1
|
Tang Z, Zhu Y, Hu X, Lui K, Li S, Song X, Cai C, Guan X. Improving Intestinal Barrier Function in Sepsis by Partially Hydrolysed Guar Gum via the Suppression of the NF-κB/MLCK Pathway. Mol Biotechnol 2025; 67:2035-2045. [PMID: 38789715 DOI: 10.1007/s12033-024-01180-z] [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: 08/02/2023] [Accepted: 04/12/2024] [Indexed: 05/26/2024]
Abstract
Partially hydrolyzed guar gum (PHGG) protects against intestinal barrier dysfunction and can ameliorate some intestinal diseases. However, whether PHGG has a role in protecting intestinal barrier function (IBF) during sepsis remains unclear. This study aimed to investigate the role and probable mechanism of PHGG in the intestinal mucosa in sepsis. A rat sepsis model was constructed using cecal ligation and puncture (CLP). FITC-dextran 4 (FD-4) flux, serum inflammatory mediator levels, tight junction (TJ) levels, jejunum mucosa pathology, and epithelial intercellular junction ultrastructure were monitored to evaluate the effect of PHGG on IBF. Caco-2 monolayers were used to study the impact and mechanism of PHGG on lipopolysaccharide (LPS)-induced barrier dysfunction in vitro. The expression of zonula occludens protein-1 and occludin and the location of P65 were studied by immunofluorescence. Nuclear factor kappa B (NF-κB) and myosin light chain kinase 3 (MLCK) pathway-related protein expression was verified by quantitative reverse transcriptase polymerase chain reaction or western blotting. The results indicated that the jejunal mucosa structure was destroyed, the villi were disrupted and shortened, and neutrophil infiltration was evident in the septic rats. Compared to Sham group, spetic rats had increased Chiu's score, serum inflammatory mediator levels, and FD-4 flux but decreased TJ and gap junction density. In addition, the expression of MLCK, p-MLC, and TJ proteins and the expression of P65 in the nucleus were increased in septic rats. Furthermore, compared to those in the Control group, LPS-treated Caco-2 cells showed lower cell viability and transepithelial electrical resistance, while had higher FD-4 flux and the expression of MLCK, p-MLC, TJ proteins and P65 in the nucleus. PHGG pretreatment reversed the above effects induced by CLP or LPS treatment. Moreover, SN50, an NF-κB inhibitor, attenuated the above effects of LPS on Caco-2 cells. Overall, PHGG reduced inflammation, increased TJ protein expression and localization, and relieved damage to the TJ structure and intestinal permeability through suppression of the NF-κB/MLCK pathway. This study provides new insights into the role of PHGG in sepsis therapy.
Collapse
Affiliation(s)
- Zhaoxia Tang
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yanping Zhu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China
| | - Xiaoguang Hu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China
| | - Kayin Lui
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China
| | - Shuhe Li
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China
| | - Xiaodong Song
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China
| | - Changjie Cai
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China.
| | - Xiangdong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Wang J, Wang Y, Huang C, Chen Y, Li X, Jiang Z. Decursin protects against DSS-induced experimental colitis in mice by inhibiting the cGAS-STING signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5119-5127. [PMID: 39520553 DOI: 10.1007/s00210-024-03589-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
While studies have shown that Angelica gigas Nakai (A. gigas) can alleviate ulcerative colitis in mice, the therapeutic role of its main active ingredient, decursin, is uncertain. Therefore, we aimed to investigate the protective effect and mechanism of decursin against inflammatory bowel disease (IBD) in vivo using mice. IBD was simulated via induction with 3% dextran sodium sulfate (DSS), with or without daily treatment with decursin (10 mg/kg or 20 mg/kg) or 5-amino salicylic acid (5-ASA; 100 mg/kg) for 14 days. Mice were weighed and monitored daily for disease activity index (DAI) scoring. Colon tissues were collected for histopathological staining analysis, and serum was collected for ELISA measurement of proinflammatory cytokines. Western blotting was employed to analyze colonic expression levels of the tight junction-related proteins ZO-1, Occludin, and Claudin 1, as well as cGAS-STING signaling pathway-associated proteins. The expression levels of major proteins were verified using immunohistochemistry and immunofluorescence. Compared with the control group, DSS-induced mice showed decreased body weight, increased DAI scores, shortening of the colon, disrupted colon tissue structure, increased serum levels of proinflammatory cytokines, increased expression of factors involved in activating the cGAS-STING signaling pathway, and reduced expression of ZO-1, Occludin, and Claudin 1. Under decursin treatment, the pathological state of IBD was less severe, proinflammatory factors were downregulated, and activation of the cGAS-STING signaling pathway was inhibited. Our findings indicate that decursin helps restore the intestinal mucosal barrier and prevents activation of the cGAS-STING signaling cascade, alleviating experimental IBD in mice.
Collapse
Affiliation(s)
- Jiamin Wang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Yudi Wang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Caisheng Huang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Yonghu Chen
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
| | - Xuezheng Li
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, China
| | - Zhe Jiang
- College of Pharmacy, Yanbian University Hospital, Yanbian University, Yanji, Jilin, 133000, China.
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin, 133002, China.
| |
Collapse
|
3
|
Li ZY, Luo HY, Xu F, Xu Y, Ma CH, Zhang SL, Xu S, Ma YY, Li N, Miao CY. Metrnl protects intestinal barrier function by regulating tight junctions via the IKKβ/IκBα/NFκB/MLCK/MLC signaling pathway. Cell Death Discov 2025; 11:155. [PMID: 40199887 PMCID: PMC11979045 DOI: 10.1038/s41420-025-02457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/10/2025] Open
Abstract
Meteorin-like (Metrnl), also known as Subfatin, IL-41, or Cometin, is a secreted protein predominantly expressed in the intestinal epithelium. The intestinal barrier, primarily consisting of epithelial cells connected by tight junctions, is essential for maintaining gut homeostasis by preventing harmful substances from entering the body. Despite Metrnl's high expression in the intestine, its role in barrier function remains unclear. In this study, we investigated Metrnl's role in intestinal barrier function using both loss-of-function (using global and intestinal epithelium-specific knockout mice) and gain-of-function (using intestinal epithelium-specific overexpression mice) approaches. Our findings showed that Metrnl deficiency disrupted tight junctions between enterocytes and exacerbated endotoxin-induced barrier dysfunction. Mechanistically, Metrnl deficiency triggered activation of the IKKβ/IκBα/NFκB signaling pathway, leading to increased MLCK expression and MLC phosphorylation. The NFκB inhibitor PDTC reversed this effect both in vivo and in vitro. Macrophages played an essential role in Metrnl's intestinal barrier protective effects during endotoxemia, but were not necessary in burn-induced barrier injury, suggesting potential differences in mechanism between these conditions. Notably, recombinant Metrnl protein administration protected against barrier dysfunction, and genetic overexpression of Metrnl in enterocytes preserved barrier function and alleviated DSS-induced colitis. These findings establish Metrnl as a key regulator of intestinal barrier integrity through the IKKβ/IκBα/NFκB/MLCK/MLC pathway, highlighting its potential therapeutic value in treating barrier dysfunction disorders. Intestinal barrier dysfunction triggers, such as endotoxin and severe burns, may induce the release of Metrnl from vascular endothelium. This leads to an increase in circulating Metrnl. Both circulating Metrnl and local Metrnl inhibit inflammation and the IKKβ/IκBα/NFκB/MLCK/MLC signaling pathway in enterocytes, thereby protecting tight junctions from disruption caused by endotoxin or burns.
Collapse
Affiliation(s)
- Zhi-Yong Li
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
- Department of Pathology, Faculty of Medical Imaging, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| | - Heng-Yu Luo
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Fei Xu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Yao Xu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Chun-Hui Ma
- Department of Pathology, Faculty of Medical Imaging, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- Department of Immunology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Yuan-Yuan Ma
- Senior Department of Hematology, The Fifth Medical Center of People's Liberation Army(PLA), General Hospital, Beijing, 100010, China
| | - Nan Li
- Department of Immunology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
4
|
Lee S, Wischmeyer PE, Mintz CD, Serbanescu MA. Recent Insights into the Evolving Role of the Gut Microbiome in Critical Care. Crit Care Clin 2025; 41:379-396. [PMID: 40021286 DOI: 10.1016/j.ccc.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
This review explores the evolving understanding of gut microbiota's role in critical illness, focusing on how acute illness and exposures in intensive care unit (ICU) environment negatively impact the gut microbiota and the implications of these changes on host responses in critically-ill patients. Focusing on recent findings from clinical and preclinical studies, we discuss the effects of inflammation, enteral nutrient deprivation, and antibiotics on gut microbial dynamics. This review aims to enhance comprehension of microbial dynamics in the ICU and their implications for clinical outcomes and therapeutic strategies.
Collapse
Affiliation(s)
- Seoho Lee
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Paul E Wischmeyer
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA
| | - Cyrus D Mintz
- Department of Anesthesiology and Critical Care, Johns Hopkins University School of Medicine, Phipps 455 1800 Orleans Street, Baltimore, MD 21212, USA
| | - Mara A Serbanescu
- Department of Anesthesiology, Duke University School of Medicine, 5692 HAFS Box 3094, 2301 Erwin Road, Durham, NC 27710, USA.
| |
Collapse
|
5
|
Liu X, Li C, Hong C, Chen Y, Nan C, Liang S, Chen H. Expression profiling of circular RNAs in sepsis-induced acute gastrointestinal injury: insights into potential biomarkers and mechanisms. Cytotechnology 2025; 77:43. [PMID: 39867824 PMCID: PMC11759752 DOI: 10.1007/s10616-025-00704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/07/2025] [Indexed: 01/28/2025] Open
Abstract
This study aimed to investigate the role of circular RNAs (circRNAs) in sepsis-induced acute gastrointestinal injury (AGI), focusing on their potential as biomarkers and their involvement in disease progression. Peripheral blood samples from 14 patients with sepsis-induced AGI and healthy volunteers were collected. RNA sequencing was performed to profile circRNA and miRNA expression. Differential expression analysis identified key regulatory RNAs. Functional enrichment analysis was conducted to explore biological pathways, and circRNA-miRNA interaction networks were constructed. Significant differences in circRNA and miRNA expression profiles were observed between sepsis-induced AGI patients and healthy controls. Several circRNAs, including hsa_circ_0008381 and hsa_circ_0071375, exhibited stepwise expression increases correlating with AGI severity. Functional enrichment analysis indicated that the host genes of differentially expressed circRNAs are involved in key biological processes like protein ubiquitination, organelle maintenance, and cellular signaling pathways such as mitochondrial biogenesis and lipid metabolism. CircRNA-miRNA interaction networks suggested their role as miRNA sponges, regulating key downstream processes. This study demonstrated the potential of circRNAs as diagnostic biomarkers and therapeutic targets for sepsis-induced AGI. Further research is warranted to validate their clinical utility and unravel their mechanistic roles in AGI progression.
Collapse
Affiliation(s)
- Xiaojun Liu
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| | - Chenxi Li
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| | - Chengying Hong
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| | - Yuting Chen
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| | - Chuanchuan Nan
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| | - Silin Liang
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| | - Huaisheng Chen
- Department of Critical Care Medicine, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, North Dongmen Road, Luohu District, Shenzhen, 518020 Guangdong China
| |
Collapse
|
6
|
Yumoto T, Oami T, Liang Z, Burd EM, Ford ML, Turner JR, Coopersmith CM. INTESTINAL EPITHELIAL-SPECIFIC OCCLUDIN DELETION WORSENS GUT PERMEABILITY AND SURVIVAL FOLLOWING SEPSIS. Shock 2025; 63:597-605. [PMID: 39637366 DOI: 10.1097/shk.0000000000002531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
ABSTRACT Sepsis induces intestinal hyperpermeability, which is associated with higher mortality. Occludin is a tight junction protein that plays a critical role in regulating disease-associated intestinal barrier loss. This study examined the role of intestinal occludin on gut barrier function and survival in a preclinical model of sepsis. Intestinal epithelial-specific occludin knockout (occludin KO IEC ) mice and wild type controls were subjected to intra-abdominal sepsis and sacrificed at predetermined endpoints for mechanistic studies or followed for survival. Occludin KO IEC mice had a significant increase in intestinal permeability, which was induced only in the setting of sepsis as knockout mice and control mice had similar baseline permeability. The worsened barrier was specific to the leak pathway of permeability, without changes in either the pore or unrestricted pathways. Increased sepsis-induced permeability was associated with increased levels of the tight junction ZO-1 in occludin KO IEC mice. Occludin KO IEC mice also had significant increases in systemic cytokines IL-6 and MCP-1 and increased bacteremia. Furthermore, occludin KO IEC mice had higher levels of jejunal IL-1β and MCP-1 as well as increased MCP-1 and IL-17A in the peritoneal fluid although peritoneal bacteria levels were unchanged. Notably, 7-day mortality was significantly higher in occludin KO IEC mice following sepsis. Occludin thus plays a critical role in preserving gut barrier function and mediating survival during sepsis, associated with alterations in inflammation and bacteremia. Agents that preserve occludin function may represent a new therapeutic strategy in the treatment of sepsis.
Collapse
Affiliation(s)
| | | | - Zhe Liang
- Department of Surgery and Emory Critical Care Center, Emory University, School of Medicine, Atlanta, Georgia
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory University, School of Medicine, Atlanta, Georgia
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia
| | - Jerrold R Turner
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, School of Medicine, Atlanta, Georgia
| |
Collapse
|
7
|
Oami T, Shimazui T, Yumoto T, Otani S, Hayashi Y, Coopersmith CM. Gut integrity in intensive care: alterations in host permeability and the microbiome as potential therapeutic targets. J Intensive Care 2025; 13:16. [PMID: 40098052 PMCID: PMC11916345 DOI: 10.1186/s40560-025-00786-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND The gut has long been hypothesized to be the "motor" of critical illness, propagating inflammation and playing a key role in multiple organ dysfunction. However, the exact mechanisms through which impaired gut integrity potentially contribute to worsened clinical outcome remain to be elucidated. Critical elements of gut dysregulation including intestinal hyperpermeability and a perturbed microbiome are now recognized as potential therapeutic targets in critical care. MAIN BODY The gut is a finely tuned ecosystem comprising ~ 40 trillion microorganisms, a single cell layer intestinal epithelia that separates the host from the microbiome and its products, and the mucosal immune system that actively communicates in a bidirectional manner. Under basal conditions, these elements cooperate to maintain a finely balanced homeostasis benefitting both the host and its internal microbial community. Tight junctions between adjacent epithelial cells selectively transport essential molecules while preventing translocation of pathogens. However, critical illness disrupts gut barrier function leading to increased gut permeability, epithelial apoptosis, and immune activation. This disruption is further exacerbated by a shift in the microbiome toward a "pathobiome" dominated by pathogenic microbes with increased expression of virulence factors, which intensifies systemic inflammation and accelerates organ dysfunction. Research has highlighted several potential therapeutic targets to restore gut integrity in the host, including the regulation of epithelial cell function, modulation of tight junction proteins, and inhibition of epithelial apoptosis. Additionally, microbiome-targeted therapies, such as prebiotics, probiotics, fecal microbiota transplantation, and selective decontamination of the digestive tract have also been extensively investigated to promote restoration of gut homeostasis in critically ill patients. Future research is needed to validate the potential efficacy of these interventions in clinical settings and to determine if the gut can be targeted in an individualized fashion. CONCLUSION Increased gut permeability and a disrupted microbiome are common in critical illness, potentially driving dysregulated systemic inflammation and organ dysfunction. Therapeutic strategies to modulate gut permeability and restore the composition of microbiome hold promise as novel treatments for critically ill patients.
Collapse
Affiliation(s)
- Takehiko Oami
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Takashi Shimazui
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tetsuya Yumoto
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shunsuke Otani
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yosuke Hayashi
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, 101 Woodruff Circle, Suite WMB 5105, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Fan Y, Meng S, Song Y, Zhang Y, Song Y, Chen Z, Xie K. Interaction, diagnosis, and treatment of lung microbiota-NLRP3 inflammasome-target organ axis in sepsis. Int Immunopharmacol 2025; 149:114222. [PMID: 39923579 DOI: 10.1016/j.intimp.2025.114222] [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: 11/01/2024] [Revised: 01/14/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Sepsis is defined as a life-threatening condition caused by a dysregulated host response to infection, leading to multi-organ dysfunction, and representing a significant global health burden. The progression of sepsis is closely linked to disruptions in lung microbiota, including bacterial translocation, impaired barrier function, and local microenvironmental disturbances. Conversely, the worsening of sepsis exacerbates lung microbiota imbalances, contributing to multi-organ dysfunction. Recent culture-independent microbiological techniques have unveiled the complexity of the respiratory tract microbiome, necessitating a reassessment of the interactions between the host, microbes, and pathogenesis in sepsis. This review synthesizes current insights into the causes of microbiota dysbiosis and the regulatory mechanisms of the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, as well as their interactions during sepsis and sepsis-induced organ dysfunction. In addition, we summarize novel diagnostic and therapeutic approaches from the current study that may offer promising prospects for the management of sepsis.
Collapse
Affiliation(s)
- Yan Fan
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Shuqi Meng
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Yu Song
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Ying Zhang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Yan Song
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin China
| | - Zhe Chen
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin China.
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China; Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, China.
| |
Collapse
|
9
|
Soranno DE, Coopersmith CM, Brinkworth JF, Factora FNF, Muntean JH, Mythen MG, Raphael J, Shaw AD, Vachharajani V, Messer JS. A review of gut failure as a cause and consequence of critical illness. Crit Care 2025; 29:91. [PMID: 40011975 DOI: 10.1186/s13054-025-05309-7] [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: 10/03/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
In critical illness, all elements of gut function are perturbed. Dysbiosis develops as the gut microbial community loses taxonomic diversity and new virulence factors appear. Intestinal permeability increases, allowing for translocation of bacteria and/or bacterial products. Epithelial function is altered at a cellular level and homeostasis of the epithelial monolayer is compromised by increased intestinal epithelial cell death and decreased proliferation. Gut immunity is impaired with simultaneous activation of maladaptive pro- and anti-inflammatory signals leading to both tissue damage and susceptibility to infections. Additionally, splanchnic vasoconstriction leads to decreased blood flow with local ischemic changes. Together, these interrelated elements of gastrointestinal dysfunction drive and then perpetuate multi-organ dysfunction syndrome. Despite the clear importance of maintaining gut homeostasis, there are very few reliable measures of gut function in critical illness. Further, while multiple therapeutic strategies have been proposed, most have not been shown to conclusively demonstrate benefit, and care is still largely supportive. The key role of the gut in critical illness was the subject of the tenth Perioperative Quality Initiative meeting, a conference to summarize the current state of the literature and identify key knowledge gaps for future study. This review is the product of that conference.
Collapse
Affiliation(s)
- Danielle E Soranno
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University, Atlanta, GA, USA
| | - Jessica F Brinkworth
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Faith N F Factora
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Julia H Muntean
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Monty G Mythen
- Perioperative Medicine, University College London, London, England
| | - Jacob Raphael
- Anesthesiology and Perioperative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew D Shaw
- Intensive Care and Resuscitation, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Vidula Vachharajani
- Department of Pulmonary and Critical Care, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jeannette S Messer
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
10
|
Xiong D, Geng H, Lv X, Wang S, Jia L. Inflammatory Response and Anti-Inflammatory Treatment in Persistent Inflammation-Immunosuppression-Catabolism Syndrome (PICS). J Inflamm Res 2025; 18:2267-2281. [PMID: 39968098 PMCID: PMC11834740 DOI: 10.2147/jir.s504694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Many patients now survive their initial critical events but subsequently develop chronic critical illness (CCI). CCI is characterized by prolonged hospital stays, poor outcomes, and significant long-term mortality. The incidence of chronic critical illness (CCI) is estimated to be 34.4 cases per 100,000 population. The incidence varies significantly with age, peaking at 82.1 cases per 100,000 in individuals aged 75-79. The one-year mortality rate among CCI patients approaches 50%. A subset of these patients enters a state of persistent inflammation, immune suppression, and ongoing catabolism, a condition termed persistent inflammation, immunosuppression, and catabolism syndrome (PICS) in 2012. In recent years, some progress has been made in treating PICS. For instance, recent advancements such as the persistent expansion of MDSCs (myeloid-derived suppressor cells) and the mechanisms underlying intestinal barrier dysfunction have provided new directions for therapeutic strategies, as discussed below. Persistent inflammation, a key feature of PICS, has received comparatively little research attention. In this review, we examine the potential pathophysiological changes and molecular mechanisms underlying persistent inflammation and its role in PICS. We also discuss current therapies about inflammation and offer recommendations for managing patients with PICS.
Collapse
Affiliation(s)
- Dacheng Xiong
- Department of Intensive Care Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Intensive Care Medicine, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Huixian Geng
- Department of Intensive Care Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Intensive Care Medicine, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Xuechun Lv
- Department of Intensive Care Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Intensive Care Medicine, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Shuqi Wang
- Department of Intensive Care Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Intensive Care Medicine, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| | - Lijing Jia
- Department of Intensive Care Medicine, Hebei Medical University, Shijiazhuang, People’s Republic of China
- Department of Intensive Care Medicine, Hebei General Hospital, Shijiazhuang, People’s Republic of China
| |
Collapse
|
11
|
Calik A, Niraula A, Dong B, Blue CEC, Fenster DA, Dalloul RA. Iohexol-based assessment of intestinal permeability in broilers challenged with Eimeria maxima, Clostridium perfringens or both. Front Physiol 2024; 15:1520346. [PMID: 39759108 PMCID: PMC11695284 DOI: 10.3389/fphys.2024.1520346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Impaired intestinal integrity in broilers reduces performance and health, highlighting the importance of accurately measuring intestinal permeability (IP) to maintain gut health. The objective of this study was to evaluate the efficiency of iohexol as an IP marker in broilers challenged with Eimeria maxima, Clostridium perfringens, or both during both peak challenge (day [d] 21) and recovery (d 28) periods. One-day-old male Ross 708 birds (n = 56) were distributed into 4 treatment groups: NC (no-challenge control); EM (challenged with 5,000 E. maxima sporulated oocysts/bird on d 15); CP (challenged with 1.0 × 108 CFUs/bird of C. perfringens on d 19 and d 20); and EM + CP (challenged by co-infection of E. maxima and C. perfringens as described). On d 21 and d 28, each bird received an iohexol dose of 64.7 mg/kg body weight via oral gavage. One hour later, blood samples were collected from 14 birds (12 in EM) per group on d 21 and from 7 birds (6 in EM) on d 28. For lesion scoring and ileum collection, 7 birds per group (6 birds in EM) were sampled on each d 21 and d 28. Birds in the EM and EM + CP groups had lower body weight gain (BWG) compared to the NC and CP groups on d 19-21 (P ≤ 0.05). These birds also exhibited significantly greater lesion scores and markedly higher serum iohexol levels on d 21 (P ≤ 0.05). However, no significant differences in serum iohexol levels were observed among treatment groups following recovery on d 28. Moreover, significant differentials were observed in the mRNA abundance of key tight junction proteins (CLDN1, CLDN2, and ZO3), pro-inflammatory cytokines (IL-1β, IFNγ, and IL-22), and gut health markers (GLP2, OLFM4, and MUC2) in the EM and EM + CP groups compared to the NC and CP groups on d 21. In conclusion, this study demonstrates that iohexol is an effective marker for assessing IP in broilers under different enteric challenge conditions.
Collapse
Affiliation(s)
- Ali Calik
- Avian Immunobiology Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, United States
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Ankara University, Ankara, Türkiye
| | - Abhisek Niraula
- Avian Immunobiology Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Bingqi Dong
- Avian Immunobiology Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Candice E. C. Blue
- Avian Immunobiology Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Davis A. Fenster
- Avian Immunobiology Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - Rami A. Dalloul
- Avian Immunobiology Laboratory, Department of Poultry Science, University of Georgia, Athens, GA, United States
| |
Collapse
|
12
|
Shahid A, Chambers S, Scott-Thomas A, Bhatia M. Gut Microbiota and Liver Dysfunction in Sepsis: The Role of Inflammatory Mediators and Therapeutic Approaches. Int J Mol Sci 2024; 25:13415. [PMID: 39769181 PMCID: PMC11678143 DOI: 10.3390/ijms252413415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sepsis is a life-threatening complication caused by an uncontrolled immune response to infection that can lead to multi-organ dysfunction, including liver injury. Recent research has shown the critical role of gut microbiota in sepsis pathogenesis, with the gut-liver axis playing a crucial role in disease progression. Mechanisms such as the disruption of the gut barrier and liver injury pathways mediated by cytokines, chemokines, adhesion molecules, hydrogen sulfide (H2S). and substance P (SP) have been the focus of recent studies. Some potential biomarkers and gut microbiota-targeted therapies have shown promise as emerging tools for predicting and managing sepsis. This review describes the role of the gut-liver axis in sepsis and the potential of microbiota-targeted therapies and biomarker-driven interventions to improve sepsis outcomes.
Collapse
Affiliation(s)
| | | | | | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (A.S.); (S.C.); (A.S.-T.)
| |
Collapse
|
13
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
14
|
Huang B, Lin G, Chen F, Yang W, Zhang C, Yao Y, Zeng Q, Yang Y, Huang J. UCP2 knockout exacerbates sepsis-induced intestinal injury by promoting NLRP3-mediated pyroptosis. Int Immunopharmacol 2024; 141:112935. [PMID: 39159561 DOI: 10.1016/j.intimp.2024.112935] [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: 07/05/2024] [Revised: 07/05/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024]
Abstract
Sepsis-induced intestinal injury is a common complication that increases the morbidity and mortality associated with sepsis. UCP2, a mitochondrial membrane protein, is involved in numerous cellular processes, including metabolism, inflammation, and pyroptosis. According to our previous studies, UCP2 expression increases in septic intestinal tissue. However, its function in intestinal damage is not known. This work investigated UCP2's role in intestinal injury caused by sepsis. A sepsis mouse model was established in wild-type and UCP2-knockout (UCP2-KO) animals using cecal ligation and puncture (CLP). MCC950, an NLRP3 inflammasome inhibitor, was injected intraperitoneally 3 h before CLP surgery. Overall, significantly higher levels of UCP2 were observed in the intestines of septic mice. UCP2-KO mice subjected to CLP exhibited exacerbated intestinal damage, characterized by enhanced mucosal erosion, inflammatory cell infiltration, and increased intestinal permeability. Furthermore, UCP2 knockout significantly increased oxidative stress, inflammation, and pyroptosis in the CLP mouse intestines. Interestingly, MCC950 not only inhibited pyroptosis but also reversed inflammation, oxidative stress as well as damage to intestinal tissues as a result of UCP2 knockout. Our results highlighted the protective functions of UCP2 in sepsis-associated intestinal injury through modulation of inflammation and oxidative stress via NLRP3 inflammasome-induced pyroptosis.
Collapse
Affiliation(s)
- Bolun Huang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Gangxi Lin
- The School of Clinical Medicine, Fujian Medical University, Fuzhou 350007, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361003, China
| | - Feiyan Chen
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Wenmin Yang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Chunmin Zhang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Yu Yao
- Department of Otolaryngology Head & Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qiyi Zeng
- Department of Pediatrics, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiyu Yang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| | - Jinda Huang
- Department of Pediatric Intensive Care Unit, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China.
| |
Collapse
|
15
|
Yumoto T, Coopersmith CM. Targeting AMP-activated protein kinase in sepsis. Front Endocrinol (Lausanne) 2024; 15:1452993. [PMID: 39469575 PMCID: PMC11513325 DOI: 10.3389/fendo.2024.1452993] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Sepsis is a global health challenge marked by limited clinical options and high mortality rates. AMP-activated protein kinase (AMPK) is a cellular energy sensor that mediates multiple crucial metabolic pathways that may be an attractive therapeutic target in sepsis. Pre-clinical experimental studies have demonstrated that pharmacological activation of AMPK can offer multiple potential benefits during sepsis, including anti-inflammatory effects, induction of autophagy, promotion of mitochondrial biogenesis, enhanced phagocytosis, antimicrobial properties, and regulation of tight junction assembly. This review aims to discuss the existing evidence supporting the therapeutic potential of AMPK activation in sepsis management.
Collapse
Affiliation(s)
- Tetsuya Yumoto
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
- Department of Emergency, Critical Care and Disaster Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Craig M. Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
16
|
Piccioni A, Spagnuolo F, Candelli M, Voza A, Covino M, Gasbarrini A, Franceschi F. The Gut Microbiome in Sepsis: From Dysbiosis to Personalized Therapy. J Clin Med 2024; 13:6082. [PMID: 39458032 PMCID: PMC11508704 DOI: 10.3390/jcm13206082] [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: 08/28/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Sepsis is a complex clinical syndrome characterized by an uncontrolled inflammatory response to an infection that may result in septic shock and death. Recent research has revealed a crucial link between sepsis and alterations in the gut microbiota, showing that the microbiome could serve an essential function in its pathogenesis and prognosis. In sepsis, the gut microbiota undergoes significant dysbiosis, transitioning from a beneficial commensal flora to a predominance of pathobionts. This transformation can lead to a dysfunction of the intestinal barrier, compromising the host's immune response, which contributes to the severity of the disease. The gut microbiota is an intricate system of protozoa, fungi, bacteria, and viruses that are essential for maintaining immunity and metabolic balance. In sepsis, there is a reduction in microbial heterogeneity and a predominance of pathogenic bacteria, such as proteobacteria, which can exacerbate inflammation and negatively influence clinical outcomes. Microbial compounds, such as short-chain fatty acids (SCFAs), perform a crucial task in modulating the inflammatory response and maintaining intestinal barrier function. However, the role of other microbiota components, such as viruses and fungi, in sepsis remains unclear. Innovative therapeutic strategies aim to modulate the gut microbiota to improve the management of sepsis. These include selective digestive decontamination (SDD), probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT), all of which have shown potential, although variable, results. The future of sepsis management could benefit greatly from personalized treatment based on the microbiota. Rapid and easy-to-implement tests to assess microbiome profiles and metabolites associated with sepsis could revolutionize the disease's diagnosis and management. These approaches could not only improve patient prognosis but also reduce dependence on antibiotic therapies and promote more targeted and sustainable treatment strategies. Nevertheless, there is still limited clarity regarding the ideal composition of the microbiota, which should be further characterized in the near future. Similarly, the benefits of therapeutic approaches should be validated through additional studies.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Fabio Spagnuolo
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Antonio Voza
- Department of Emergency Medicine, IRCCS-Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Medical and Surgical Science Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
17
|
Bendas G, Gobec M, Schlesinger M. Modulating Immune Responses: The Double-Edged Sword of Platelet CD40L. Semin Thromb Hemost 2024. [PMID: 39379039 DOI: 10.1055/s-0044-1791512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The CD40-CD40L receptor ligand pair plays a fundamental role in the modulation of the innate as well as the adaptive immune response, regulating monocyte, T and B cell activation, and antibody isotype switching. Although the expression and function of the CD40-CD40L dyad is mainly attributed to the classical immune cells, the majority of CD40L is expressed by activated platelets, either in a membrane-bound form or shed as soluble molecules in the circulation. Platelet-derived CD40L is involved in the communication with different immune cell subpopulations and regulates their functions effectively. Thus, platelet CD40L contributes to the containment and clearance of bacterial and viral infections, and additionally guides leukocytes to sites of infection. However, platelet CD40L promotes inflammatory cellular responses also in a pathophysiological context. For example, in HIV infections, platelet CD40L is supportive of neuronal inflammation, damage, and finally HIV-related dementia. In sepsis, platelet CD40L can induce extensive endothelial and epithelial damage resulting in barrier dysfunction of the gut, whereby the translocation of microbiota into the circulation further aggravates the uncontrolled systemic inflammation. Nevertheless, a distinct platelet subpopulation expressing CD40L under septic conditions can attenuate systemic inflammation and reduce mortality in mice. This review focuses on recent findings in the field of platelet CD40L biology and its physiological and pathophysiological implications, and thereby highlights platelets as vital immune cells that are essential for a proper immune surveillance. In this context, platelet CD40L proves to be an interesting target for various inflammatory diseases. However, either an agonism or a blockade of CD40L needs to be well balanced since both the approaches can cause severe adverse events, ranging from hyperinflammation to immune deficiency. Thus, an interference in CD40L activities should be likely done in a context-dependent and timely restricted manner.
Collapse
Affiliation(s)
- Gerd Bendas
- Department of Pharmacy, University of Bonn, Bonn, Germany
| | - Martina Gobec
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Martin Schlesinger
- Department of Pharmacy, University of Bonn, Bonn, Germany
- Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| |
Collapse
|
18
|
Wang J, Wang XY, Yuan ZY, Wang XH, Guan YY, Zhu JX, Huang WF, Liu Q, Xu GH, Yi LT. Blueberry extract attenuates DSS-induced inflammatory bowel disease in mice through inhibiting ER stress-mediated colonic apoptosis in mice. Food Funct 2024; 15:9541-9551. [PMID: 39225067 DOI: 10.1039/d4fo00194j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic, debilitating condition with limited therapeutic options. Dietary components like blueberries have emerged as potential modulators of inflammation and tissue repair in gastrointestinal diseases. This study investigated endoplasmic reticulum (ER) stress-mediated apoptosis mediated protective effects of blueberries in ameliorating dextran sulfate sodium (DSS)-induced IBD. Firstly, a total of 86 anthocyanin compounds were identified in blueberry extract by LC-MS spectroscopy, including 35 cyanidin, 9 delphinidin, 14 malvidin, 10 peonidin, and 9 petunidin. Then, the animal study showed that blueberry supplementation notably ameliorated DSS-induced IBD symptoms, as evidenced by improved histopathological scores and a reduced disease activity index (DAI) score. Additionally, blueberries attenuated ER stress by inhibiting the colonic PERK/eIF2α/ATF4/CHOP signaling pathway. Furthermore, blueberries inhibited the expression of the pro-apoptotic protein, caspase-3, and decreased colonic apoptosis, as evidenced by TUNEL assay results. However, it did not affect the expression of anti-apoptotic proteins, bcl-2 and bcl-xl. Finally, blueberries enhanced the intestinal barrier by upregulating ZO-1, claudin-1, occludin, and E-cadherin. In conclusion, blueberries demonstrate therapeutic potential against DSS-induced IBD-like symptoms in mice, possibly by regulating ER stress-mediated apoptosis pathways. These findings suggest that blueberries might be an effective dietary intervention for IBD management.
Collapse
Affiliation(s)
- Jun Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Xin-Yu Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Zhong-Yu Yuan
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Xiao-Han Wang
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Yu-Ying Guan
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Ji-Xiao Zhu
- Research Center of Traditional Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, PR China
| | - Wei-Feng Huang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen 361003, Fujian Province, PR China
| | - Qing Liu
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
| | - Guang-Hui Xu
- Xiamen Medicine Research Institute, Xiamen 361008, Fujian Province, PR China
| | - Li-Tao Yi
- Department of Chemical and Pharmaceutical Engineering, College of Chemical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China.
- Institute of Pharmaceutical Engineering, Huaqiao University, Xiamen 361021, Fujian Province, PR China
- Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen, 361021, Fujian Province, PR China
| |
Collapse
|
19
|
Li Z, Wang Y, Huang W, Shi X, Ma T, Yu X. miR-155 induces sepsis-associated damage to the intestinal mucosal barrier via sirtuin 1/nuclear factor-κB-mediated intestinal pyroptosis. Acta Biochim Biophys Sin (Shanghai) 2024. [PMID: 39262326 DOI: 10.3724/abbs.2024124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024] Open
Abstract
Sepsis is a life-threatening state of organ dysfunction caused by systemic inflammation and a dysfunctional response to host infections that can induce severe intestinal mucosal damage. Pyroptosis is mediated by the activated NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome after stimulation by various inflammatory factors during sepsis. The inflammatory response is a major driver of intestinal damage during sepsis. Intestinal mucosal barrier dysfunction in sepsis is associated with pyroptosis, a type of programmed inflammatory cell death. Several studies have confirmed the role of miR-155 in sepsis and other diseases. However, the effect of miR-155 on intestinal pyroptosis in the context of intestinal mucosal barrier dysfunction during sepsis remains unclear. Thus, a model of sepsis in Sprague-Dawley rats is established using cecal ligation and puncture (CLP), and a series of molecular biological methods are used in this study. The results show that the expression of miR-155 is increased and that of sirtuin 1 (SIRT1) is decreased in the intestinal tissues of patients with sepsis. miR-155 expression is negatively correlated with SIRT1 expression. Increased miR-155 expression significantly inhibits SIRT1 activity and upregulates the expressions of NOD-like receptor family pyrin domain-containing 3 (NLRP3), caspase-1, apoptosis-associated speck-like protein containing a CARD (ASC), interleukin-1β (IL-1β) and interleukin-18 (IL-18) to promote pyroptosis. The inhibition of miR-155 expression is associated with increased SIRT1 expression, promotes the deacetylation of p65, and significantly downregulates p65 acetylation. Herein, we propose that miR-155 induces pyroptosis in the intestine partly by regulating SIRT1, thereby reducing the deacetylation of the nuclear factor (NF)-κB subunit p65 and increasing NF-κB signaling activity in sepsis, leading to intestinal barrier damage.
Collapse
Affiliation(s)
- Zhihua Li
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Yi Wang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
- Department of Critical Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Weiwei Huang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Xingyu Shi
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Tao Ma
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - Xiangyou Yu
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
- Department of Critical Medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
20
|
Zhai Z, Yang Y, Chen S, Wu Z. Long-Term Exposure to Polystyrene Microspheres and High-Fat Diet-Induced Obesity in Mice: Evaluating a Role for Microbiota Dysbiosis. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:97002. [PMID: 39226184 PMCID: PMC11370995 DOI: 10.1289/ehp13913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 07/23/2024] [Accepted: 08/08/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Microplastics (MPs) have become a global environmental problem, emerging as contaminants with potentially alarming consequences. However, long-term exposure to polystyrene microspheres (PS-MS) and its effects on diet-induced obesity are not yet fully understood. OBJECTIVES We aimed to investigate the effect of PS-MS exposure on high-fat diet (HFD)-induced obesity and underlying mechanisms. METHODS In the present study, C57BL/6J mice were fed a normal diet (ND) or a HFD in the absence or presence of PS-MS via oral administration for 8 wk. Antibiotic depletion of the microbiota and fecal microbiota transplantation (FMT) were performed to assess the influence of PS-MS on intestinal microbial ecology. We performed 16S rRNA sequencing to dissect microbial discrepancies and investigated the dysbiosis-associated intestinal integrity and inflammation in serum. RESULTS Compared with HFD mice, mice fed the HFD with PS-MS exhibited higher body weight, liver weight, metabolic dysfunction-associated steatotic liver disease (MASLD) activity scores, and mass of white adipose tissue, as well as higher blood glucose and serum lipid concentrations. Furthermore, 16S rRNA sequencing of the fecal microbiota revealed that mice fed the HFD with PS-MS had greater α -diversity and greater relative abundances of Lachnospiraceae, Oscillospiraceae, Bacteroidaceae, Akkermansiaceae, Marinifilaceae, Deferribacteres, and Desulfovibrio, but lower relative abundances of Atopobiaceae, Bifidobacterium, and Parabacteroides. Mice fed the HFD with PS-MS exhibited lower expression of MUC2 mucin and higher levels of lipopolysaccharide and inflammatory cytokines [tumor necrosis factor-α (TNF-α ), interleukin-6 (IL-6), IL-1β , and IL-17A] in serum. Correlation analyses revealed that differences in the microbial flora of mice exposed to PS-MS were associated with obesity. Interestingly, microbiota-depleted mice did not show the same PS-MS-associated differences in Muc2 and Tjp1 expression in the distal colon, expression of inflammatory cytokines in serum, or obesity outcomes between HFD and HFD + PS-MS. Importantly, transplantation of feces from HFD + PS-MS mice to microbiota-depleted HFD-fed mice resulted in a lower expression of mucus proteins, higher expression of inflammatory cytokines, and obesity outcomes, similar to the findings in HFD + PS-MS mice. CONCLUSIONS Our findings provide a new gut microbiota-driven mechanism for PS-MS-induced obesity in HFD-fed mice, suggesting the need to reevaluate the adverse health effects of MPs commonly found in daily life, particularly in susceptible populations. https://doi.org/10.1289/EHP13913.
Collapse
Affiliation(s)
- Zhian Zhai
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Ying Yang
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
| | - Sheng Chen
- State Key Lab of Chemical Biology and Drug Discovery, Hong Kong Polytechnic University, Kowloon, Hong Kong, China
- Department of Food Science and Nutrition, Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Zhenlong Wu
- Department of Companion Animal Science, State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
21
|
Cao YY, Wang ZH, Pan YJ, Qi YP, Chen Q, Qin XM, Wang T, Shen GG, Jiang XG, Lu WH. The dual effects of dexmedetomidine on intestinal barrier and intestinal motility during sepsis. Surgery 2024; 176:379-385. [PMID: 38762380 DOI: 10.1016/j.surg.2024.03.047] [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: 10/13/2023] [Revised: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Sepsis, characterized by dysregulated host responses to infection, remains a critical global health concern, with high morbidity and mortality rates. The gastrointestinal tract assumes a pivotal role in sepsis due to its dual functionality as a protective barrier against injurious agents and as a regulator of motility. Dexmedetomidine, an α2-adrenergic agonist commonly employed in critical care settings, exhibits promise in influencing the maintenance of intestinal barrier integrity during sepsis. However, its impact on intestinal motility, a crucial component of intestinal function, remains incompletely understood. METHODS In this study, we investigated dexmedetomidine's multifaceted effects on intestinal barrier function and motility during sepsis using both in vitro and in vivo models. Sepsis was induced in Sprague-Dawley rats via cecal ligation and puncture. Rats were treated with dexmedetomidine post-cecal ligation and puncture, and various parameters were assessed to elucidate dexmedetomidine's impact. RESULTS Our findings revealed a dichotomous influence of dexmedetomidine on intestinal physiology. In septic rats, dexmedetomidine administration resulted in improved intestinal barrier integrity, as evidenced by reduced mucosal hyper-permeability and morphological alterations. However, a contrasting effect was observed on intestinal motility, as dexmedetomidine treatment inhibited both the frequency and amplitude of contractions in isolated intestinal strips and decreased the distance of ink migration in vivo. Additionally, dexmedetomidine suppressed the secretion of pro-motility hormones while having no influence on hormones that inhibit intestinal peristalsis. CONCLUSION The study revealed that during sepsis, dexmedetomidine exhibited protective effects on barrier integrity, although concurrently it hindered intestinal motility, partly attributed to its modulation of pro-motility hormone secretion. These findings underscore the necessity of a comprehensive understanding of dexmedetomidine's impact on multiple facets of gastrointestinal physiology in sepsis management, offering potential implications for therapeutic strategies and patient care.
Collapse
Affiliation(s)
- Ying-Ya Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Zhong-Han Wang
- Department of Anesthesiology, The People's Hospital of Bozhou, Bozhou, Anhui, China
| | - You-Jun Pan
- Department of Critical Care Medicine, Wuhu Hospital, East China Normal University (The Second People's Hospital, Wuhu), Wuhu, Anhui, China
| | - Yu-Peng Qi
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Qun Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Xue-Mei Qin
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Tong Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Guang-Gui Shen
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Xiao-Gan Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China.
| | - Wei-Hua Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China.
| |
Collapse
|
22
|
Bajgai B, Suri M, Singh H, Hanifa M, Bhatti JS, Randhawa PK, Bali A. Naringin: A flavanone with a multifaceted target against sepsis-associated organ injuries. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155707. [PMID: 38788393 DOI: 10.1016/j.phymed.2024.155707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/16/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Sepsis causes multiple organ dysfunctions and raises mortality and morbidity rates through a dysregulated host response to infection. Despite the growing research interest over the last few years, no satisfactory treatment exists. Naringin, a naturally occurring bioflavonoid with vast therapeutic potential in citrus fruits and Chinese herbs, has received much attention for treating sepsis-associated multiple organ dysfunctions. PURPOSE The review describes preclinical evidence of naringin from 2011 to 2024, particularly emphasizing the mechanism of action mediated by naringin against sepsis-associated specific injuries. The combination therapy, safety profile, drug interactions, recent advancements in formulation, and future perspectives of naringin are also discussed. METHODS In vivo and in vitro studies focusing on the potential role of naringin and its mechanism of action against sepsis-associated organ injuries were identified and summarised in the present manuscript, which includes contributions from 2011 to 2024. All the articles were extracted from the Medline database using PubMed, Science Direct, and Web of Science with relevant keywords. RESULTS Research findings revealed that naringin modulates many signaling cascades, such as Rho/ROCK and PPAR/STAT1, PIP3/AKT and KEAP1/Nrf2, and IkB/NF-kB and MAPK/Nrf2/HO-1, to potentially protect against sepsis-induced intestinal, cardiac, and lung injury, respectively. Furthermore, naringin treatment exhibits anti-inflammatory, anti-apoptotic, and antioxidant action against sepsis harm, highlighting naringin's promising effects in septic settings. Naringin could be employed as a treatment against sepsis, based on studies on combination therapy, synergistic effects, and toxicological investigation that show no reported severe side effects. CONCLUSION Naringin might be a promising therapeutic approach for preventing sepsis-induced multiple organ failure. Naringin should be used alongside other therapeutic therapies with caution despite its great therapeutic potential and lower toxicity. Nonetheless, clinical studies are required to comprehend the therapeutic benefits of naringin against sepsis.
Collapse
Affiliation(s)
- Bivek Bajgai
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Manisha Suri
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Harshita Singh
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Mohd Hanifa
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Jasvinder Singh Bhatti
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kaur Randhawa
- Department of Pharmaceutical Sciences, Amritsar Group of Colleges, Amritsar, Punjab, 143001, India; Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Anjana Bali
- Laboratory of Neuroendocrinology, Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
23
|
Nofi CP, Prince JM, Aziz M, Wang P. The Novel MFG-E8-derived Oligopeptide, MOP3, Improves Outcomes in a Preclinical Murine Model of Neonatal Sepsis. J Pediatr Surg 2024; 59:1282-1290. [PMID: 38582704 DOI: 10.1016/j.jpedsurg.2024.03.025] [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: 02/21/2024] [Accepted: 03/04/2024] [Indexed: 04/08/2024]
Abstract
INTRODUCTION Neonatal sepsis is a devastating inflammatory condition that remains a leading cause of morbidity and mortality. Milk fat globule-EGF-factor VIII (MFG-E8) is a glycoprotein that reduces inflammation, whereas extracellular cold-inducible RNA binding protein (eCIRP) worsens inflammation. This study aimed to determine the therapeutic potential of a novel MFG-E8-derived oligopeptide 3 (MOP3) designed to clear eCIRP and protect against inflammation, organ injury, and mortality in neonatal sepsis. METHODS C57BL6 mouse pups were injected intraperitoneally with cecal slurry (CS) and treated with MOP3 (20 μg/g) or vehicle. 10 h after injection, blood, lungs, and intestines were collected for analyses, and in a 7-day experiment, pups were monitored for differences in mortality. RESULTS MOP3 treatment protected septic pups from inflammation by reducing eCIRP, IL-6, TNFα, and LDH. MOP3 reduced lung and intestinal inflammation and injury as assessed by reductions in tissue mRNA levels of inflammatory markers, histopathologic injury, and apoptosis in lung and intestines. MOP3 also significantly improved 7-day overall survival for CS-septic mouse pups compared to vehicle (75% vs. 46%, respectively). CONCLUSION Deriving from MFG-E8 and designed to clear eCIRP, MOP3 protects against sepsis-induced inflammation, organ injury, and mortality in a preclinical model of neonatal sepsis, implicating it as an exciting potential new therapeutic. LEVEL OF EVIDENCE Level 1.
Collapse
Affiliation(s)
- Colleen P Nofi
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA.
| | - Jose M Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA; Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, USA; Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, USA; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA; Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| |
Collapse
|
24
|
da Silveira BC, da Silva Platner F, da Rosa LB, Silva MLC, da Silva KS, de Oliveira NMT, Moffa EB, Silva KF, Lima-Neto LG, Maria-Ferreira D, Cordeiro LMC, Gois MB, Fernandes ES. Oral Treatment with the Pectin Fibre Obtained from Yellow Passion Fruit Peels Worsens Sepsis Outcome in Mice by Affecting the Intestinal Barrier. Pharmaceuticals (Basel) 2024; 17:863. [PMID: 39065714 PMCID: PMC11279511 DOI: 10.3390/ph17070863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The biological activities of plant-derived soluble dietary fibres (SDFs) have been widely investigated. Pectin from yellow passion fruit (YPF-peSDF) peels was suggested as a protective macromolecule in ulcers and colitis due to its antioxidant and anti-inflammatory properties. Sepsis has high mortality and morbidity and is characterised by inflammatory and oxidative stress imbalances. Evidence suggests that pectins may aid sepsis treatment; however, the effects of YPF-peSDF on sepsis remain unclear. Herein, polymicrobial sepsis was induced by cecal-ligation and puncture in mice treated with YPF-peSDF (1 and 10 mg/kg; gavage). YPF-peSDF accelerated mortality, reaching 100% in 24 h. Inflammation was present in the colons and small intestines (SI) of both vehicle- and fibre-treated mice. Although crypt depth and width, and villus height were preserved in the SI of septic mice administered YPF-peSDF, they exhibited exacerbated muscle layer atrophy and mucosa and submucosa hypertrophy, along with shortened enterocytes. Larger crypts and shorter enterocytes were noted in their colons in comparison with vehicle-controls. YPF-peSDF also reduced inflammatory cell numbers and exacerbated IL-6 levels in peritoneal lavage fluid (PELF) samples. YPF-peSDF modulated SI but not colon cytokines. Lipoperoxidation and antioxidant capacity levels were attenuated in PELF samples. Overall, in contrast to previous evidence, YPF-peSDF worsened polymicrobial sepsis outcomes in mice.
Collapse
Affiliation(s)
- Bruna C. da Silveira
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Fernanda da Silva Platner
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Liza B. da Rosa
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Matheus L. C. Silva
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Karien S. da Silva
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Natalia M. T. de Oliveira
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Eduardo B. Moffa
- College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada;
| | - Karinny F. Silva
- Programa de Pós-Graduação, Universidade Ceuma, São Luís 65075-120, MA, Brazil; (K.F.S.); (L.G.L.-N.)
| | - Lídio G. Lima-Neto
- Programa de Pós-Graduação, Universidade Ceuma, São Luís 65075-120, MA, Brazil; (K.F.S.); (L.G.L.-N.)
| | - Daniele Maria-Ferreira
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| | - Lucimara M. C. Cordeiro
- Departmento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Curitiba 81531-980, PR, Brazil;
| | - Marcelo B. Gois
- Faculdade de Ciências da Saúde, Universidade Federal de Rondonópolis, Rondonópolis 78740-393, MT, Brazil;
| | - Elizabeth S. Fernandes
- Programa de Pós-Graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba 80230-020, PR, Brazil; (B.C.d.S.); (F.d.S.P.); (L.B.d.R.); (M.L.C.S.); (K.S.d.S.); (N.M.T.d.O.); (D.M.-F.)
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, PR, Brazil
| |
Collapse
|
25
|
Wang L, Chen Y, Wu H, Yu HH, Ma L. Slit2-Robo4 signal pathway and tight junction in intestine mediate LPS-induced inflammation in mice. Eur J Med Res 2024; 29:349. [PMID: 38937814 PMCID: PMC11209965 DOI: 10.1186/s40001-024-01894-5] [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: 01/14/2024] [Accepted: 05/21/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Sepsis is one of the most common clinical diseases, which is characterized by a serious and uncontrollable inflammatory response. LPS-induced inflammation is a critical pathological event in sepsis, but the underlying mechanism has not yet been fully elucidated. METHODS The animal model was established for two batches. In the first batch of experiments, Adult C57BL/6J mice were randomly divided into control group and LPS (5 mg/kg, i.p.)group . In the second batch of experiments, mice were randomly divided into control group, LPS group, and LPS+VX765(10 mg/kg, i.p., an inhibitor of NLRP3 inflammasome) group. After 24 hours, mice were anesthetized with isoflurane, blood and intestinal tissue were collected for tissue immunohistochemistry, Western blot analysis and ELISA assays. RESULTS The C57BL/6J mice injected with LPS for twenty-four hours could exhibit severe inflammatory reaction including an increased IL-1β, IL-18 in serum and activation of NLRP3 inflammasome in intestine. The injection of VX765 could reverse these effects induced by LPS. These results indicated that the increased level of IL-1β and IL-18 in serum induced by LPS is related to the increased intestinal permeability and activation of NLRP3 inflammasome. In the second batch of experiments, results of western blot and immunohistochemistry showed that Slit2 and Robo4 were significant decreased in intestine of LPS group, while the expression of VEGF was significant increased. Meanwhile, the protein level of tight junction protein ZO-1, occludin, and claudin-5 were significantly lower than in control group, which could also be reversed by VX765 injection. CONCLUSIONS In this study, we revealed that Slit2-Robo4 signaling pathway and tight junction in intestine may be involved in LPS-induced inflammation in mice, which may account for the molecular mechanism of sepsis.
Collapse
Affiliation(s)
- Lv Wang
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - Yingtai Chen
- Emergency Department, Baoshan Branch of Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200444, People's Republic of China
| | - Hao Wu
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China
| | - He-Hua Yu
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China.
| | - Linhao Ma
- Department of Emergency and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, People's Republic of China.
| |
Collapse
|
26
|
Yao Y, Ba T, Bao B, Zhang S, Kong L. Sepsis as a Potential Risk Factor for Upper Gastrointestinal Bleeding in Critically Ill Patients: A Systematic Review and Meta-analysis. J Intensive Care Med 2024:8850666241252048. [PMID: 38813775 DOI: 10.1177/08850666241252048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Purpose: Sepsis is a common and critical condition in intensive care units (ICUs) known to complicate patient outcomes. Previous studies have indicated an association between sepsis and various ICU morbidities, including upper gastrointestinal bleeding (UGIB). However, the extent of this relationship and its implications in ICU settings remain inadequately quantified. This study aims to elucidate the association between sepsis and the risk of UGIB in ICU patients. Methods: A comprehensive meta-analysis was conducted, encompassing nine studies with a total of nearly 9000 participants. These studies reported events for both sepsis and nonsepsis patients separately. Pooled odds ratios (ORs) were calculated to assess the risk of UGIB in septic versus nonseptic ICU patients. Subgroup analyses were conducted based on age and study design, and both unadjusted and adjusted ORs were examined. Results: The pooled OR indicated a significant association between sepsis and UGIB (OR = 3.276, 95% CI: 1.931 to 5.557). Moderate heterogeneity was observed (I² = 43.9%). The association was significant in adults (pooled OR = 4.083) but not in children. No difference in association was found based on the study design. Unadjusted and adjusted ORs differed slightly, indicating the influence of confounding factors. Conclusion: This meta-analysis reveals a significant association between sepsis and an increased risk of UGIB in ICU patients, particularly in adults. These findings highlight the need for vigilant monitoring and proactive management of septic ICU patients to mitigate the risk of UGIB. Future research should focus on understanding the underlying mechanisms and developing tailored preventive strategies.
Collapse
Affiliation(s)
- Yanfen Yao
- Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Shandong University, Jinan, China
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tejin Ba
- Department of Emergency, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Bagenna Bao
- Department of Emergency, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Shuanglin Zhang
- Department of Emergency, International Mongolian Hospital of Inner Mongolia, Hohhot, China
| | - Li Kong
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Emergency Center, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
27
|
Ziaka M, Exadaktylos A. Exploring the lung-gut direction of the gut-lung axis in patients with ARDS. Crit Care 2024; 28:179. [PMID: 38802959 PMCID: PMC11131229 DOI: 10.1186/s13054-024-04966-4] [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: 03/12/2024] [Accepted: 05/22/2024] [Indexed: 05/29/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) represents a life-threatening inflammatory reaction marked by refractory hypoxaemia and pulmonary oedema. Despite advancements in treatment perspectives, ARDS still carries a high mortality rate, often due to systemic inflammatory responses leading to multiple organ dysfunction syndrome (MODS). Indeed, the deterioration and associated mortality in patients with acute lung injury (LI)/ARDS is believed to originate alongside respiratory failure mainly from the involvement of extrapulmonary organs, a consequence of the complex interaction between initial inflammatory cascades related to the primary event and ongoing mechanical ventilation-induced injury resulting in multiple organ failure (MOF) and potentially death. Even though recent research has increasingly highlighted the role of the gastrointestinal tract in this process, the pathophysiology of gut dysfunction in patients with ARDS remains mainly underexplored. This review aims to elucidate the complex interplay between lung and gut in patients with LI/ARDS. We will examine various factors, including systemic inflammation, epithelial barrier dysfunction, the effects of mechanical ventilation (MV), hypercapnia, and gut dysbiosis. Understanding these factors and their interaction may provide valuable insights into the pathophysiology of ARDS and potential therapeutic strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic of Geriatric Medicine, Center of Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
28
|
Huang FC, Huang SC. Unveiling the Novel Benefits of Co-Administering Butyrate and Active Vitamin D3 in Mice Subjected to Chemotherapy-Induced Gut-Derived Pseudomonas aeruginosa Sepsis. Biomedicines 2024; 12:1026. [PMID: 38790988 PMCID: PMC11118095 DOI: 10.3390/biomedicines12051026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/22/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer patients face increased susceptibility to invasive infections, primarily due to ulcerative lesions on mucosal surfaces and immune suppression resulting from chemotherapy. Pseudomonas aeruginosa (P. aeruginosa) bacteremia is notorious for its rapid progression into fatal sepsis, posing a significant threat to cancer patients, particularly those experiencing chemotherapy-induced neutropenia. This bacterial infection contributes significantly to morbidity and mortality rates among such individuals. Our latest report showed the mutually beneficial effects of postbiotic butyrate on 1,25-dihydroxyvitamin D3 (1,25D3)-controlled innate immunity during Salmonella colitis. Hence, we investigated the impact of butyrate and 1,25D3 on chemotherapy-induced gut-derived P. aeruginosa sepsis in mice. The chemotherapy-induced gut-derived P. aeruginosa sepsis model was established through oral administration of 1 × 107 CFU of the P. aeruginosa wild-type strain PAO1 in C57BL/6 mice undergoing chemotherapy. Throughout the infection process, mice were orally administered butyrate and/or 1,25D3. Our observations revealed that the combined action of butyrate and 1,25D3 led to a reduction in the severity of colitis and the invasion of P. aeruginosa into the liver and spleen of the mice. This reduction was attributed to an enhancement in the expression of defensive cytokines and antimicrobial peptides within the cecum, coupled with decreased levels of zonulin and claudin-2 proteins in the mucosal lining. These effects were notably more pronounced when compared to treatments administered individually. This study unveils a promising alternative therapy that involves combining postbiotics and 1,25D3 for treating chemotherapy-induced gut-derived P. aeruginosa sepsis.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shun-Chen Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| |
Collapse
|
29
|
Sun J, Li J, Deng Y, Yin X, Huangfu X, Ye Z, Zhou X, Chen Y, Yuan S, Wang X. The beneficial effects of neutrophil elastase inhibitor on gastrointestinal dysfunction in sepsis. Clin Transl Sci 2024; 17:e13829. [PMID: 38769746 PMCID: PMC11106555 DOI: 10.1111/cts.13829] [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: 01/27/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/22/2024] Open
Abstract
To investigate the effects of neutrophil elastase inhibitor (sivelestat sodium) on gastrointestinal function in sepsis. A reanalysis of the data from previous clinical trials conducted at our center was performed. Septic patients were divided into either the sivelestat group or the non-sivelestat group. The gastrointestinal dysfunction score (GIDS), feeding intolerance (FI) incidence, serum levels of intestinal barrier function and inflammatory biomarkers were recorded. The clinical severity and outcome variables were also documented. A total of 163 septic patients were included. The proportion of patients with GIDS ≥2 in the sivelestat group was reduced relative to that in the non-sivelestat group (9.6% vs. 22.5%, p = 0.047) on the 7th day of intensive care unit (ICU) admission. The FI incidence was also remarkably reduced in the sivelestat group in contrast to that in the non-sivelestat group (21.2% vs. 37.8%, p = 0.034). Furthermore, the sivelestat group had fewer days of FI [4 (3, 4) vs. 5 (4-6), p = 0.008]. The serum levels of d-lactate (p = 0.033), intestinal fatty acid-binding protein (p = 0.005), interleukin-6 (p = 0.001), white blood cells (p = 0.007), C-reactive protein (p = 0.001), and procalcitonin (p < 0.001) of the sivelestat group were lower than those of the non-sivelestat group. The sivelestat group also demonstrated longer ICU-free days [18 (0-22) vs. 13 (0-17), p = 0.004] and ventilator-free days [22 (1-24) vs. 16 (1-19), p = 0.002] compared with the non-sivelestat group. In conclusion, sivelestat sodium administration appears to improve gastrointestinal dysfunction, mitigate dysregulated inflammation, and reduce disease severity in septic patients.
Collapse
Affiliation(s)
- Jia‐Kui Sun
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Jing‐Jing Li
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Yi‐Hang Deng
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xiang Yin
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xiao‐Tian Huangfu
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Zi‐Yu Ye
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xue‐Hui Zhou
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Yong‐Ming Chen
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Shou‐Tao Yuan
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| | - Xiang Wang
- Department of Critical Care Medicine, Nanjing First HospitalNanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
30
|
Dvornikova KA, Platonova ON, Bystrova EY. The Role of TRP Channels in Sepsis and Colitis. Int J Mol Sci 2024; 25:4784. [PMID: 38731999 PMCID: PMC11084600 DOI: 10.3390/ijms25094784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
To date, several members of the transient receptor potential (TRP) channels which provide a wide array of roles have been found in the gastrointestinal tract (GI). The goal of earlier research was to comprehend the intricate signaling cascades that contribute to TRP channel activation as well as how these receptors' activity affects other systems. Moreover, there is a large volume of published studies describing the role of TRP channels in a number of pathological disorders, including inflammatory bowel disease (IBD) and sepsis. Nevertheless, the generalizability of these results is subject to certain limitations. For instance, the study of IBD relies on various animal models and experimental methods, which are unable to precisely imitate the multifactorial chronic disease. The diverse pathophysiological mechanisms and unique susceptibility of animals may account for the inconsistency of the experimental data collected. The main purpose of this study was to conduct a comprehensive review and analysis of existing studies on transient receptor potential (TRP) channels implicating specific models of colitis and sepsis, with particular emphasis on their involvement in pathological disorders such as IBD and sepsis. Furthermore, the text endeavors to evaluate the generalizability of experimental findings, taking into consideration the limitations posed by animal models and experimental methodologies. Finally, we also provide an updated schematic of the most important and possible molecular signaling pathways associated with TRP channels in IBD and sepsis.
Collapse
Affiliation(s)
| | | | - Elena Y. Bystrova
- I.P. Pavlov Institute of Physiology RAS, 199034 St. Petersburg, Russia; (K.A.D.); (O.N.P.)
| |
Collapse
|
31
|
Yan Z, Niu L, Wang S, Gao C, Pan S. Intestinal Piezo1 aggravates intestinal barrier dysfunction during sepsis by mediating Ca 2+ influx. J Transl Med 2024; 22:332. [PMID: 38575957 PMCID: PMC10996241 DOI: 10.1186/s12967-024-05076-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
INTRODUCTION Intestinal barrier dysfunction is a pivotal factor in sepsis progression. The mechanosensitive ion channel Piezo1 is associated with barrier function; however, its role in sepsis-induced intestinal barrier dysfunction remains poorly understood. METHODS The application of cecal ligation and puncture (CLP) modeling was performed on both mice of the wild-type (WT) variety and those with Villin-Piezo1flox/flox genetic makeup to assess the barrier function using in vivo FITC-dextran permeability measurements and immunofluorescence microscopy analysis of tight junctions (TJs) and apoptosis levels. In vitro, Caco-2 monolayers were subjected to TNF-α incubation. Moreover, to modulate Piezo1 activation, GsMTx4 was applied to inhibit Piezo1 activation. The barrier function, intracellular calcium levels, and mitochondrial function were monitored using calcium imaging and immunofluorescence techniques. RESULTS In the intestinal tissues of CLP-induced septic mice, Piezo1 protein levels were notably elevated compared with those in normal mice. Piezo1 has been implicated in the sepsis-mediated disruption of TJs, apoptosis of intestinal epithelial cells, elevated intestinal mucosal permeability, and systemic inflammation in WT mice, whereas these effects were absent in Villin-Piezo1flox/flox CLP mice. In Caco-2 cells, TNF-α prompted calcium influx, an effect reversed by GsMTx4 treatment. Elevated calcium concentrations are correlated with increased accumulation of reactive oxygen species, diminished mitochondrial membrane potential, and TJ disruption. CONCLUSIONS Thus, Piezo1 is a potential contributor to sepsis-induced intestinal barrier dysfunction, influencing apoptosis and TJ modification through calcium influx-mediated mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zimeng Yan
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Lei Niu
- Department of Emergency, Shanghai Jiahui International Hospital, No. 689, Guiping Rd., Shanghai, China
| | - Shangyuan Wang
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China.
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, Shanghai, China.
| |
Collapse
|
32
|
Heimfarth L, Dos Santos KS, Monteiro BS, de Souza Oliveira AK, Coutinho HDM, Menezes IRA, Dos Santos MRV, de Souza Araújo AA, Picot L, de Oliveira Júnior RG, Grougnet R, de Souza Siqueira Quintans J, Quintans-Júnior LJ. The protective effects of naringenin, a citrus flavonoid, non-complexed or complexed with hydroxypropyl-β-cyclodextrin against multiorgan damage caused by neonatal endotoxemia. Int J Biol Macromol 2024; 264:130500. [PMID: 38428770 DOI: 10.1016/j.ijbiomac.2024.130500] [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: 09/30/2023] [Revised: 02/16/2024] [Accepted: 02/17/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Endotoxemia is a severe and dangerous clinical syndrome that results in elevated morbidity, especially in intensive care units. Neonates are particularly susceptible to endotoxemia due to their immature immune systems. There are few effective treatments for neonatal endotoxemia. One group of compounds with potential in the treatment of neonatal inflammatory diseases such as endotoxemia is the flavonoids, mainly due to their antioxidant and anti-inflammatory properties. Among these, naringenin (NGN) is a citrus flavonoid which has already been reported to have anti-inflammatory, antioxidant, anti-nociceptive and anti-cancer effects. Unfortunately, its clinical application is limited by its low solubility and bioavailability. However, cyclodextrins (CDs) have been widely used to improve the solubility of nonpolar drugs and enhance the bioavailability of these natural products. OBJECTIVE We, therefore, aimed to investigate the effects of NGN non-complexed and complexed with hydroxypropyl-β-cyclodextrin (HPβCD) on neonatal endotoxemia injuries in a rodent model and describe the probable molecular mechanisms involved in NGN activities. METHOD We used exposure to a bacterial lipopolysaccharide (LPS) to induce neonatal endotoxemia in the mice. RESULTS It was found that NGN (100 mg/kg i.p.) exposure during the neonatal period reduced leukocyte migration and decreased pro-inflammatory cytokine (TNF-α, IL-1β and IL-6) levels in the lungs, heart, kidneys or cerebral cortex. In addition, NGN upregulated IL-10 production in the lungs and kidneys of neonate mice. The administration of NGN also enhanced antioxidant enzyme catalase and SOD activity, reduced lipid peroxidation and protein carbonylation and increased the reduced sulfhydryl groups in an organ-dependent manner, attenuating the oxidative damage caused by LPS exposure. NGN decreased ERK1/2, p38MAPK and COX-2 activation in the lungs of neonate mice. Moreover, NGN complexed with HPβCD was able to increase the animal survival rate. CONCLUSION NGN attenuated inflammatory and oxidative damage in the lungs, heart and kidneys caused by neonatal endotoxemia through the MAPK signaling pathways regulation. Our results show that NGN has beneficial effects against neonatal endotoxemia and could be useful in the treatment of neonatal inflammatory injuries.
Collapse
Affiliation(s)
- Luana Heimfarth
- Laboratory of Neuroscience and Pharmacological Assay (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe CEP: 49100-000, Brazil
| | - Katielen Silvana Dos Santos
- Laboratory of Neuroscience and Pharmacological Assay (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe CEP: 49100-000, Brazil
| | - Brenda Souza Monteiro
- Laboratory of Neuroscience and Pharmacological Assay (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe CEP: 49100-000, Brazil
| | - Anne Karoline de Souza Oliveira
- Laboratory of Neuroscience and Pharmacological Assay (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe CEP: 49100-000, Brazil
| | | | - Irwin R A Menezes
- Universidade Regional do Cariri - URCA, Departmento de Química Biológica, Crato, CE, Brazil
| | | | | | - Laurent Picot
- UMR CNRS 7266 LIENSs, La Rochelle Université, 17042 La Rochelle, France
| | - Raimundo Gonçalves de Oliveira Júnior
- Laboratoire de Pharmacognosie-UMR CNRS 8638, Faculté de Pharmacie, Université Paris Cité, Paris, France; CiTCoM UMR 8038 CNRS, Faculté Pharmacie, Université Paris Cité, 75006, Paris, France
| | - Raphaël Grougnet
- Laboratoire de Pharmacognosie-UMR CNRS 8638, Faculté de Pharmacie, Université Paris Cité, Paris, France
| | - Jullyana de Souza Siqueira Quintans
- Laboratory of Neuroscience and Pharmacological Assay (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe CEP: 49100-000, Brazil; Graduate Program of Health Sciences, Federal University of Sergipe, Aracaju, Sergipe CEP 49060-025, Brazil
| | - Lucindo José Quintans-Júnior
- Laboratory of Neuroscience and Pharmacological Assay (LANEF), Department of Physiology, Federal University of Sergipe, São Cristóvão, Sergipe CEP: 49100-000, Brazil; Graduate Program of Health Sciences, Federal University of Sergipe, Aracaju, Sergipe CEP 49060-025, Brazil
| |
Collapse
|
33
|
Oami T, Abtahi S, Shimazui T, Chen CW, Sweat YY, Liang Z, Burd EM, Farris AB, Roland JT, Tsukita S, Ford ML, Turner JR, Coopersmith CM. Claudin-2 upregulation enhances intestinal permeability, immune activation, dysbiosis, and mortality in sepsis. Proc Natl Acad Sci U S A 2024; 121:e2217877121. [PMID: 38412124 PMCID: PMC10927519 DOI: 10.1073/pnas.2217877121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/16/2024] [Indexed: 02/29/2024] Open
Abstract
Intestinal epithelial expression of the tight junction protein claudin-2, which forms paracellular cation and water channels, is precisely regulated during development and in disease. Here, we show that small intestinal epithelial claudin-2 expression is selectively upregulated in septic patients. Similar changes occurred in septic mice, where claudin-2 upregulation coincided with increased flux across the paracellular pore pathway. In order to define the significance of these changes, sepsis was induced in claudin-2 knockout (KO) and wild-type (WT) mice. Sepsis-induced increases in pore pathway permeability were prevented by claudin-2 KO. Moreover, claudin-2 deletion reduced interleukin-17 production and T cell activation and limited intestinal damage. These effects were associated with reduced numbers of neutrophils, macrophages, dendritic cells, and bacteria within the peritoneal fluid of septic claudin-2 KO mice. Most strikingly, claudin-2 deletion dramatically enhanced survival in sepsis. Finally, the microbial changes induced by sepsis were less pathogenic in claudin-2 KO mice as survival of healthy WT mice injected with cecal slurry collected from WT mice 24 h after sepsis was far worse than that of healthy WT mice injected with cecal slurry collected from claudin-2 KO mice 24 h after sepsis. Claudin-2 upregulation and increased pore pathway permeability are, therefore, key intermediates that contribute to development of dysbiosis, intestinal damage, inflammation, ineffective pathogen control, and increased mortality in sepsis. The striking impact of claudin-2 deletion on progression of the lethal cascade activated during sepsis suggests that claudin-2 may be an attractive therapeutic target in septic patients.
Collapse
Affiliation(s)
- Takehiko Oami
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba260-8670, Japan
| | - Shabnam Abtahi
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02115
| | - Takashi Shimazui
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
- Department of Emergency and Critical Care Medicine, Chiba University Graduate School of Medicine, Chiba260-8670, Japan
| | - Ching-Wen Chen
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
| | - Yan Y. Sweat
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02115
| | - Zhe Liang
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
| | - Eileen M. Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA30322
| | - Alton B. Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA30322
| | - Joe T. Roland
- Epithelial Biology Center, Department of Surgery, Vanderbilt University Medical Center, Nashville, TN37240
| | - Sachiko Tsukita
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo173-0003, Japan
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, GA30322
| | - Jerrold R. Turner
- Laboratory of Mucosal Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA02115
| | - Craig M. Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA30322
| |
Collapse
|
34
|
Chang B, Wang Y, Tu W, Zhang Z, Pu Y, Xie L, Yuan F, Gao Y, Xu N, Yao Q. Regulatory effects of mangiferin on LPS-induced inflammatory responses and intestinal flora imbalance during sepsis. Food Sci Nutr 2024; 12:2068-2080. [PMID: 38455195 PMCID: PMC10916552 DOI: 10.1002/fsn3.3907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/19/2023] [Accepted: 12/08/2023] [Indexed: 03/09/2024] Open
Abstract
Studies suggest that mangiferin (MAF) has good therapeutic effects on chronic bronchitis and hepatitis. Also, it is one of the antiviral ingredients in Anemarrhena asphodeloides Bunge. However, its effect on the LPS-induced inflammation and intestinal flora during sepsis remains unclear yet. In the present study, LPS-stimulated inflammation RAW264.7 cells and LPS-induced sepsis mice were used to evaluate the efficacy of MAF in vitro and in vivo. 16S rDNA sequencing was performed to analyze the characteristics of intestinal flora of the sepsis mice. It has been demonstrated that MAF (12.5 and 25 μg/mL) significantly inhibited protein expressions of TLR4, MyD88, NF-κB, and TNF-α in the LPS-treated cells and reduced the supernatant TNF-α and IL-6 levels. In vivo, MAF (20 mg/kg) markedly protected the sepsis mice and reduced the serum TNF-α and IL-6 levels. Also, MAF significantly downregulated the protein expressions of TLR4, NF-κB, and MyD88 in the livers. Importantly, MAF significantly attenuated the pathological injuries of the livers and small intestines. Further, MAF significantly increased proportion of Bacteroidota and decreased the proportions of Firmicutes, Desulfobacterota, Actinobacteriota, and Proteobacteria at phylum level, and it markedly reduced the proportions of Escherichia-Shigella, Pseudoalteromonas, Staphylococcus at genus level. Moreover, MAF affects some metabolism-related pathways such as citrate cycle (TCA cycle), lipoic acid metabolism, oxidative phosphorylation, bacterial chemotaxis, fatty acid biosynthesis, and peptidoglycan biosynthesis of the intestinal flora. Thus, it can be concluded that MAF as a treatment reduces the inflammatory responses in vitro and in vivo by inhibiting the TLR4/ MyD88/NF-κB pathway, and corrects intestinal flora imbalance during sepsis to some degree.
Collapse
Affiliation(s)
- Bo‐tao Chang
- Department of PostgraduateGuizhou University of Traditional Chinese MedicineGuiyangChina
| | - Yang Wang
- Department of General SurgeryThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Wen‐lian Tu
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Zhi‐qing Zhang
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Yan‐fang Pu
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Li Xie
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Fang Yuan
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Ying Gao
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
- The First Affiliated Hospital, Guizhou University of Traditional Chinese MedicineGuiyangChina
| | - Ning Xu
- Department of Clinical LaboratoryThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
| | - Qi Yao
- Department of PharmacyThe First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and TechnologyKunmingChina
- The First Affiliated Hospital, Guizhou University of Traditional Chinese MedicineGuiyangChina
| |
Collapse
|
35
|
Wang G, Ma F, Zhang W, Xin Y, Ping K, Wang Y, Dong J. Malvidin alleviates LPS-induced septic intestinal injury through the nuclear factor erythroid 2-related factor 2/reactive oxygen species/NLRP3 inflammasome pathway. Inflammopharmacology 2024; 32:893-901. [PMID: 38100033 DOI: 10.1007/s10787-023-01378-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 03/03/2024]
Abstract
Emerging evidence suggests that the gastrointestinal tract plays a crucial role in the pathophysiology of sepsis, a leading cause of mortality among patients admitted to the intensive care unit (ICU). Malvidin, belonging to the flavonoid family of compounds, exhibits a range of capabilities including anti-inflammatory and antioxidant properties. Studies have demonstrated that Malvidin exhibits a dose-dependent effect in mitigating sepsis-induced intestinal injury. The advantageous impact of Malvidin in safeguarding against sepsis-induced intestinal injury is associated with its capacity to counteract oxidative stress, inhibit cellular apoptosis, diminish the secretion of pro-inflammatory cytokines, and regulate the synthesis of inflammasomes. The findings indicate that Malvidin, a natural compound, exhibits protective effects on the gut by activating the nuclear factor erythroid 2-related factor 2/reactive oxygen species/NLRP3 inflammasome pathway. These results have significant implications for potential clinical applications and offer valuable insights into the treatment of sepsis-induced intestinal injury.
Collapse
Affiliation(s)
- Guanglu Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Fenfen Ma
- Department of Medicine Laboratory, Department of Cardiology, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang, China
| | - Wei Zhang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yue Xin
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Kaixin Ping
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yan Wang
- Department of Medicine Laboratory, Department of Cardiology, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| |
Collapse
|
36
|
Wang G, Ma F, Xie K, Li X, Tan X, Xia Y, Wang Y, Dong J. Liensinine alleviates mouse intestinal injury induced by sepsis through inhibition of oxidative stress, inflammation, and cell apoptosis. Int Immunopharmacol 2024; 127:111335. [PMID: 38101222 DOI: 10.1016/j.intimp.2023.111335] [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: 07/25/2023] [Revised: 11/29/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023]
Abstract
Sepsis is a clinical syndrome triggered by an imbalanced host response to pathogens that can lead to multiple organ dysfunction. The immune response and barrier function of the gut play an important role in the pathogenesis and progression of sepsis. This study aimed to explore the potential role of natural alkaloid Liensinine in the treatment of intestinal injury caused by sepsis and its possible molecular mechanism. In this study, a mouse model of sepsis was established by injecting LPS to explore the protective effect of Liensinine on intestinal injury in sepsis. The results showed that Liensinine could reduce the intestinal damage caused by LPS and increase the number of goblet cells. Furthermore, it decreased the release of inflammatory cytokines by inhibiting NF-kB phosphorylation and NLRP3 inflammasome synthesis. Liensinine also reduced the oxidative stress and ROS accumulation caused by LPS, and played an anti-oxidative stress role by regulating the Nrf2/keap1 signaling pathway. In addition, Liensinine alleviated the inhibition of intestinal autophagy caused by LPS by inhibiting the PI3K/Akt/mTOR pathway. And then it reduced the excessive apoptosis of intestinal cells. This study provides valuable insights for sepsis prevention and treatment, offering a potential therapeutic candidate to protect against intestinal injury and regulate the inflammatory response in sepsis.
Collapse
Affiliation(s)
- Guanglu Wang
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China; Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China
| | - Fenfen Ma
- Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China
| | - Kunmei Xie
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China; Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China
| | - Xueqing Li
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xuelian Tan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yan Xia
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yan Wang
- Department of Medicine Laboratory, Department of Cardiac Function Examination, The Second People's Hospital of Lianyungang Affiliated to Kangda College of Nanjing Medical University, The Second People's Hospital of Lianyungang City, Lianyungang 222000, China.
| | - Jingquan Dong
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Co-Innovation Center of Jiangsu Marine Bio-industry Technology, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China.
| |
Collapse
|
37
|
Huang FC, Huang SC. The Hazards of Probiotics on Gut-Derived Pseudomonas aeruginosa Sepsis in Mice Undergoing Chemotherapy. Biomedicines 2024; 12:253. [PMID: 38397855 PMCID: PMC10886725 DOI: 10.3390/biomedicines12020253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 02/25/2024] Open
Abstract
Pseudomonas aeruginosa (P. aeruginosa) is a leading cause of nosocomial infections associated with a high mortality rate and represents a serious threat to human health and the increasing frequency of antimicrobial resistance. Cancer patients are more vulnerable to invasive infection due to ulcerative lesions in mucosal surfaces and immune suppression secondary to chemotherapy. In our in vitro study, we observed that probiotics have the potential to yield beneficial effects on intestinal epithelial cells infected with P. aeruginosa. Additionally, probiotics were found to confer advantageous effects on the innate immunity of mice suffering from Salmonella-induced colitis. As a result, we sought to investigate the impact of probiotics on gut-derived P. aeruginosa sepsis induced by chemotherapy. Following chemotherapy, gut-derived P. aeruginosa sepsis was induced in female C57BL/6 mice aged 6-8 weeks, which were raised under specific-pathogen-free (SPF) conditions in an animal center. Prior to the induction of the sepsis model, the mice were administered 1 × 108 colony-forming units (CFU) of the probiotics, namely Lactobacillus rhamnosus GG (LGG) and Bifidobacterium longum (BL) via oral gavage. We observed that LGG or BL amplified the inflammatory mRNA expression in mice undergoing chemotherapy and suffering from gut-derived P. aeruginosa sepsis. This led to a heightened severity of colitis, as indicated by histological examination. Meanwhile, there was a notable decrease in the expression of antimicrobial peptide mRNA along with reduced levels of zonulin and claudin-2 protein staining within mucosal tissue. These alterations facilitated the translocation of bacteria to the liver, spleen, and bloodstream. To our astonishment, the introduction of probiotics exacerbated gut-derived P. aeruginosa sepsis in mice undergoing chemotherapy. Conclusively, we must be prudent when using probiotics in mice receiving chemotherapy complicated with gut-derived P. aeruginosa sepsis.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| | - Shun-Chen Huang
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
38
|
Xu H, You J, He W, Pei L, Han Y, Wang X, Tian Z, Zheng X, Wu E, Ling Y. Dynamic changes in the migratory microbial components of colon tissue during different periods of sepsis in an LPS-induced rat model. Front Cell Infect Microbiol 2024; 13:1330087. [PMID: 38287976 PMCID: PMC10822926 DOI: 10.3389/fcimb.2023.1330087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Previous studies have shown that bacterial translocation may play an important role in worsening gastrointestinal injury during sepsis. However, the dynamics of specific microbiota components in intestinal tissues at different sepsis stages remain unclear. Rats receiving intraperitoneal lipopolysaccharide (LPS) were sacrificed at 12 h and 48 h post-injection. Routine blood, serum cytokines, and microbiota in colon tissue, colonic contents, and lung tissue at different time points were assessed. Migratory microbial components in colonic tissue at 12 h and 48 h post-LPS were identified using source tracking, characteristic component identification, and abundance difference analyses. Colonic tissue microbiota changed dynamically over time after LPS injection, involving translocation of microbial components from colon contents and lung tissue at different time points. Bacteria migrating to colon tissue at 12 h sepsis were mainly from colonic contents, while those at 48 h were predominantly from the lung tissue. The migratory microbial components in colon tissue were widely associated with blood indicators and colonizing genus abundance and microbiota functionality in colon tissue. In this study, the temporal dynamics of bacterial translocation from various sources into colon tissues at different sepsis progression stages were characterized for the first time, and the species composition of these migrating microbes was delineated. These bacterial migrants may contribute to the pathophysiological processes in sepsis through direct interactions or indirectly by modulating colonic microbiota community structure and function.
Collapse
Affiliation(s)
- Hao Xu
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Jia You
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wenqin He
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Lingpeng Pei
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Yue Han
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Xueer Wang
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Zhigang Tian
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiwei Zheng
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Enqi Wu
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Yaqin Ling
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| |
Collapse
|
39
|
Liu B, Deng Y, Duan Z, Chu C, Wang X, Yang C, Li J, Ding W. Neutrophil extracellular traps promote intestinal barrier dysfunction by regulating macrophage polarization during trauma/hemorrhagic shock via the TGF-β signaling pathway. Cell Signal 2024; 113:110941. [PMID: 37890686 DOI: 10.1016/j.cellsig.2023.110941] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/01/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The mechanism by which neutrophil extracellular traps (NETs) may cause intestinal barrier dysfunction in response to trauma/hemorrhagic shock (T/HS) remains unclear. In this study, the roles and mechanisms of NETs in macrophage polarization were examined to determine whether this process plays a role in tissue damage associated with T/HS. Rat models of T/HS and macrophage polarization were developed and the levels of NETs formation in the intestinal tissue of T/HS rats were assessed. NET formation was inhibited in models of T/HS to examine the effect on intestinal inflammation and barrier injury. The proportions of pro-inflammatory and anti-inflammatory macrophages in the damaged intestinal tissues were measured. Finally, high-throughput sequencing was performed to investigate the underlying mechanisms involved in this process. The study revealed that the level of NETs formation was increased and that inhibition of NETs formation alleviated the intestinal inflammation and barrier injury. Moreover, the number of pro-inflammatory macrophages increased and the number of anti-inflammatory macrophages decreased. RNA sequencing analysis indicated that NETs formation decreased the expression of transforming growth factor-beta receptor 2 (TGFBR2), bioinformatic analyses revealed that TGFBR2 was significantly enriched in the transforming growth factor-beta (TGF-β) signaling pathway. Verification experiments showed that NETs impeded macrophage differentiation into the anti-inflammatory/M2 phenotype and inhibited TGFBR2 and TGF-β expression in macrophages. However, treatment with DNase I and overexpression of TGFBR2, and inhibition of TGF-β promoted and prevented this process, respectively. NETs may regulate the macrophage polarization process by promoting intestinal barrier dysfunction in T/HS rats through the TGFBR2-mediated TGF-β signaling pathway.
Collapse
Affiliation(s)
- Baochen Liu
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yunxuan Deng
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zehua Duan
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chengnan Chu
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xingyu Wang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chao Yang
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jieshou Li
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Weiwei Ding
- Department of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
40
|
Abstract
The remarkable diversity of lymphocytes, essential components of the immune system, serves as an ingenious mechanism for maximizing the efficient utilization of limited host defense resources. While cell adhesion molecules, notably in gut-tropic T cells, play a central role in this mechanism, the counterbalancing molecular details have remained elusive. Conversely, we've uncovered the molecular pathways enabling extracellular vesicles secreted by lymphocytes to reach the gut's mucosal tissues, facilitating immunological regulation. This discovery sheds light on immune fine-tuning, offering insights into immune regulation mechanisms.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Higashimurayama, Tokyo, Japan
| |
Collapse
|
41
|
Ren Z, Zheng Z, Feng X. Role of gut microbes in acute lung injury/acute respiratory distress syndrome. Gut Microbes 2024; 16:2440125. [PMID: 39658851 PMCID: PMC11639474 DOI: 10.1080/19490976.2024.2440125] [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: 07/04/2024] [Revised: 10/31/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024] Open
Abstract
Acute lung injury (ALI) is an acute, diffuse inflammatory lung condition triggered by factors of severe infections, trauma, shock, burns, ischemia-reperfusion, and mechanical ventilation. It is primarily characterized by refractory hypoxemia and respiratory distress. The more severe form, acute respiratory distress syndrome (ARDS), can progress to multi-organ failure and has a high mortality rate. Despite extensive research, the exact pathogenesis of ALI and ARDS remains complex and not fully understood. Recent advancements in studying the gut microecology of patients have revealed the critical role that gut microbes play in ALI/ARDS onset and progression. While the exact mechanisms are still under investigation, evidence increasingly points to the influence of gut microbes and their metabolites on ALI/ARDS. This review aims to summarize the role of gut microbes and their metabolites in ALI/ARDS caused by various triggers. Moreover, it explores potential mechanisms and discusses how gut microbe-targeting interventions might offer new clinical strategies for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Zixuan Ren
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhihuan Zheng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiujing Feng
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
42
|
Cao X, Zhao H, Liang Z, Cao Y, Min M. Long-term administration of probiotics prevents gastrointestinal mucosal barrier dysfunction in septic mice partly by upregulating the 5-HT degradation pathway. Open Med (Wars) 2023; 18:20230869. [PMID: 38152336 PMCID: PMC10751891 DOI: 10.1515/med-2023-0869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/22/2023] [Accepted: 11/12/2023] [Indexed: 12/29/2023] Open
Abstract
Sepsis can impair gastrointestinal (GI) barrier integrity. Oral probiotics (PT) can maintain the balance of GI microflora and improve GI function. 5-Hydroxytryptamine (5-HT) is a key promoter of GI injury caused by sepsis. However, the mechanism by which PT attenuates sepsis by regulating 5-HT is not fully understood. In this study, C57BL6 mice were intragastric administrated with normal saline (NC) or PT once a day for 4 weeks before cecal ligation and puncture (CLP). Compared with NC-CLP mice, PT-CLP mice had lower clinical score, higher body temperature. The survival rate of PT-CLP mice was significantly improved. The levels of inflammatory cytokines and 5-HT were obviously decreased in PT-CLP mice, and GI peristalsis and barrier function were enhanced. Moreover, sepsis downregulated the expression of tight junction proteins, while PT pretreatment could maintain them at the level of sham operation group. Furthermore, PT pretreatment increased the expression of serotonin transporter and monoamine oxidase A. PT administration could inhibit NF-κB activity, and activate ERK activity. In conclusion, long-term supplementation of PT before CLP can prevent sepsis-induced GI mucosal barrier dysfunction in mice, which may be partially mediated by upregulating the 5-HT degradation pathway via activating ERK signaling.
Collapse
Affiliation(s)
- Xiaopeng Cao
- Department of Gastroenterology, The First Medical Center of PLA General Hospital, Beijing, 100048China
| | - Hui Zhao
- Department of Gastroenterology, The First Medical Center of PLA General Hospital, Beijing, 100048China
| | - Zhimin Liang
- Department of Gastroenterology, The First Medical Center of PLA General Hospital, Beijing, 100048China
| | - Yi Cao
- Department of Global Health, Milken Institute School of Public Health, The George Washington University, WashingtonDC, 20052USA
| | - Min Min
- Department of Gastroenterology, The Fifth Medical Center of PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100039China
| |
Collapse
|
43
|
Lauwers C, De Bruyn L, Langouche L. Impact of critical illness on cholesterol and fatty acids: insights into pathophysiology and therapeutic targets. Intensive Care Med Exp 2023; 11:84. [PMID: 38015312 PMCID: PMC10684846 DOI: 10.1186/s40635-023-00570-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/21/2023] [Indexed: 11/29/2023] Open
Abstract
Critical illness is characterized by a hypercatabolic response encompassing endocrine and metabolic alterations. Not only the uptake, synthesis and metabolism of glucose and amino acids is majorly affected, but also the homeostasis of lipids and cholesterol is altered during acute and prolonged critical illness. Patients who suffer from critically ill conditions such as sepsis, major trauma, surgery or burn wounds display an immediate and sustained reduction in low plasma LDL-, HDL- and total cholesterol concentrations, together with a, less pronounced, increase in plasma free fatty acids. The severity of these alterations is associated with severity of illness, but the underlying pathophysiological mechanisms are multifactorial and only partly clarified. This narrative review aims to provide an overview of the current knowledge of how lipid and cholesterol uptake, synthesis and metabolism is affected during critical illness. Reduced nutritional uptake, increased scavenging of lipoproteins as well as an increased conversion to cortisol or other cholesterol-derived metabolites might all play a role in the decrease in plasma cholesterol. The acute stress response to critical illness creates a lipolytic cocktail, which might explain the increase in plasma free fatty acids, although reduced uptake and oxidation, but also increased lipogenesis, especially in prolonged critical illness, will also affect the circulating levels. Whether a disturbed lipid homeostasis warrants intervention or should primarily be interpreted as a signal of severity of illness requires further research.
Collapse
Affiliation(s)
- Caroline Lauwers
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lauren De Bruyn
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium
| | - Lies Langouche
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, O&N1 Box 503, 3000, Leuven, Belgium.
| |
Collapse
|
44
|
Liu J, Chen B, Jiang M, Cui T, Lv B, Fu Z, Li X, Du Y, Guo J, Zhong X, Zou Y, Zhao X, Yang W, Gao X. Polygonatum odoratum polysaccharide attenuates lipopolysaccharide-induced lung injury in mice by regulating gut microbiota. Food Sci Nutr 2023; 11:6974-6986. [PMID: 37970373 PMCID: PMC10630852 DOI: 10.1002/fsn3.3622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 11/17/2023] Open
Abstract
Polygonatum odoratum is appreciated for its edible and medicinal benefits especially for lung protection. However, the contained active components have been understudied, and further research is required to fully exploit its potential application. We aimed to probe into the beneficial effects of Polygonatum odoratum polysaccharide (POP) in lipopolysaccharide-induced lung inflammatory injury mice. POP treatment could ameliorate the survival rate, pulmonary function, lung pathological lesions, and immune inflammatory response. POP treatment could repair intestinal barrier, and modulate the composition of gut microbiota, especially reducing the abundance of Klebsiella, which were closely associated with the therapeutic effects of POP. Investigation of the underlying anti-inflammatory mechanism showed that POP suppressed the generation of pro-inflammatory molecules in lung by inhibiting iNOS+ M1 macrophages. Collectively, POP is a promising multi-target microecological regulator to prevent and treat the immuno-inflammation and lung injury by modulating gut microbiota.
Collapse
Affiliation(s)
- Jia‐rui Liu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Bo‐xue Chen
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Mei‐ting Jiang
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Tian‐yi Cui
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Bin Lv
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Zhi‐fei Fu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xue Li
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yao‐dong Du
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Jin‐he Guo
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xin‐qin Zhong
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ya‐dan Zou
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xin Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Wen‐zhi Yang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xiu‐mei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| |
Collapse
|
45
|
Kobritz M, Nofi C, Sfakianos M, Coppa G, Aziz M, Wang P. Targeting sting to reduce sepsis-induced acute intestinal injury. Surgery 2023; 174:1071-1077. [PMID: 37517896 PMCID: PMC10529857 DOI: 10.1016/j.surg.2023.06.032] [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: 03/28/2023] [Revised: 06/08/2023] [Accepted: 06/18/2023] [Indexed: 08/01/2023]
Abstract
BACKGROUND Sepsis is a dysregulated host response to infection syndrome leading to life-threatening organ dysfunction. Sepsis-induced intestinal dysfunction is a key element in the progression to multisystem organ failure. The stimulator of interferon genes is an intracellular protein implicated in intestinal injury in sepsis. H151, a small molecule inhibitor of stimulator of interferon genes, has not yet been studied as a potential therapeutic in sepsis. We hypothesize that H151 therapeutically reduces sepsis-induced acute intestinal injury. METHODS Male mice underwent cecal ligation and puncture and were treated with intraperitoneal H151 (10 mg/kg body weight) or vehicle. Intestines and serum were collected for analysis 20 hours after cecal ligation and puncture. Oral gavage of mice with FITC-dextran was performed 15 hours after cecal ligation and puncture. Five hours after gavage, serum was collected, and intestinal permeability was assessed. Mice were monitored for 10 days after cecal ligation and puncture to assess survival. RESULTS Zonula occludens 1 tight junctional protein expression was reduced after cecal ligation and puncture and recovered with H151 treatment. This was associated with a 62.3% reduction in intestinal permeability as assessed by fluorimetry. After cecal ligation and puncture, treatment with H151 was associated with a 58.7% reduction in intestinal histopathologic injury (P < .05) and a 56.6% reduction in intestinal apoptosis (P < .05). Intestinal myeloperoxidase activity was decreased by 70.8% after H151 treatment (P < .05). Finally, H151 improved 10-day survival from 33% to 80% after cecal ligation and puncture (P = .011). CONCLUSION H151, a novel stimulator of interferon genes inhibitor, reduces intestinal injury, inflammation, and permeability when administered as a treatment for cecal ligation and puncture-induced sepsis. Thus, targeting stimulator of interferon genes shows promise as a therapeutic strategy to ameliorate sepsis-induced acute intestinal injury.
Collapse
Affiliation(s)
- Molly Kobritz
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Colleen Nofi
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Maria Sfakianos
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Gene Coppa
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
| | - Monowar Aziz
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| |
Collapse
|
46
|
Chen TT, Lv JJ, Chen L, Li M, Liu LP. Heparanase inhibition leads to improvement in patients with acute gastrointestinal injuries induced by sepsis. World J Gastroenterol 2023; 29:5154-5165. [PMID: 37744293 PMCID: PMC10514756 DOI: 10.3748/wjg.v29.i35.5154] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Patients with sepsis are at high risk for acute gastrointestinal injury (AGI), but the diagnosis and treatment of AGI due to sepsis are unsatisfactory. Heparanase (HPA) plays an important role in septic AGI (S-AGI), but its specific mechanism is not completely understood, and few clinical reports are available. AIM To explore the effect and mechanism of HPA inhibition in S-AGI patients. METHODS In our prospective clinical trial, 48 patients with S-AGI were randomly assigned to a control group to receive conventional treatment, whereas 47 patients were randomly assigned to an intervention group to receive conventional treatment combined with low molecular weight heparin. AGI grade, sequential organ failure assessment score, acute physiology and chronic health evaluation II score, D-dimer, activated partial thromboplastin time (APTT), anti-Xa factor, interleukin-6, tumour necrosis factor-α, HPA, syndecan-1 (SDC-1), LC3B (autophagy marker), intestinal fatty acid binding protein, D-lactate, motilin, gastrin, CD4/CD8, length of intensive care unit (ICU) stay, length of hospital stay and 28-d survival on the 1st, 3rd and 7th d after treatment were compared. Correlations between HPA and AGI grading as well as LC3B were compared. Receiver operator characteristic (ROC) curves were generated to evaluate the diagnostic value of HPA, intestinal fatty acid binding protein and D-lactate in S-AGI. RESULTS Serum HPA and SCD-1 levels were significantly reduced in the intervention group compared with the control group (P < 0.05). In addition, intestinal fatty acid-binding protein, D-lactate, AGI grade, motilin, and gastrin levels and sequential organ failure assessment score were significantly decreased (P < 0.05) in the intervention group. However, LC3B, APTT, anti-Xa factor, and CD4/CD8 were significantly increased (P < 0.05) in the intervention group. No significant differences in interleukin-6, tumour necrosis factor-α, d-dimer, acute physiology and chronic health evaluation II score, length of ICU stay, length of hospital stay, or 28-d survival were noted between the two groups (P > 0.05). Correlation analysis revealed a significant negative correlation between HPA and LC3B and a significant positive correlation between HPA and AGI grade. ROC curve analysis showed that HPA had higher specificity and sensitivity in diagnosis of S-AGI. CONCLUSION HPA has great potential as a diagnostic marker for S-AGI. Inhibition of HPA activity reduces SDC-1 shedding and alleviates S-AGI symptoms. The inhibitory effect of HPA in gastrointestinal protection may be achieved by enhanced autophagy.
Collapse
Affiliation(s)
- Ting-Ting Chen
- The First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Jia-Jun Lv
- The First Clinical Medical School of Lanzhou University, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ling Chen
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Min Li
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Li-Ping Liu
- Department of Emergency Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
47
|
Ding N, Xiao H, Zhen L, Li H, Zhang Z, Ge J, Jia H. Systemic cytokines inhibition with Imp7 siRNA nanoparticle ameliorates gut injury in a mouse model of ventilator-induced lung injury. Biomed Pharmacother 2023; 165:115237. [PMID: 37516020 DOI: 10.1016/j.biopha.2023.115237] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/22/2023] [Accepted: 07/25/2023] [Indexed: 07/31/2023] Open
Abstract
Mechanical ventilation (MV) may negatively affect the lungs and cause the release of inflammatory mediators, resulting in extra-pulmonary organ dysfunction. Studies have revealed systemically elevated levels of proinflammatory cytokines in animal models of ventilator-induced lung injury (VILI); however, whether these cytokines have an effect on gut injury and the mechanisms involved remain unknown. In this study, VILI was generated in mice with high tidal volume mechanical ventilation (20 ml/kg). Tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6 concentrations in serum and gut measured by ELISA showed significant elevation in the VILI mice. Significant increases in gut injury and PANoptosis were observed in the VILI mice, which were positively correlated with the serum levels of TNF-α, IL-1β, and IL-6. The VILI mice displayed intestinal barrier defects, decreased expressions of occludin and zonula occludin-1 (ZO-1), and increased expression of claudin-2 and the activation of myosin light chain (MLC). Importantly, intratracheal administration of Imp7 siRNA nanoparticle effectively inhibited cytokines production and protected mice from VILI-induced gut injury. These data provide evidence of systemic cytokines contributing to gut injury following VILI and highlight the possibility of targeting cytokines inhibition via Imp7 siRNA nanoparticle as a potential therapeutic intervention for alleviating gut injury following VILI.
Collapse
Affiliation(s)
- Ning Ding
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China.
| | - Hui Xiao
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Lixiao Zhen
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Huiqing Li
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Zengzhen Zhang
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Anesthesiology, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China
| | - Junke Ge
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiyan Jia
- Key Laboratory of Intensive Care Rehabilitation of Shandong, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250031, China; Department of Intensive Care Medicine, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
48
|
Sun Y, Song J, Lan X, Ma F, Jiang M, Jiang C. Calcium-Sensitive Receptors Alters Intestinal Microbiota Metabolites Especially SCFAs and Ameliorates Intestinal Barrier Damage in Neonatal Rat Endotoxemia. Infect Drug Resist 2023; 16:5707-5717. [PMID: 37667808 PMCID: PMC10475303 DOI: 10.2147/idr.s420689] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023] Open
Abstract
Purpose The calcium-sensing receptor (CaSR) acts as a major modulator of tissue responses related to calcium homeostasis and expresses highly in the mammalian intestine. Endotoxemia tends to impair intestinal barrier function and poses significant obstacles in clinical treatment. This work is designed to decipher whether CaSR can protect lipopolysaccharide (LPS)-induced intestinal barrier dysfunction in neonatal rats by targeting intestinal metabolites. Patient and Methods In this study, we utilized gas chromatography (GC) combined with liquid chromatography-mass spectrometry (LC-MS) to quantitatively analyze SCFAs and metabolites in fecal samples of 24 neonatal rats with LPS induced endotoxemia. Results Our results showed that CaSR alleviated endotoxin damage to the intestinal tight junction structure and upregulated the levels of butyric acid, propionic acid, valeric acid, and isovaleric acid in short-chain fatty acids (SCFAs). Non-targeted metabolomics analysis indicated that CaSR improved intestinal metabolic disorders by regulating glycerophospholipid metabolism, α-linolenic acid metabolism, as well as sphingolipids metabolism. Conclusion CaSR can alter intestinal microbiota metabolites, especially SCFAs, and improve intestinal barrier damage in neonatal rat endotoxemia.
Collapse
Affiliation(s)
- Yan Sun
- Department of Neonatology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Jiayu Song
- Department of Neonatology, Zhuhai Women and Children’s Hospital, Zhuhai, Guangdong, 519060, People’s Republic of China
| | - Xue Lan
- Department of Neonatology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Fei Ma
- Department of Neonatology, Zhuhai Women and Children’s Hospital, Zhuhai, Guangdong, 519060, People’s Republic of China
| | - Mingyu Jiang
- Department of Pediatrics, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
| | - Chunming Jiang
- Department of Neonatology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People’s Republic of China
- Department of Neonatology, Zhuhai Women and Children’s Hospital, Zhuhai, Guangdong, 519060, People’s Republic of China
| |
Collapse
|
49
|
Abenavoli L, Scarpellini E, Paravati MR, Scarlata GGM, Boccuto L, Tilocca B, Roncada P, Luzza F. Gut Microbiota and Critically Ill Patients: Immunity and Its Modulation via Probiotics and Immunonutrition. Nutrients 2023; 15:3569. [PMID: 37630759 PMCID: PMC10459644 DOI: 10.3390/nu15163569] [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: 06/30/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Critically ill patients have a hyper-inflammatory response against various offending injuries that can result in tissue damage, organ failure, and fatal prognosis. The origin of this detrimental, uncontrolled inflammatory cascade can be found also within our gut. In detail, one of the main actors is our gut microbiota with its imbalance, namely gut dysbiosis: learning about the microbiota's dysfunction and pathophysiology in the frame of critical patients is of crucial and emerging importance in the management of the systemic inflammatory response syndrome (SIRS) and the multiple organ dysfunction syndrome (MODS). Multiple pieces of evidence indicate that the bacteria that populate our gut efficiently modulate the immune response. Treatment and pretreatment with probiotics have shown promising preliminary results to attenuate systemic inflammation, especially in postoperative infections and ventilation performance. Finally, it is emerging how immunonutrition may exert a possible impact on the health status of patients in intensive care. Thus, this manuscript reviews evidence from the literature on gut microbiota composition, its derangement in critically ill patients, its pathophysiological role, and the described and emerging opportunities arising from its modulation.
Collapse
Affiliation(s)
- Ludovico Abenavoli
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (M.R.P.); (G.G.M.S.); (B.T.); (P.R.); (F.L.)
| | - Emidio Scarpellini
- Translationeel Onderzoek van Gastro-Enterologische Aandoeningen (T.A.R.G.I.D.), Gasthuisberg University 11 Hospital, KU Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Maria Rosaria Paravati
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (M.R.P.); (G.G.M.S.); (B.T.); (P.R.); (F.L.)
| | - Giuseppe Guido Maria Scarlata
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (M.R.P.); (G.G.M.S.); (B.T.); (P.R.); (F.L.)
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC 29634, USA;
- School of Health Research, Clemson University, Clemson, SC 29634, USA
| | - Bruno Tilocca
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (M.R.P.); (G.G.M.S.); (B.T.); (P.R.); (F.L.)
| | - Paola Roncada
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (M.R.P.); (G.G.M.S.); (B.T.); (P.R.); (F.L.)
| | - Francesco Luzza
- Department of Health Sciences, University “Magna Graecia”, 88100 Catanzaro, Italy; (M.R.P.); (G.G.M.S.); (B.T.); (P.R.); (F.L.)
| |
Collapse
|
50
|
Oami T, Yumoto T, Shimazui T, Sarmiento S, Klingensmith NJ, Chen CW, Otani S, Liang Z, Burd EM, Mahdi ZK, Ford ML, Coopersmith CM. CHRONIC ETHANOL USE WORSENS GUT PERMEABILITY AND ALTERS TIGHT JUNCTION EXPRESSION IN A MURINE SEPSIS MODEL. Shock 2023; 60:280-290. [PMID: 37405872 PMCID: PMC10526701 DOI: 10.1097/shk.0000000000002162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
ABSTRACT Alcohol use disorder is associated with increased mortality in septic patients. Murine studies demonstrate that ethanol/sepsis is associated with changes in gut integrity. This study examined intestinal permeability after ethanol/sepsis and investigated mechanisms responsible for alterations in barrier function. Mice were randomized to drink either 20% ethanol or water for 12 weeks and then were subjected to either sham laparotomy or cecal ligation and puncture (CLP). Intestinal permeability was disproportionately increased in ethanol/septic mice via the pore, leak, and unrestricted pathways. Consistent with increased permeability in the leak pathway, jejunal myosin light chain (MLC) kinase (MLCK) expression and the ratio of phospho-MLC to total MLC were both increased in ethanol/CLP. Gut permeability was altered in MLCK -/- mice in water/CLP; however, permeability was not different between WT and MLCK -/- mice in ethanol/CLP. Similarly, jejunal IL-1β levels were decreased while systemic IL-6 levels were increased in MLCK -/- mice in water/CLP but no differences were identified in ethanol/CLP. While we have previously shown that mortality is improved in MLCK -/- mice after water/CLP, mortality was significantly worse in MLCK -/- mice after ethanol/CLP. Consistent with an increase in the pore pathway, claudin 4 levels were also selectively decreased in ethanol/CLP WT mice. Furthermore, mRNA expression of jejunal TNF and IFN-γ were both significantly increased in ethanol/CLP. The frequency of CD4 + cells expressing TNF and IL-17A and the frequency of CD8 + cells expressing IFN-γ in Peyer's Patches were also increased in ethanol/CLP. Thus, there is an ethanol-specific worsening of gut barrier function after CLP that impacts all pathways of intestinal permeability, mediated, in part, via changes to the tight junction. Differences in the host response in the setting of chronic alcohol use may play a role in future precision medicine approaches toward the treatment of sepsis.
Collapse
Affiliation(s)
| | | | | | - Sofia Sarmiento
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia
| | - Nathan J Klingensmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia
| | - Ching-Wen Chen
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia
| | | | - Zhe Liang
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia
| | - Eileen M Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Zaid K Mahdi
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Mandy L Ford
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia
| | - Craig M Coopersmith
- Department of Surgery and Emory Critical Care Center, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|