treatment in both serum and peritoneal cells. These mice also show shorter survival after LPS injection, and S100A8 supplementation prolonged the survival. While myelomonocytic cells-specific S100A8-deficient mice (Lyz2 cre :S100A8 floxed/floxed ) exhibit shorter survival after LPS treatment, S100A8 supplementation prolonged the survival. Thus, myelomonocytic cell-derived S100A8 is crucial for protection from sepsis, and S100A8 supplementation improves sepsis, particularly in mice with obesity and diabetes.
Collapse
Affiliation(s)
- Daisuke Miyashita
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Ryota Inoue
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Takahiro Tsuno
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Tomoko Okuyama
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Mayu Kyohara
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Chigusa Nakahashi-Oda
- Department of Immunology, Faculty of Medicine, and R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Japan
| | - Kuniyuki Nishiyama
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Setsuko Fukushima
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
| | - Yutaro Inada
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Yu Togashi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, and R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
| | - Jun Shirakawa
- Laboratory of Diabetes and Metabolic Disorders, Institute for Molecular and Cellular Regulation (IMCR), Gunma University, Maebashi, Japan
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama-City University, Yokohama, Japan
- Corresponding author
| |
Collapse
|
17
|
Udompornpitak K, Charoensappakit A, Sae-Khow K, Bhunyakarnjanarat T, Dang CP, Saisorn W, Visitchanakun P, Phuengmaung P, Palaga T, Ritprajak P, Tungsanga S, Leelahavanichkul A. Obesity Exacerbates Lupus Activity in Fc Gamma Receptor IIb Deficient Lupus Mice Partly through Saturated Fatty Acid-Induced Gut Barrier Defect and Systemic Inflammation. J Innate Immun 2022; 15:240-261. [PMID: 36219976 PMCID: PMC10643905 DOI: 10.1159/000526206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/21/2022] [Indexed: 11/19/2022] Open
Abstract
The prevalence of obesity is increasing, and the coexistence of obesity and systemic lupus erythematosus (lupus) is possible. A high-fat diet (HFD) was orally administered for 6 months in female 8-week-old Fc gamma receptor IIb deficient (FcgRIIb-/-) lupus or age and gender-matched wild-type (WT) mice. Lupus nephritis (anti-dsDNA, proteinuria, and increased creatinine), gut barrier defect (fluorescein isothiocyanate dextran), serum lipopolysaccharide (LPS), serum interleukin (IL)-6, liver injury (alanine transaminase), organ fibrosis (liver and kidney pathology), spleen apoptosis (activated caspase 3), and aorta thickness (but not weight gain and lipid profiles) were more prominent in HFD-administered FcgRIIb-/- mice than the obese WT, without injury in regular diet-administered mice (both FcgRIIb-/- and WT). In parallel, combined palmitic acid (PA; a saturated fatty acid) with LPS (PA + LPS) induced higher tumor necrotic factor-α, IL-6, and IL-10 in the supernatant, inflammatory genes (inducible nitric oxide synthase and IL-1β), reactive oxygen species (dihydroethidium), and glycolysis with reduced mitochondrial activity (extracellular flux analysis) when compared with the activation by each molecule alone in both FcgRIIb-/- and WT macrophages. However, the alterations of these parameters were more prominent in PA + LPS-administered FcgRIIb-/- than in the WT cells. In conclusion, obesity accelerated inflammation in FcgRIIb-/- mice, partly due to the more potent responses from the loss of inhibitory FcgRIIb against PA + LPS with obesity-induced gut barrier defect.
Collapse
Affiliation(s)
- Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Awirut Charoensappakit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kritsanawan Sae-Khow
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Cong Phi Dang
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Peerapat Visitchanakun
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornpimol Phuengmaung
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Patcharee Ritprajak
- Research Unit in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Somkanya Tungsanga
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of General Internal Medicine, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence on Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
18
|
Lampova B, Doskocil I, Kourimska L, Kopec A. N-3 polyunsaturated fatty acids may affect the course of COVID-19. Front Immunol 2022; 13:957518. [PMID: 36238306 PMCID: PMC9551352 DOI: 10.3389/fimmu.2022.957518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
The highly infectious coronavirus disease (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is a new coronavirus that has been spreading since late 2019 and has caused millions of deaths worldwide. COVID-19 continues to spread rapidly worldwide despite high vaccination coverage; therefore, it is crucial to focus on prevention. Most patients experience only mild symptoms of COVID-19. However, in some cases, serious complications can develop mainly due to an exaggerated immune response; that is, a so-called cytokine storm, which can lead to acute respiratory distress syndrome, organ failure, or, in the worst cases, death. N-3 polyunsaturated fatty acids and their metabolites can modulate inflammatory responses, thus reducing the over-release of cytokines. It has been hypothesized that supplementation of n-3 polyunsaturated fatty acids could improve clinical outcomes in critically ill COVID-19 patients. Some clinical trials have shown that administering n-3 polyunsaturated fatty acids to critically ill patients can improve their health and shorten the duration of their stay in intensive care. However, previous clinical studies have some limitations; therefore, further studies are required to confirm these findings.
Collapse
Affiliation(s)
- Barbora Lampova
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Ivo Doskocil
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
- *Correspondence: Ivo Doskocil,
| | - Lenka Kourimska
- Department of Microbiology, Nutrition and Dietetics, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Aneta Kopec
- Department of Human Nutrition and Dietetics, Faculty of Food Technology, The University of Agriculture in Krakow, Krakow, Poland
| |
Collapse
|
19
|
Schena CA, de’Angelis GL, Carra MC, Bianchi G, de’Angelis N. Antimicrobial Challenge in Acute Care Surgery. Antibiotics (Basel) 2022; 11:1315. [PMID: 36289973 PMCID: PMC9598495 DOI: 10.3390/antibiotics11101315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/24/2022] [Accepted: 09/26/2022] [Indexed: 12/07/2022] Open
Abstract
The burden of infections in acute care surgery (ACS) is huge. Surgical emergencies alone account for three million admissions per year in the United States (US) with estimated financial costs of USD 28 billion per year. Acute care facilities and ACS patients represent boost sanctuaries for the emergence, development and transmission of infections and multi-resistant organisms. According to the World Health Organization, healthcare-associated infections affected around 4 million cases in Europe and 1.7 million in the US alone in 2011 with 39,000 and 99,000 directly attributable deaths, respectively. In this scenario, antimicrobial resistance arose as a public-health emergency that worsens patients' morbidity and mortality and increases healthcare costs. The optimal patient care requires the application of comprehensive evidence-based policies and strategies aiming at minimizing the impact of healthcare associated infections and antimicrobial resistance, while optimizing the treatment of intra-abdominal infections. The present review provides a snapshot of two hot topics, such as antimicrobial resistance and systemic inflammatory response, and three milestones of infection management, such as source control, infection prevention, and control and antimicrobial stewardship.
Collapse
Affiliation(s)
- Carlo Alberto Schena
- Unit of Digestive and HPB Surgery, CARE Department, Henri Mondor Hospital, AP-HP, 94010 Créteil, France
| | - Gian Luigi de’Angelis
- Gastroenterology and Endoscopy Unit, Department of Medicine and Surgery, University Hospital of Parma, 43126 Parma, Italy
| | - Maria Clotilde Carra
- Rothschild Hospital, AP-HP, Université Paris Cité, U.F.R. of Odontology, 75006 Paris, France
| | - Giorgio Bianchi
- Unit of Digestive and HPB Surgery, CARE Department, Henri Mondor Hospital, AP-HP, 94010 Créteil, France
| | - Nicola de’Angelis
- Unit of Digestive and HPB Surgery, CARE Department, Henri Mondor Hospital, AP-HP, 94010 Créteil, France
| |
Collapse
|
20
|
Hartel JC, Merz N, Grösch S. How sphingolipids affect T cells in the resolution of inflammation. Front Pharmacol 2022; 13:1002915. [PMID: 36176439 PMCID: PMC9513432 DOI: 10.3389/fphar.2022.1002915] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
The concept of proper resolution of inflammation rather than counteracting it, gained a lot of attention in the past few years. Re-assembly of tissue and cell homeostasis as well as establishment of adaptive immunity after inflammatory processes are the key events of resolution. Neutrophiles and macrophages are well described as promotors of resolution, but the role of T cells is poorly reviewed. It is also broadly known that sphingolipids and their imbalance influence membrane fluidity and cell signalling pathways resulting in inflammation associated diseases like inflammatory bowel disease (IBD), atherosclerosis or diabetes. In this review we highlight the role of sphingolipids in T cells in the context of resolution of inflammation to create an insight into new possible therapeutical approaches.
Collapse
Affiliation(s)
- Jennifer Christina Hartel
- Institute of Clinical Pharmacology, Goethe-University Frankfurt. Frankfurt am Main, Frankfurt, Germany
- Department of Life Sciences, Goethe-University Frankfurt, Frankfurt, Germany
| | - Nadine Merz
- Institute of Clinical Pharmacology, Goethe-University Frankfurt. Frankfurt am Main, Frankfurt, Germany
| | - Sabine Grösch
- Institute of Clinical Pharmacology, Goethe-University Frankfurt. Frankfurt am Main, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
- *Correspondence: Sabine Grösch,
| |
Collapse
|
21
|
Lupu L, Horst K, Greven J, Mert Ü, Ludviksen JA, Pettersen K, Lau C, Li Y, Palmer A, Qin K, Zhang X, Mayer B, van Griensven M, Huber-Lang M, Hildebrand F, Mollnes TE. Simultaneous C5 and CD14 inhibition limits inflammation and organ dysfunction in pig polytrauma. Front Immunol 2022; 13:952267. [PMID: 36059503 PMCID: PMC9433645 DOI: 10.3389/fimmu.2022.952267] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Dysfunctional complement activation and Toll-like receptor signaling immediately after trauma are associated with development of trauma-induced coagulopathy and multiple organ dysfunction syndrome. We assessed the efficacy of the combined inhibition therapy of complement factor C5 and the TLR co-receptor CD14 on thrombo-inflammation and organ damage in an exploratory 72-h polytrauma porcine model, conducted under standard surgical and intensive care management procedures. Twelve male pigs were subjected to polytrauma, followed by resuscitation (ATLS® guidelines) and operation of the femur fracture (intramedullary nailing technique). The pigs were allocated to combined C5 and CD14 inhibition therapy group (n=4) and control group (n=8). The therapy group received intravenously C5 inhibitor (RA101295) and anti-CD14 antibody (rMil2) 30 min post-trauma. Controls received saline. Combined C5 and CD14 inhibition reduced the blood levels of the terminal complement complex (TCC) by 70% (p=0.004), CRP by 28% (p=0.004), and IL-6 by 52% (p=0.048). The inhibition therapy prevented the platelet consumption by 18% and TAT formation by 77% (p=0.008). Moreover, the norepinephrine requirements in the treated group were reduced by 88%. The inhibition therapy limited the organ damage, thereby reducing the blood lipase values by 50% (p=0.028), LDH by 30% (p=0.004), AST by 33%, and NGAL by 30%. Immunofluorescent analysis of the lung tissue revealed C5b-9 deposition on blood vessels in five from the untreated, and in none of the treated animals. In kidney and liver, the C5b-9 deposition was similarly detected mainly the untreated as compared to the treated animals. Combined C5 and CD14 inhibition limited the inflammatory response, the organ damage, and reduced the catecholamine requirements after experimental polytrauma and might be a promising therapeutic approach.
Collapse
Affiliation(s)
- Ludmila Lupu
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Klemens Horst
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Johannes Greven
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Ümit Mert
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | | | | | - Corinna Lau
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway
| | - Yang Li
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Kang Qin
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Xing Zhang
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Martijn van Griensven
- Department Cell Biology-Inspired Tissue Engineering (cBITE), MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma Immunology, University Hospital Ulm, Ulm, Germany
| | - Frank Hildebrand
- Department of Orthopedics, Trauma and Reconstructive Surgery, Rheinisch-Westfalische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital Bodø, Bodø, Norway
- Department of Immunology, Oslo University Hospital, and University of Oslo, Oslo, Norway
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
- *Correspondence: Tom Eirik Mollnes,
| |
Collapse
|
22
|
Charoensappakit A, Sae-Khow K, Leelahavanichkul A. Gut Barrier Damage and Gut Translocation of Pathogen Molecules in Lupus, an Impact of Innate Immunity (Macrophages and Neutrophils) in Autoimmune Disease. Int J Mol Sci 2022; 23:ijms23158223. [PMID: 35897790 PMCID: PMC9367802 DOI: 10.3390/ijms23158223] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/08/2023] Open
Abstract
The gut barrier is a single cell layer that separates gut micro-organisms from the host, and gut permeability defects result in the translocation of microbial molecules from the gut into the blood. Despite the silent clinical manifestation, gut translocation of microbial molecules can induce systemic inflammation that might be an endogenous exacerbating factor of systemic lupus erythematosus. In contrast, circulatory immune-complex deposition and the effect of medications on the gut, an organ with an extremely large surface area, of patients with active lupus might cause gut translocation of microbial molecules, which worsens lupus severity. Likewise, the imbalance of gut microbiota may initiate lupus and/or interfere with gut integrity which results in microbial translocation and lupus exacerbation. Moreover, immune hyper-responsiveness of innate immune cells (macrophages and neutrophils) is demonstrated in a lupus model from the loss of inhibitory Fc gamma receptor IIb (FcgRIIb), which induces prominent responses through the cross-link between activating-FcgRs and innate immune receptors. The immune hyper-responsiveness can cause cell death, especially apoptosis and neutrophil extracellular traps (NETosis), which possibly exacerbates lupus, partly through the enhanced exposure of the self-antigens. Leaky gut monitoring and treatments (such as probiotics) might be beneficial in lupus. Here, we discuss the current information on leaky gut in lupus.
Collapse
Affiliation(s)
- Awirut Charoensappakit
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kritsanawan Sae-Khow
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
23
|
Zhao C, Deng H, Chen X. Harnessing immune response using reactive oxygen Species-Generating/Eliminating inorganic biomaterials for disease treatment. Adv Drug Deliv Rev 2022; 188:114456. [PMID: 35843505 DOI: 10.1016/j.addr.2022.114456] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/27/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022]
Abstract
With the increasing understanding of various biological functions mediated by reactive oxygen species (ROS) in the immune system, a number of studies have been designed to develop ROS-generating/eliminating strategies to selectively modulate immunogenicity for disease treatment. These strategies potentially exploit ROS-modulating inorganic biomaterials to harness host immunity to maximize the therapeutic potency by eliciting a favorable immune response. Inorganic biomaterial-guided in vivo ROS scavenging can exhibit several effects to: i) reduce the secretion of pro-inflammatory factors, ii) induce the phenotypic transition of macrophages from inflammatory M1 to immunosuppressive M2 phase, iii) minimize the recruitment and infiltration of immune cells. and/or iv) suppress the activation of nuclear factor kappa-B (NF-κB) pathway. Inversely, ROS-generating inorganic biomaterials have been found to be capable of: i) inducing immunogenic cell death (ICD), ii) reprograming tumor-associated macrophages from M2 to M1 phenotypes, iii) activating inflammasomes to stimulate tumor immunogenicity, and/or iv) recruiting phagocytes for antimicrobial therapy. This review provides a systematic and up-to-date overview on the progress related to ROS-nanotechnology mediated immunomodulation. We highlight how the ROS-generating/eliminating inorganic biomaterials can converge with immunomodulation and ultimately elicit an effective immune response against inflammation, autoimmune diseases, and/or cancers. We expect that contents presented in this review will be beneficial for the future advancements of ROS-based nanotechnology and its potential applications in this evolving field.
Collapse
Affiliation(s)
- Caiyan Zhao
- Engineering Research Center of Molecular & Neuroimaging, Ministry of Education School of Life Science and Technology, Xidian University, Xi'an, Shaanxi 710126, China
| | - Hongzhang Deng
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore; Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore 119074, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| |
Collapse
|
24
|
Du HB, Jiang SB, Zhao ZA, Zhang H, Zhang LM, Wang Z, Guo YX, Zhai JY, Wang P, Zhao ZG, Niu CY, Jiang LN. TLR2/TLR4-Enhanced TIPE2 Expression Is Involved in Post-Hemorrhagic Shock Mesenteric Lymph-Induced Activation of CD4+T Cells. Front Immunol 2022; 13:838618. [PMID: 35572554 PMCID: PMC9101470 DOI: 10.3389/fimmu.2022.838618] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
Purpose Post hemorrhagic shock mesenteric lymph (PHSML) return contributes to CD4+ T cell dysfunction, which leads to immune dysfunction and uncontrolled inflammatory response. Tumor necrosis factor α induced protein 8 like-2 (TIPE2) is one of the essential proteins to maintain the immune homeostasis. This study investigated the role of TIPE2 in regulation of CD4+ T lymphocyte function in interaction of PHSML and TLR2/TLR4. Methods The splenic CD4+ T cells were isolated from various mice (WT, TLR2-/-, TLR4-/-) by immunomagnetic beads, and stimulated with PHSML, normal lymphatic fluid (NML), respectively. Application of TIPE2-carrying interfering fragments of lentivirus were transfected to WT, TLR4-/-, and TLR2-/- CD4+ T cells, respectively. After interference of TIPE2, they were stimulated with PHSML and NML for the examinations of TIPE2, TLR2, and TLR4 mRNA expressions, proliferation, activation molecules on surface, and cytokine secretion function. Results PHSML stimulation significantly upregulated TIPE2, TLR2, and TLR4 mRNA expressions, decreased proliferation, CD25 expression, and IFN-γ secretion, and increased the secretion ability of IL-4 in WT CD4+ T cells. TIPE2 silencing enhanced proliferative capacity, upregulated CD25 expression, and increased IFNγ secretion in CD4+ T cells. PHSML stimulated TLR2-/-CD4+ T or TLR4-/-CD4+ T cells of which TIPE2 were silenced. TLR2 or TLR4 knockout attenuated PHSML-induced CD4+ T cells dysfunction; PHSML stimulation of silent TIPE2-expressing TLR2-/-CD4+ T or TLR4-/-CD4+ T revealed that the coexistence of low TIPE2 expression with lack of TLR2 or TLR4 eliminated this beneficial effect. Conclusion TIPE2 improves the PHSML-mediated CD4+T cells dysfunction by regulating TLR2/TLR4 pathway, providing a new intervention target following hemorrhagic shock-induced immune dysfunction.
Collapse
Affiliation(s)
- Hui-Bo Du
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| | - Sun-Ban Jiang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Zhen-Ao Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| | - Hong Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| | - Li-Min Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| | - Zhao Wang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Ya-Xiong Guo
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| | - Jia-Yi Zhai
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Peng Wang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| | - Chun-Yu Niu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Li-Na Jiang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, China
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, China
- Key Laboratory of Microcirculation and Shock in Zhangjiakou City, Zhangjiakou, China
| |
Collapse
|
25
|
Naqvi I, Giroux N, Olson L, Morrison SA, Llanga T, Akinade TO, Zhu Y, Zhong Y, Bose S, Arvai S, Abramson K, Chen L, Que L, Kraft B, Shen X, Lee J, Leong KW, Nair SK, Sullenger B. DAMPs/PAMPs induce monocytic TLR activation and tolerance in COVID-19 patients; nucleic acid binding scavengers can counteract such TLR agonists. Biomaterials 2022; 283:121393. [PMID: 35349874 PMCID: PMC8797062 DOI: 10.1016/j.biomaterials.2022.121393] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022]
Abstract
Millions of COVID-19 patients have succumbed to respiratory and systemic inflammation. Hyperstimulation of toll-like receptor (TLR) signaling is a key driver of immunopathology following infection by viruses. We found that severely ill COVID-19 patients in the Intensive Care Unit (ICU) display hallmarks of such hyper-stimulation with abundant agonists of nucleic acid-sensing TLRs present in their blood and lungs. These nucleic acid-containing Damage and Pathogen Associated Molecular Patterns (DAMPs/PAMPs) can be depleted using nucleic acid-binding microfibers to limit the patient samples' ability to hyperactivate such innate immune receptors. Single-cell RNA-sequencing revealed that CD16+ monocytes from deceased but not recovered ICU patients exhibit a TLR-tolerant phenotype and a deficient anti-viral response after ex vivo TLR stimulation. Plasma proteomics confirmed such myeloid hyperactivation and revealed DAMP/PAMP carrier consumption in deceased patients. Treatment of these COVID-19 patient samples with MnO nanoparticles effectively neutralizes TLR activation by the abundant nucleic acid-containing DAMPs/PAMPs present in their lungs and blood. Finally, MnO nanoscavenger treatment limits the ability of DAMPs/PAMPs to induce TLR tolerance in monocytes. Thus, treatment with microfiber- or nanoparticle-based DAMP/PAMP scavengers may prove useful for limiting SARS-CoV-2 induced hyperinflammation, preventing monocytic TLR tolerance, and improving outcomes in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Ibtehaj Naqvi
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | - Nicholas Giroux
- Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA; Duke University, Graduate School, USA
| | - Lyra Olson
- Duke University, Graduate School, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA
| | - Sarah Ahn Morrison
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | | | - Tolu O Akinade
- Columbia University, Department of Biomedical Engineering, USA
| | - Yuefei Zhu
- Columbia University, Department of Biomedical Engineering, USA
| | - Yiling Zhong
- Columbia University, Department of Biomedical Engineering, USA
| | - Shree Bose
- Duke University, Graduate School, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA
| | - Stephanie Arvai
- Duke University Center for Genomic and Computational Biology, RNA Sequencing Core, USA
| | - Karen Abramson
- Duke University Center for Genomic and Computational Biology, RNA Sequencing Core, USA
| | - Lingye Chen
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Loretta Que
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Bryan Kraft
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Xiling Shen
- Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA
| | - Jaewoo Lee
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | - Kam W Leong
- Columbia University, Department of Biomedical Engineering, USA
| | - Smita K Nair
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA; Duke University School of Medicine, Department of Pathology, USA; Duke University School of Medicine, Department of Neurosurgery, USA.
| | - Bruce Sullenger
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA; Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA; Duke University School of Medicine, Department of Neurosurgery, USA.
| |
Collapse
|
26
|
Early Plasma Nuclear DNA, Mitochondrial DNA, and Nucleosome Concentrations Are Associated With Acute Kidney Injury in Critically Ill Trauma Patients. Crit Care Explor 2022; 4:e0663. [PMID: 35372847 PMCID: PMC8963825 DOI: 10.1097/cce.0000000000000663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Circulating nucleic acids, alone and in complex with histones as nucleosomes, have been proposed to link systemic inflammation and coagulation after trauma to acute kidney injury (AKI). We sought to determine the association of circulating nucleic acids measured at multiple time points after trauma with AKI risk.
Collapse
|
27
|
Shi Y, Fan Y, Chen T. Editorial: Acute-On-Chronic Liver Failure: Natural History, Mechanism, and Treatment. Front Med (Lausanne) 2022; 9:867436. [PMID: 35360750 PMCID: PMC8961725 DOI: 10.3389/fmed.2022.867436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/07/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Yu Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuchen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Chen
- Department and Institute of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
HMGB1 Inhibition to Ameliorate Organ Failure and Increase Survival in Trauma. Biomolecules 2022; 12:biom12010101. [PMID: 35053249 PMCID: PMC8773879 DOI: 10.3390/biom12010101] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 12/15/2022] Open
Abstract
Several preclinical and clinical reports have demonstrated that levels of circulating high mobility group box 1 protein (HMGB1) are increased early after trauma and are associated with systemic inflammation and clinical outcomes. However, the mechanisms of the interaction between HMGB1 and inflammatory mediators that lead to the development of remote organ damage after trauma remain obscure. HMGB1 and inflammatory mediators were analyzed in plasma from 54 combat casualties, collected on admission to a military hospital in Iraq, and at 8 and 24 h after admission. In total, 45 (83%) of these patients had traumatic brain injury (TBI). Nine healthy volunteers were enrolled as controls. HMGB1 plasma levels were significantly increased in the first 8 h after admission, and were found to be associated with systemic inflammatory responses, injury severity score, and presence of TBI. These data provided the rationale for designing experiments in rats subjected to blast injury and hemorrhage, to explore the effect of HMGB1 inhibition by CX-01 (2-O, 3-O desulfated heparin). Animals were cannulated, then recovered for 5–7 days before blast injury in a shock tube and volume-controlled hemorrhage. Blast injury and hemorrhage induced an early increase in HMGB1 plasma levels along with severe tissue damage and high mortality. CX-01 inhibited systemic HMGB1 activity, decreased local and systemic inflammatory responses, significantly reduced tissue and organ damage, and tended to increase survival. These data suggest that CX-01 has potential as an adjuvant treatment for traumatic hemorrhage.
Collapse
|
29
|
Ondee T, Pongpirul K, Janchot K, Kanacharoen S, Lertmongkolaksorn T, Wongsaroj L, Somboonna N, Ngamwongsatit N, Leelahavanichkul A. Lactiplantibacillus plantarum dfa1 Outperforms Enterococcus faecium dfa1 on Anti-Obesity in High Fat-Induced Obesity Mice Possibly through the Differences in Gut Dysbiosis Attenuation, despite the Similar Anti-Inflammatory Properties. Nutrients 2021; 14:nu14010080. [PMID: 35010955 PMCID: PMC8746774 DOI: 10.3390/nu14010080] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/22/2022] Open
Abstract
Fat reduction and anti-inflammation are commonly claimed properties of probiotics. Lactiplantibacillus plantarum and Enterococcus faecium were tested in high fat-induced obesity mice and in vitro experiments. After 16 weeks of probiotics, L. plantarum dfa1 outperforms E. faecium dfa1 on the anti-obesity property as indicated by body weight, regional fat accumulation, serum cholesterol, inflammatory cytokines (in blood and colon tissue), and gut barrier defect (FITC-dextran assay). With fecal microbiome analysis, L. plantarum dfa1 but not E. faecium dfa1 reduced fecal abundance of pathogenic Proteobacteria without an alteration in total Gram-negative bacteria when compared with non-probiotics obese mice. With palmitic acid induction, the condition media from both probiotics similarly attenuated supernatant IL-8, improved enterocyte integrity and down-regulated cholesterol absorption-associated genes in Caco-2 cell (an enterocyte cell line) and reduced supernatant cytokines (TNF-α and IL-6) with normalization of cell energy status (extracellular flux analysis) in bone-marrow-derived macrophages. Due to the anti-inflammatory effect of the condition media of both probiotics on palmitic acid-activated enterocytes was neutralized by amylase, the active anti-inflammatory molecules might, partly, be exopolysaccharides. As L. plantarum dfa1 out-performed E. faecium dfa1 in anti-obesity property, possibly through the reduced fecal Proteobacteria, with a similar anti-inflammatory exopolysaccharide; L. plantarum is a potentially better option for anti-obesity than E. faecium.
Collapse
Affiliation(s)
- Thunnicha Ondee
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (T.O.); (K.J.)
| | - Krit Pongpirul
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (T.O.); (K.J.)
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Bumrungrad International Hospital, Bangkok 10110, Thailand
- Correspondence: (K.P.); (A.L.)
| | - Kantima Janchot
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (T.O.); (K.J.)
| | - Suthicha Kanacharoen
- Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Thanapat Lertmongkolaksorn
- Research Management and Development Division, Office of the President, Mahidol University, Nakhon Pathom 73170, Thailand;
| | - Lampet Wongsaroj
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (L.W.); (N.S.)
| | - Naraporn Somboonna
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; (L.W.); (N.S.)
- Microbiome Research Unit for Probiotics in Food and Cosmetics, Chulalongkorn University, Bangkok 10330, Thailand
| | - Natharin Ngamwongsatit
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom 73170, Thailand;
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (K.P.); (A.L.)
| |
Collapse
|
30
|
Li C, He Q, Qian H, Liu J. Overview of the pathogenesis of COVID-19 (Review). Exp Ther Med 2021; 22:1011. [PMID: 34345293 PMCID: PMC8311250 DOI: 10.3892/etm.2021.10444] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/21/2021] [Indexed: 01/08/2023] Open
Abstract
At present, the pathogenesis of the novel coronavirus disease 2019 (COVID-19) has not been fully elucidated. Clinical and experimental findings from studies investigating COVID-19 have suggested that the immune-inflammatory response has a crucial role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The present article aimed to systematically review the available literature on the pathogenesis of COVID-19. Severe COVID-19 is characterized by organ dysfunction, hypercytokinemia and lymphopenia. It is assumed that the direct cytopathological damage of host cells and the dysregulated immune response caused by SARS-CoV-2 may be the primary underlying mechanisms of COVID-19. Based on the published literature, this review attempts to provide an integrated view of the immunological mechanisms and the potential pathogenesis of COVID-19, providing an in-depth summary of the host-pathogen interaction and host immune responses. It is of great importance to elucidate the possible pathogenesis of COVID-19 to determine the direction of future research.
Collapse
Affiliation(s)
- Chao Li
- Department of Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Qifang He
- Department of Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| | - Hebu Qian
- Department of Critical Care Medicine, Suzhou Ninth People's Hospital, The Affiliated Wujiang Hospital of Nantong University, Suzhou, Jiangsu 215001, P.R. China
| | - Jun Liu
- Department of Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215001, P.R. China
| |
Collapse
|
31
|
Moriyama K, Nishida O. Targeting Cytokines, Pathogen-Associated Molecular Patterns, and Damage-Associated Molecular Patterns in Sepsis via Blood Purification. Int J Mol Sci 2021; 22:8882. [PMID: 34445610 PMCID: PMC8396222 DOI: 10.3390/ijms22168882] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 01/14/2023] Open
Abstract
Sepsis is characterized by a dysregulated immune response to infections that causes life-threatening organ dysfunction and even death. When infections occur, bacterial cell wall components (endotoxin or lipopolysaccharide), known as pathogen-associated molecular patterns, bind to pattern recognition receptors, such as toll-like receptors, to initiate an inflammatory response for pathogen elimination. However, strong activation of the immune system leads to cellular dysfunction and ultimately organ failure. Damage-associated molecular patterns (DAMPs), which are released by injured host cells, are well-recognized triggers that result in the elevation of inflammatory cytokine levels. A cytokine storm is thus amplified and sustained in this vicious cycle. Interestingly, during sepsis, neutrophils transition from powerful antimicrobial protectors into dangerous mediators of tissue injury and organ dysfunction. Thus, the concept of blood purification has evolved to include inflammatory cells and mediators. In this review, we summarize recent advances in knowledge regarding the role of lipopolysaccharides, cytokines, DAMPs, and neutrophils in the pathogenesis of sepsis. Additionally, we discuss the potential of blood purification, especially the adsorption technology, for removing immune cells and molecular mediators, thereby serving as a therapeutic strategy against sepsis. Finally, we describe the concept of our immune-modulating blood purification system.
Collapse
Affiliation(s)
- Kazuhiro Moriyama
- Laboratory for Immune Response and Regulatory Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan
| | - Osamu Nishida
- Department of Anesthesiology and Critical Care Medicine, Fujita Health University School of Medicine, Toyoake 470-1192, Japan;
| |
Collapse
|
32
|
Tu Q, Cotta M, Raman S, Graham N, Schlapbach L, Roberts JA. Individualized precision dosing approaches to optimize antimicrobial therapy in pediatric populations. Expert Rev Clin Pharmacol 2021; 14:1383-1399. [PMID: 34313180 DOI: 10.1080/17512433.2021.1961578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction:Severe infections continue to impose a major burden on critically ill children and mortality rates remain stagnant. Outcomes rely on accurate and timely delivery of antimicrobials achieving target concentrations in infected tissue. Yet, developmental aspects, disease-related variables, and host factors may severely alter antimicrobial pharmacokinetics in pediatrics. The emergence of antimicrobial resistance increases the need for improved treatment approaches.Areas covered:This narrative review explores why optimization of antimicrobial therapy in neonates, infants, children, and adolescents is crucial and summarizes the possible dosing approaches to achieve antimicrobial individualization. Finally, we outline a roadmap toward scientific evidence informing the development and implementation of precision antimicrobial dosing in critically ill children.The literature search was conducted on PubMed using the following keywords: neonate, infant, child, adolescent, pediatrics, antimicrobial, pharmacokinetic, pharmacodynamic target, Bayes dosing software, optimizing, individualizing, personalizing, precision dosing, drug monitoring, validation, attainment, and software implementation. Further articles were sought from the references of the above searched articles.Expert opinion:Recently, technological innovations have emerged that enabled the development of individualized antimicrobial dosing approaches in adults. More work is required in pediatrics to make individualized antimicrobial dosing approaches widely operationalized in this population.
Collapse
Affiliation(s)
- Quyen Tu
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Department of Pharmacy, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Menino Cotta
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Sainath Raman
- Department of Paediatric Intensive Care Medicine, Queensland Children's Hospital, Brisbane, QLD, Australia.,Centre for Children's Health Research (CCHR), The University of Queensland, Brisbane, QLD, Australia
| | - Nicolette Graham
- Department of Pharmacy, Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Luregn Schlapbach
- Department of Paediatric Intensive Care Medicine, Queensland Children's Hospital, Brisbane, QLD, Australia.,Department of Intensive Care and Neonatology, The University Children's Hospital Zurich, Switzerland
| | - Jason A Roberts
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia.,Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| |
Collapse
|
33
|
Zhao Y, Pu M, Zhang J, Wang Y, Yan X, Yu L, He Z. Recent advancements of nanomaterial-based therapeutic strategies toward sepsis: bacterial eradication, anti-inflammation, and immunomodulation. NANOSCALE 2021; 13:10726-10747. [PMID: 34165483 DOI: 10.1039/d1nr02706a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sepsis is a life threatening disease that is caused by a dysregulated host immune response to infection, resulting in tissue damage and organ dysfunction, which account for a high in-hospital mortality (approximately 20%). However, there are still no effective and specific therapeutics for clinical sepsis management. Nanomaterial-based strategies have emerged as promising tools for improving the therapeutic efficacy of sepsis by combating lethal bacterial infection, modulating systemic inflammatory response, preventing multiple organ failure, etc. This review has comprehensively summarized the recent advancements in nanomaterial-based strategies for the management of sepsis and severe complications, in which those nanosystems act either as inherent therapeutics or as nanocarriers for the precise delivery of agents. These formulations mechanically possess antibacterial, anti-inflammatory, immunomodulatory, and anti-oxidative effects, achieving multifunctional synergistic treatment efficacy against sepsis. Furthermore, several cell membrane-derived biomimetic nanoplatforms have been used as decoys to trap and neutralize the pathogenic toxins. The critical role of other adjuvant therapies in sepsis management, including the combination of nanotechnology and stem cell therapy, is also highlighted. Overall, this review provides insights into innovative nanotechnology-based strategies applied in sepsis treatment.
Collapse
Affiliation(s)
- Yi Zhao
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Minju Pu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Jingwen Zhang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Yanan Wang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Xuefeng Yan
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Liangmin Yu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Zhiyu He
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| |
Collapse
|
34
|
Sultan ZW, Jaeckel ER, Krause BM, Grady SM, Murphy CA, Sanders RD, Banks MI. Electrophysiological signatures of acute systemic lipopolysaccharide-induced inflammation: potential implications for delirium science. Br J Anaesth 2021; 126:996-1008. [PMID: 33648701 PMCID: PMC8132883 DOI: 10.1016/j.bja.2020.12.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Novel preventive therapies are needed for postoperative delirium, which especially affects older patients. A mouse model is presented that captures inflammation-associated cortical slow wave activity (SWA) observed in patients, allowing exploration of the mechanistic role of prostaglandin-adenosine signalling. METHODS EEG and cortical cytokine measurements (interleukin 6, monocyte chemoattractant protein-1) were obtained from adult and aged mice. Behaviour, SWA, and functional connectivity were assayed before and after systemic administration of lipopolysaccharide (LPS)+piroxicam (cyclooxygenase inhibitor) or LPS+caffeine (adenosine receptor antagonist). To avoid the confounder of inflammation-driven changes in movement which alter SWA and connectivity, electrophysiological recordings were classified as occurring during quiescence or movement, and propensity score matching was used to match distributions of movement magnitude between baseline and post-LPS administration. RESULTS LPS produces increases in cortical cytokines and behavioural quiescence. In movement-matched data, LPS produces increases in SWA (likelihood-ratio test: χ2(4)=21.51, P<0.001), but not connectivity (χ2(4)=6.39, P=0.17). Increases in SWA associate with interleukin 6 (P<0.001) and monocyte chemoattractant protein-1 (P=0.001) and are suppressed by piroxicam (P<0.001) and caffeine (P=0.046). Aged animals compared with adult animals show similar LPS-induced SWA during movement, but exaggerated cytokine response and increased SWA during quiescence. CONCLUSIONS Cytokine-SWA correlations during wakefulness are consistent with observations in patients with delirium. Absence of connectivity effects after accounting for movement changes suggests decreased connectivity in patients is a biomarker of hypoactivity. Exaggerated effects in quiescent aged animals are consistent with increased hypoactive delirium in older patients. Prostaglandin-adenosine signalling may link inflammation to neural changes and hence delirium.
Collapse
Affiliation(s)
- Ziyad W Sultan
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth R Jaeckel
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Bryan M Krause
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Sean M Grady
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Caitlin A Murphy
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert D Sanders
- Specialty of Anaesthetics, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Anaesthetics, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Matthew I Banks
- Department of Anesthesiology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
35
|
Udompornpitak K, Bhunyakarnjanarat T, Charoensappakit A, Dang CP, Saisorn W, Leelahavanichkul A. Lipopolysaccharide-Enhanced Responses against Aryl Hydrocarbon Receptor in FcgRIIb-Deficient Macrophages, a Profound Impact of an Environmental Toxin on a Lupus-Like Mouse Model. Int J Mol Sci 2021; 22:ijms22084199. [PMID: 33919603 PMCID: PMC8073880 DOI: 10.3390/ijms22084199] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/15/2021] [Accepted: 04/15/2021] [Indexed: 01/06/2023] Open
Abstract
Fc gamma receptor IIb (FcgRIIb) is the only inhibitory-FcgR in the FcgR family, and FcgRIIb-deficient (FcgRIIb−/−) mice develop a lupus-like condition with hyper-responsiveness against several stimulations. The activation of aryl hydrocarbon receptor (Ahr), a cellular environmental sensor, might aggravate activity of the lupus-like condition. As such, 1,4-chrysenequinone (1,4-CQ), an Ahr-activator, alone did not induce supernatant cytokines from macrophages, while the 24 h pre-treatment by lipopolysaccharide (LPS), a representative inflammatory activator, prior to 1,4-CQ activation (LPS/1,4-CQ) predominantly induced macrophage pro-inflammatory responses. Additionally, the responses from FcgRIIb−/− macrophages were more prominent than wild-type (WT) cells as determined by (i) supernatant cytokines (TNF-α, IL-6, and IL-10), (ii) expression of the inflammation associated genes (NF-κB, aryl hydrocarbon receptor, iNOS, IL-1β and activating-FcgRIV) and cell-surface CD-86 (a biomarker of M1 macrophage polarization), and (iii) cell apoptosis (Annexin V), with the lower inhibitory-FcgRIIb expression. Moreover, 8-week-administration of 1,4-CQ in 8 week old FcgRIIb−/− mice, a genetic-prone lupus-like model, enhanced lupus characteristics as indicated by anti-dsDNA, serum creatinine, proteinuria, endotoxemia, gut-leakage (FITC-dextran), and glomerular immunoglobulin deposition. In conclusion, an Ahr activation worsened the disease severity in FcgRIIb−/− mice possibly through the enhanced inflammatory responses. The deficiency of inhibitory-FcgRIIb in these mice, at least in part, prominently enhanced the pro-inflammatory responses. Our data suggest that patients with lupus might be more vulnerable to environmental pollutants.
Collapse
Affiliation(s)
- Kanyarat Udompornpitak
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (T.B.); (A.C.); (C.P.D.); (W.S.)
| | - Thansita Bhunyakarnjanarat
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (T.B.); (A.C.); (C.P.D.); (W.S.)
| | - Awirut Charoensappakit
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (T.B.); (A.C.); (C.P.D.); (W.S.)
| | - Cong Phi Dang
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (T.B.); (A.C.); (C.P.D.); (W.S.)
| | - Wilasinee Saisorn
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (T.B.); (A.C.); (C.P.D.); (W.S.)
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (T.B.); (A.C.); (C.P.D.); (W.S.)
- Correspondence: ; Tel.: +66-2-256-4251; Fax: +66-2-252-6920
| |
Collapse
|
36
|
Toldi J, Nemeth D, Hegyi P, Molnar Z, Solymar M, Farkas N, Alizadeh H, Rumbus Z, Pakai E, Garami A. Macrophage migration inhibitory factor as a diagnostic and predictive biomarker in sepsis: meta-analysis of clinical trials. Sci Rep 2021; 11:8051. [PMID: 33850259 PMCID: PMC8044150 DOI: 10.1038/s41598-021-87613-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 03/31/2021] [Indexed: 12/29/2022] Open
Abstract
The hunt for useful sepsis biomarkers is ongoing. Macrophage migration inhibitory factor (MIF) was implicated as a biomarker in sepsis, but its diagnostic and prognostic value has remained unclear in human studies. Here, we aimed at clarifying the value of MIF as a sepsis biomarker with the meta-analysis of clinical trials. PubMed, EMBASE, and Cochrane Central Register of Controlled Trials databases were searched until December 2019. From the included studies, blood MIF levels and indicators of disease severity were extracted in septic and control patient groups. Twenty-one eligible studies were identified, including data from 1876 subjects (of which 1206 had sepsis). In the septic patients, blood MIF levels were significantly higher than in healthy controls with a standardized mean difference (SMD) of 1.47 (95% confidence interval, CI: 0.96-1.97; p < 0.001) and also higher than in patient groups with nonseptic systemic inflammation (SMD = 0.94; CI: 0.51-1.38; p < 0.001). Markedly greater elevation in blood MIF level was found in the more severe forms of sepsis and in nonsurvivors than in less severe forms and in survivors with SMDs of 0.84 (CI: 0.45-1.24) and 0.75 (CI: 0.40-1.11), respectively (p < 0.001 for both). In conclusion, blood MIF level is more elevated in systemic inflammation caused by infection (i.e., sepsis) compared to noninfectious causes. In more severe forms of sepsis, including fatal outcome, MIF levels are higher than in less severe forms. These results suggest that MIF can be a valuable diagnostic and prognostic biomarker in sepsis given that well-designed clinical trials validate our findings.
Collapse
Affiliation(s)
- Janos Toldi
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
- Department of Anesthesiology and Intensive Care, Medical School, University of Pecs, Pecs, Hungary
| | - David Nemeth
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Peter Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Zsolt Molnar
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Department of Anesthesiology and Intensive Therapy, Poznan University of Medical Sciences, Poznan, Poland
| | - Margit Solymar
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pecs, Pecs, Hungary
| | - Hussain Alizadeh
- Division of Hematology, First Department of Medicine, University of Pecs, Pecs, Hungary
| | - Zoltan Rumbus
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Eszter Pakai
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
| |
Collapse
|
37
|
Lactobacillus acidophilus LA5 improves saturated fat-induced obesity mouse model through the enhanced intestinal Akkermansia muciniphila. Sci Rep 2021; 11:6367. [PMID: 33737543 PMCID: PMC7973717 DOI: 10.1038/s41598-021-85449-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, a major healthcare problem worldwide, induces metabolic endotoxemia through the gut translocation of lipopolysaccharides (LPS), a major cell wall component of Gram-negative bacteria, causing a chronic inflammatory state. A combination of several probiotics including Lactobacillus acidophilus 5 (LA5), a potent lactic acid-producing bacterium, has previously been shown to attenuate obesity. However, data on the correlation between a single administration of LA5 versus microbiota alteration might be helpful for the probiotic adjustment. LA5 was administered daily together with a high-fat diet (HFD) for 8 weeks in mice. Furthermore, the condition media of LA5 was also tested in a hepatocyte cell-line (HepG2 cells). Accordingly, LA5 attenuated obesity in mice as demonstrated by weight reduction, regional fat accumulation, lipidemia, liver injury (liver weight, lipid compositions, and liver enzyme), gut permeability defect, endotoxemia, and serum cytokines. Unsurprisingly, LA5 improved these parameters and acidified fecal pH leads to the attenuation of fecal dysbiosis. The fecal microbiome analysis in obese mice with or without LA5 indicated; (i) decreased Bacteroidetes (Gram-negative anaerobes that predominate in non-healthy conditions), (ii) reduced total fecal Gram-negative bacterial burdens (the sources of gut LPS), (iii) enhanced Firmicutes (Gram-positive bacteria with potential benefits) and (iv) increased Verrucomycobia, especially Akkermansia muciniphila, a bacterium with the anti-obesity property. With LA5 administration, A. muciniphila in the colon were more than 2,000 folds higher than the regular diet mice as determined by 16S rRNA. Besides, LA5 produced anti-inflammatory molecules with a similar molecular weight to LPS that reduced cytokine production in LPS-activated HepG2 cells. In conclusion, LA5 attenuated obesity through (i) gut dysbiosis attenuation, partly through the promotion of A. muciniphila (probiotics with the difficulty in preparation processes), (ii) reduced endotoxemia, and (iii) possibly decreased liver injury by producing the anti-inflammatory molecules.
Collapse
|
38
|
Zhou Y, Li M, Lv T, Huang M, Cheng B, Zhang Y, Zhu J. Gastrodin Inhibits Virus Infection by Promoting the Production of Type I Interferon. Front Pharmacol 2021; 11:608707. [PMID: 33776755 PMCID: PMC7990098 DOI: 10.3389/fphar.2020.608707] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 12/30/2020] [Indexed: 01/18/2023] Open
Abstract
Type I interferon (IFN-I) plays a critical role in the antiviral immune response. However, viruses have developed different strategies to suppress the production of IFN-I for its own escape and amplification. Therefore, promoting the production of IFN-I is an effective strategy against virus infection. Gastrodin (GTD), a phenolic glucoside extracted from Gastrodia elata Blume, has been reported to play a protective role in some central nervous system -related diseases and is beneficial for the recovery of diseases by inhibiting inflammation. However, the effect of GTD on virus infection is largely unknown. Here we found GTD treatment increased the survival rate of mice infected with vesicular stomatitis virus (VSV) or herpes simplex virus-1 (HSV-1). The production of IFN-I was increased in GTD-treated mice or macrophages compared to the control group, during virus infection. Furthermore, the activation of interferon regulatory factor 3 (IRF3) was promoted by GTD in macrophages upon VSV and HSV-1 infection. Our results demonstrated that GTD could inhibit the VSV and HSV-1 infection by promoting the production of IFN-I in macrophages and might provide an effective strategy against virus infection.
Collapse
Affiliation(s)
- Yunlian Zhou
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mengyao Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tingyi Lv
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Meixia Huang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Beilei Cheng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanyuan Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Zhu
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|
39
|
Bhunyakarnjanarat T, Udompornpitak K, Saisorn W, Chantraprapawat B, Visitchanakun P, Dang CP, Issara-Amphorn J, Leelahavanichkul A. Prominent Indomethacin-Induced Enteropathy in Fcgriib Defi-cient lupus Mice: An Impact of Macrophage Responses and Immune Deposition in Gut. Int J Mol Sci 2021; 22:1377. [PMID: 33573095 PMCID: PMC7866536 DOI: 10.3390/ijms22031377] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 02/06/2023] Open
Abstract
A high dose of NSAIDs, a common analgesic, might induce lupus activity through several NSAIDs adverse effects including gastrointestinal permeability defect (gut leakage) and endotoxemia. Indomethacin (25 mg/day) was orally administered for 7 days in 24-wk-old Fc gamma receptor IIb deficient (FcgRIIb-/-) mice, an asymptomatic lupus model (increased anti-dsDNA without lupus nephritis), and age-matched wild-type (WT) mice. Severity of indomethacin-induced enteropathy in FcgRIIb-/- mice was higher than WT mice as demonstrated by survival analysis, intestinal injury (histology, immune-deposition, and intestinal cytokines), gut leakage (FITC-dextran assay and endotoxemia), serum cytokines, and lupus characteristics (anti-dsDNA, renal injury, and proteinuria). Prominent responses of FcgRIIb-/- macrophages toward lipopolysaccharide (LPS) compared to WT cells due to the expression of only activating-FcgRs without inhibitory-FcgRIIb were demonstrated. Extracellular flux analysis indicated the greater mitochondria activity (increased respiratory capacity and respiratory reserve) in FcgRIIb-/- macrophages with a concordant decrease in glycolysis activity when compared to WT cells. In conclusion, gut leakage-induced endotoxemia is more severe in indomethacin-administered FcgRIIb-/- mice than WT, possibly due to the enhanced indomethacin toxicity from lupus-induced intestinal immune-deposition. Due to a lack of inhibitory-FcgRIIb expression, mitochondrial function, and cytokine production of FcgRIIb-/- macrophages were more prominent than WT cells. Hence, lupus disease-activation from NSAIDs-enteropathy-induced gut leakage is possible.
Collapse
Affiliation(s)
- Thansita Bhunyakarnjanarat
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok 10330, Thailand;
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Kanyarat Udompornpitak
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Wilasinee Saisorn
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Bhumdhanin Chantraprapawat
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Peerapat Visitchanakun
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Cong Phi Dang
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Jiraphorn Issara-Amphorn
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok 10330, Thailand; (K.U.); (W.S.); (B.C.); (P.V.); (C.P.D.); (J.I.-A.)
- Department of Microbiology, Immunology Unit, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
40
|
Stavropoulou E, Kantartzi K, Tsigalou C, Konstantinidis T, Romanidou G, Voidarou C, Bezirtzoglou E. Focus on the Gut-Kidney Axis in Health and Disease. Front Med (Lausanne) 2021; 7:620102. [PMID: 33553216 PMCID: PMC7859267 DOI: 10.3389/fmed.2020.620102] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
The recent new developments in technology with culture-independent techniques including genome sequencing methodologies shed light on the identification of microbiota bacterial species and their role in health and disease. Microbiome is actually reported as an important predictive tool for evaluating characteristic shifts in case of disease. Our present review states the development of different renal diseases and pathologies linked to the intestinal dysbiosis, which impacts on host homeostasis. The gastrointestinal-kidney dialogue provides intriguing features in the pathogenesis of several renal diseases. Without any doubt, investigation of this interconnection consists one of the most cutting-edge areas of research with potential implications on our health.
Collapse
Affiliation(s)
- Elisavet Stavropoulou
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Department of Infectious Diseases, Central Institute, Valais Hospital, Sion, Switzerland
| | - Konstantia Kantartzi
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christina Tsigalou
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | | | | | - Eugenia Bezirtzoglou
- Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
41
|
Stanfield BA, Purves T, Palmer S, Sullenger B, Welty-Wolf K, Haines K, Agarwal S, Kasotakis G. IL-10 and class 1 histone deacetylases act synergistically and independently on the secretion of proinflammatory mediators in alveolar macrophages. PLoS One 2021; 16:e0245169. [PMID: 33471802 PMCID: PMC7816993 DOI: 10.1371/journal.pone.0245169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022] Open
Abstract
Introduction Anti-inflammatory cytokine IL-10 suppresses pro-inflammatory IL-12b expression after Lipopolysaccharide (LPS) stimulation in colonic macrophages, as part of the innate immunity Toll-Like Receptor (TLR)-NF-κB activation system. This homeostatic mechanism limits excess inflammation in the intestinal mucosa, as it constantly interacts with the gut flora. This effect is reversed with Histone Deacetylase 3 (HDAC3), a class I HDAC, siRNA, suggesting it is mediated through HDAC3. Given alveolar macrophages’ prominent role in Acute Lung Injury (ALI), we aim to determine whether a similar regulatory mechanism exists in the typically sterile pulmonary microenvironment. Methods Levels of mRNA and protein for IL-10, and IL-12b were determined by qPCR and ELISA/Western Blot respectively in naïve and LPS-stimulated alveolar macrophages. Expression of the NF-κB intermediaries was also similarly assessed. Experiments were repeated with AS101 (an IL-10 protein synthesis inhibitor), MS-275 (a selective class 1 HDAC inhibitor), or both. Results LPS stimulation upregulated all proinflammatory mediators assayed in this study. In the presence of LPS, inhibition of IL-10 and/or class 1 HDACs resulted in both synergistic and independent effects on these signaling molecules. Quantitative reverse-transcriptase PCR on key components of the TLR4 signaling cascade demonstrated significant diversity in IL-10 and related gene expression in the presence of LPS. Inhibition of IL-10 secretion and/or class 1 HDACs in the presence of LPS independently affected the transcription of MyD88, IRAK1, Rela and the NF-κB p50 subunit. Interestingly, by quantitative ELISA inhibition of IL-10 secretion and/or class 1 HDACs in the presence of LPS independently affected the secretion of not only IL-10, IL-12b, and TNFα, but also proinflammatory mediators CXCL2, IL-6, and MIF. These results suggest that IL-10 and class 1 HDAC activity regulate both independent and synergistic mechanisms of proinflammatory cytokine/chemokine signaling. Conclusions Alveolar macrophages after inflammatory stimulation upregulate both IL-10 and IL-12b production, in a highly class 1 HDAC-dependent manner. Class 1 HDACs appear to help maintain the balance between the pro- and anti-inflammatory IL-12b and IL-10 respectively. Class 1 HDACs may be considered as targets for the macrophage-initiated pulmonary inflammation in ALI in a preclinical setting.
Collapse
Affiliation(s)
- Brent A. Stanfield
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Trauma, Acute and Critical Care Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Todd Purves
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Urology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Scott Palmer
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Bruce Sullenger
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Karen Welty-Wolf
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Krista Haines
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Trauma, Acute and Critical Care Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Suresh Agarwal
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Trauma, Acute and Critical Care Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - George Kasotakis
- Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- Division of Trauma, Acute and Critical Care Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
42
|
Yan Z, Luo H, Xie B, Tian T, Li S, Chen Z, Liu J, Zhao X, Zhang L, Deng Y, Billiar TR, Jiang Y. Targeting adaptor protein SLP76 of RAGE as a therapeutic approach for lethal sepsis. Nat Commun 2021; 12:308. [PMID: 33436632 PMCID: PMC7804203 DOI: 10.1038/s41467-020-20577-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 12/07/2020] [Indexed: 01/29/2023] Open
Abstract
Accumulating evidence shows that RAGE has an important function in the pathogenesis of sepsis. However, the mechanisms by which RAGE transduces signals to downstream kinase cascades during septic shock are not clear. Here, we identify SLP76 as a binding partner for the cytosolic tail of RAGE both in vitro and in vivo and demonstrate that SLP76 binds RAGE through its sterile α motif (SAM) to mediate downstream signaling. Genetic deficiency of RAGE or SLP76 reduces AGE-induced phosphorylation of p38 MAPK, ERK1/2 and IKKα/β, as well as cytokine release. Delivery of the SAM domain into macrophages via the TAT cell-penetrating peptide blocks proinflammatory cytokine production. Furthermore, administration of TAT-SAM attenuates inflammatory cytokine release and tissue damage in mice subjected to cecal ligation and puncture (CLP) and protects these mice from the lethality of sepsis. These findings reveal an important function for SLP76 in RAGE-mediated pro-inflammatory signaling and shed light on the development of SLP76-targeted therapeutics for sepsis.
Collapse
Affiliation(s)
- Zhengzheng Yan
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Bingyao Xie
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian Tian
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shan Li
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhixia Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xuwen Zhao
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liyong Zhang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yongqiang Deng
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
43
|
The Central Role and Possible Mechanisms of Bacterial DNAs in Sepsis Development. Mediators Inflamm 2020; 2020:7418342. [PMID: 32934605 PMCID: PMC7479481 DOI: 10.1155/2020/7418342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/20/2020] [Indexed: 12/20/2022] Open
Abstract
The pathological roles of bacterial DNA have been documented many decades ago. Bacterial DNAs are different from mammalian DNAs; the latter are heavily methylated. Mammalian cells have sensors such as TLR-9 to sense the DNAs with nonmethylated CpGs and distinguish them from host DNAs with methylated CpGs. Further investigation has identified many other types of DNA sensors distributed in a variety of cellular compartments. These sensors not only sense foreign DNAs, including bacterial and viral DNAs, but also sense damaged DNAs from the host cells. The major downstream signalling pathways includeTLR-9-MyD88-IKKa-IRF-7/NF-κB pathways to increase IFN/proinflammatory cytokine production, STING-TBK1-IRF3 pathway to increase IFN-beta, and AIM2-ASC-caspas-1 pathway to release IL-1beta. The major outcome is to activate host immune response by inducing cytokine production. In this review, we focus on the roles and potential mechanisms of DNA sensors and downstream pathways in sepsis. Although bacterial DNAs play important roles in sepsis development, bacterial DNAs alone are unable to cause severe disease nor lead to death. Priming animals with bacterial DNAs facilitate other pathological factors, such as LPS and other virulent factors, to induce severe disease and lethality. We also discuss compartmental distribution of DNA sensors and pathological significance as well as the transport of extracellular DNAs into cells. Understanding the roles of DNA sensors and signal pathways will pave the way for novel therapeutic strategies in many diseases, particularly in sepsis.
Collapse
|
44
|
Koutsogiannaki S, Bu W, Hou L, Shibamura-Fujiogi M, Ishida H, Ohto U, Eckenhoff RG, Yuki K. The effect of anesthetics on toll like receptor 9. FASEB J 2020; 34:14645-14654. [PMID: 32901993 DOI: 10.1096/fj.202000791rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/17/2020] [Accepted: 08/20/2020] [Indexed: 12/11/2022]
Abstract
Toll like receptors (TLRs) are critical receptors to respond to danger signals, and their functions are relevant in the perioperative period. We previously reported that volatile anesthetics directly bound to TLR2 and TLR4 and attenuated their functions. Given that TLR9 can respond to mitochondrial DNA, a danger signal that is released upon tissue injury, we examined the role of anesthetics on TLR9 function. Our reporter assay showed that volatile anesthetics isoflurane and sevoflurane increased the activation of TLR9, while propofol attenuated it. TLR9 activation occurs via its dimerization. The dimerization is facilitated by unmethylated cytosine-phosphate-guanine (CpG) DNA as well as DNA containing cytosine at the second position from 5'-end (5'-xCx DNA). Our structural analysis using photoactivable anesthetics and rigid docking simulation showed that isoflurane and sevoflurane bound to both TLR9 dimer interface and 5'-xCx DNA binding site. Propofol bound to the TLR9 antagonist binding site. This is the first illustration that anesthetics can affect the binding of nucleic acids to their receptor. This study sets the foundation for the effect of anesthetics on TLR9 and will pave the way for future studies to determine the significance of such interactions in the clinical setting.
Collapse
Affiliation(s)
- Sophia Koutsogiannaki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, MA, USA.,Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Weiming Bu
- Department of Anesthesia and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - Lifei Hou
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, MA, USA.,Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Miho Shibamura-Fujiogi
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, MA, USA.,Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Hanako Ishida
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Umeharu Ohto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Roderic G Eckenhoff
- Department of Anesthesia and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children's Hospital, Boston, MA, USA.,Department of Anaesthesia, Harvard Medical School, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
45
|
Land WG. Role of Damage-Associated Molecular Patterns in Light of Modern Environmental Research: A Tautological Approach. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH 2020; 14:583-604. [PMID: 32837525 PMCID: PMC7415330 DOI: 10.1007/s41742-020-00276-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/30/2020] [Accepted: 08/01/2020] [Indexed: 05/06/2023]
Abstract
Two prominent models emerged as a result of intense interdisciplinary discussions on the environmental health paradigm, called the "exposome" concept and the "adverse outcome pathway" (AOP) concept that links a molecular initiating event to the adverse outcome via key events. Here, evidence is discussed, suggesting that environmental stress/injury-induced damage-associated molecular patterns (DAMPs) may operate as an essential integrating element of both environmental health research paradigms. DAMP-promoted controlled/uncontrolled innate/adaptive immune responses reflect the key events of the AOP concept. The whole process starting from exposure to a distinct environmental stress/injury-associated with the presence/emission of DAMPs-up to the manifestation of a disease may be regarded as an exposome. Clinical examples of such a scenario are briefly sketched, in particular, a model in relation to the emerging COVID-19 pandemic, where the interaction of noninfectious environmental factors (e.g., particulate matter) and infectious factors (SARS CoV-2) may promote SARS case fatality via superimposition of both exogenous and endogenous DAMPs.
Collapse
Affiliation(s)
- Walter Gottlieb Land
- German Academy for Transplantation Medicine, Munich, Germany
- Molecular ImmunoRheumatology, Laboratory of Excellence Transplantex, Faculty of Medicine, INSERM UMR_S1109, University of Strasbourg, Strasbourg, France
| |
Collapse
|
46
|
Zhou Y, Li M, Lv T, Huang M, Cheng B, Zhang Y, Zhu J. Gastrodin Inhibits Virus Infection by Promoting the Production of Type I Interferon. Front Pharmacol 2020. [PMID: 33776755 DOI: 10.3389/fphar.2020.00931/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Type I interferon (IFN-I) plays a critical role in the antiviral immune response. However, viruses have developed different strategies to suppress the production of IFN-I for its own escape and amplification. Therefore, promoting the production of IFN-I is an effective strategy against virus infection. Gastrodin (GTD), a phenolic glucoside extracted from Gastrodia elata Blume, has been reported to play a protective role in some central nervous system -related diseases and is beneficial for the recovery of diseases by inhibiting inflammation. However, the effect of GTD on virus infection is largely unknown. Here we found GTD treatment increased the survival rate of mice infected with vesicular stomatitis virus (VSV) or herpes simplex virus-1 (HSV-1). The production of IFN-I was increased in GTD-treated mice or macrophages compared to the control group, during virus infection. Furthermore, the activation of interferon regulatory factor 3 (IRF3) was promoted by GTD in macrophages upon VSV and HSV-1 infection. Our results demonstrated that GTD could inhibit the VSV and HSV-1 infection by promoting the production of IFN-I in macrophages and might provide an effective strategy against virus infection.
Collapse
Affiliation(s)
- Yunlian Zhou
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mengyao Li
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tingyi Lv
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Meixia Huang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Beilei Cheng
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yuanyuan Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Zhu
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, China
| |
Collapse
|