1
|
Zheng Q, Xu D, Wang Q, Liu L, Liu W, Wang J. Huang-Lian-Jie-Du decoction ameliorates sepsis through dynamic regulation of immune response and gut microbiota-metabolite axis. Microb Pathog 2025; 199:107246. [PMID: 39708977 DOI: 10.1016/j.micpath.2024.107246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
Sepsis remains a life-threatening condition with high mortality rates despite current therapeutic approaches. While Huang-Lian-Jie-Du Decoction (HLJDD), a traditional Chinese medicine formula, has been historically used to treat inflammatory conditions, its therapeutic potential in sepsis and underlying mechanisms remain unexplored. This study investigated HLJDD's comprehensive effects on sepsis pathophysiology using a rat cecal ligation and puncture (CLP) model. HLJDD significantly improved survival rates and demonstrated sophisticated immunomodulatory effects through temporal regulation of the biphasic immune response characteristic of sepsis. In early sepsis, HLJDD suppressed pro-inflammatory cytokines (IL-1β, IL-6) while maintaining defensive inflammation. During late sepsis, it counteracted immunosuppression by reducing IL-10 levels and CD4+CD25+ T cell populations while protecting CD4+ and CD8+ T cells from apoptosis. Notably, HLJDD demonstrated dynamic regulation of the gut microbiota-metabolite axis. It enhanced beneficial bacterial populations (Firmicutes, Lactobacillus) while suppressing potentially pathogenic species (Bacteroides, Parabacteroides). Metabolomic analysis revealed time-dependent modulation of short-chain fatty acids, with elevated levels at 12 h followed by strategic reduction at 18-30 h, coordinating with changes in SCFA-producing bacteria. This temporal metabolic regulation corresponded with improved intestinal barrier function and balanced immune responses. The study unveils HLJDD's novel mechanism of action through synchronized modulation of immune responses, gut microbiota, and metabolite profiles, presenting a multi-target therapeutic approach that addresses the complex pathophysiology of sepsis. These findings provide a strong foundation for further clinical investigation of HLJDD as an innovative treatment strategy for sepsis.
Collapse
Affiliation(s)
- Qi Zheng
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Di Xu
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Qing Wang
- Department of Interventional Surgery, Zibo Central Hospital, 54 Gongqingtuan Road, Zibo, Shandong, China
| | - Lin Liu
- Department of Interventional Surgery, Zibo Central Hospital, 54 Gongqingtuan Road, Zibo, Shandong, China
| | - Wenya Liu
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China
| | - Junsong Wang
- Center of Molecular Metabolism, Nanjing University of Science and Technology, 200 Xiaolingwei Street, Nanjing, 210094, China.
| |
Collapse
|
2
|
Rajput I, Rajendran VM, Nickerson AJ, Lodge JPA, Sandle GI. Somatostatin peptides prevent increased human colonic epithelial permeability induced by hypoxia. Am J Physiol Gastrointest Liver Physiol 2024; 327:G701-G710. [PMID: 39226584 PMCID: PMC11559641 DOI: 10.1152/ajpgi.00057.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
Mesenteric ischemia increases gut permeability and bacterial translocation. In human colon, chemical hypoxia induced by 2,4-dinitrophenol (DNP) activates basolateral intermediate conductance K+ (IK) channels (designated KCa3.1 or KCNN4) and increases paracellular shunt conductance/permeability (GS), but whether this leads to increased macromolecule permeability is unclear. Somatostatin (SOM) inhibits IK channels and prevents hypoxia-induced increases in GS. Thus, we examined whether octreotide (OCT), a synthetic SOM analog, prevents hypoxia-induced increases GS in human colon and hypoxia-induced increases in total epithelial conductance (GT) and permeability to FITC-dextran 4000 (FITC) in rat colon. The effects of serosal SOM and OCT on increases in GS induced by 100 µM DNP were compared in isolated human colon. The effects of OCT on DNP-induced increases in GT and transepithelial FITC movement were evaluated in isolated rat distal colon. GS in DNP-treated human colon was 52% greater than in controls (P = 0.003). GS was similar when 2 µM SOM was added after or before DNP treatment, in both cases being less (P < 0.05) than with DNP alone. OCT (0.2 µM) was equally effective preventing hypoxia-induced increases in GS, whether added after or before DNP treatment. In rat distal colon, DNP significantly increased GT by 18% (P = 0.016) and mucosa-to-serosa FITC movement by 43% (P = 0.01), and 0.2 µM OCT pretreatment completely prevented these changes. We conclude that OCT prevents hypoxia-induced increases in paracellular/macromolecule permeability and speculate that it may limit ischemia-induced gut hyperpermeability during abdominal surgery, thereby reducing bacterial/bacterial toxin translocation and sepsis.NEW & NOTEWORTHY Somatostatin (SOM, 2 µM) and octreotide (OCT, 0.2 µM, a long-acting synthetic analog of SOM) were equally effective in preventing chemical hypoxia-induced increases in paracellular shunt permeability/conductance in isolated human colon. In rat distal colon, chemical hypoxia significantly increased total epithelial conductance and transepithelial movement of FITC-dextran 4000, changes completely prevented by 0.2 µM OCT. OCT may prevent or limit gut ischemia during abdominal surgery, thereby decreasing the risk of bacterial/bacterial toxin translocation and sepsis.
Collapse
Affiliation(s)
- Ibrahim Rajput
- Department of Surgery, St James's University Hospital, Leeds, United Kingdom
| | - Vazhaikkurichi M Rajendran
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Andrew J Nickerson
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - J Peter A Lodge
- Department of Surgery, St James's University Hospital, Leeds, United Kingdom
| | - Geoffrey I Sandle
- Leeds Institute of Medical Research at St James's, St James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
3
|
Ma Y, Zhao Y, Zhang X. Factors affecting neutrophil functions during sepsis: human microbiome and epigenetics. J Leukoc Biol 2024; 116:672-688. [PMID: 38734968 DOI: 10.1093/jleuko/qiae107] [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: 12/07/2023] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/13/2024] Open
Abstract
Sepsis is a severe disease that occurs when the body's immune system reacts excessively to infection. The body's response, which includes an intense antibacterial reaction, can damage its tissues and organs. Neutrophils are the major components of white blood cells in circulation, play a vital role in innate immunity while fighting against infections, and are considered a feature determining sepsis classification. There is a plethora of basic research detailing neutrophil functioning, among which, the study of neutrophil extracellular traps is providing novel insights into mechanisms and treatments of sepsis. This review explores their functions, dysfunctions, and influences in the context of sepsis. The interplay between neutrophils and the human microbiome and the impact of DNA methylation on neutrophil function in sepsis are crucial areas of study. The interaction between neutrophils and the human microbiome is complex, particularly in the context of sepsis, where dysbiosis may occur. We highlight the importance of deciphering neutrophils' functional alterations and their epigenetic features in sepsis because it is critical for defining sepsis endotypes and opening up the possibility for novel diagnostic methods and therapy. Specifically, epigenetic signatures are pivotal since they will provide a novel implication for a sepsis diagnostic method when used in combination with the cell-free DNA. Research is exploring how specific patterns of DNA methylation in neutrophils, detectable in cell-free DNA, could serve as biomarkers for the early detection of sepsis.
Collapse
Affiliation(s)
- Yina Ma
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Yu Zhao
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| | - Xin Zhang
- Department of Urology Surgery, Beijing Chaoyang Hospital, Capital Medical University, Shijingshan District, Beijing 100043, China
| |
Collapse
|
4
|
Radler JB, McBride AR, Saha K, Nighot P, Holmes GM. Regional Heterogeneity in Intestinal Epithelial Barrier Permeability and Mesenteric Perfusion After Thoracic Spinal Cord Injury. Dig Dis Sci 2024; 69:3236-3248. [PMID: 39001959 DOI: 10.1007/s10620-024-08537-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/18/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Spinal cord injury (SCI) disrupts intestinal barrier function, thereby increasing antigen permeation and leading to poor outcomes. Despite the intestinal tract's anatomic and physiologic heterogeneity, studies following SCI have not comprehensively addressed intestinal pathophysiology with regional specificity. AIMS AND METHODS We used an experimental model of high thoracic SCI to investigate (1) regional mucosal oxidative stress using dihydroethidium labeling; (2) regional paracellular permeability to small- and large-molecular probes via Ussing chamber; (3) regional intestinal tight junction (TJ) protein expression; and (4) hindgut perfusion via the caudal mesenteric artery. RESULTS Dihydroethidium staining was significantly elevated within duodenal mucosa at 3-day post-SCI. Molar flux of [14C]-urea was significantly elevated in duodenum and proximal colon at 3-day post-SCI, while molar flux of [3H]-inulin was significantly elevated only in duodenum at 3-day post-SCI. Barrier permeability was mirrored by a significant increase in the expression of pore-forming TJ protein claudin-2 in duodenum and proximal colon at 3-day post-SCI. Claudin-2 expression remained significantly elevated in proximal colon at 3-week post-SCI. Expression of the barrier-forming TJ protein occludin was significantly reduced in duodenum at 3-day post-SCI. Caudal mesenteric artery flow was unchanged by SCI at 3 days or 3 weeks despite significant reductions in mean arterial pressure. CONCLUSION These data show that T3-SCI provokes elevated mucosal oxidative stress, altered expression of TJ proteins, and elevated intestinal barrier permeability in the proximal intestine. In contrast, mucosal oxidative stress and intestinal barrier permeability were unchanged in the hindgut after SCI. This regional heterogeneity may result from differential sensitivity to reduced mesenteric perfusion, though further studies are required to establish a causal link. Understanding regional differences in intestinal pathophysiology is essential for developing effective treatments and standards of care for individuals with SCI.
Collapse
Affiliation(s)
- Jackson B Radler
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA
| | - Amanda R McBride
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA
- Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
| | - Kushal Saha
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State University College of Medicine, Hershey, PA, 17033, USA
| | - Gregory M Holmes
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Dr., H109, Hershey, PA, 17033, USA.
| |
Collapse
|
5
|
Hayashi S, Moriyama T, Ito Y, Harada Y, Dodo H, Kumahara K, Yogi T, Ohashi N, Higashi R, Mori A. Proton-Pump Inhibitors and Risk of Bloodstream Infection without an Identifiable Source: a Hospital-Based Case-Control Study. Jpn J Infect Dis 2024; 77:205-212. [PMID: 38296545 DOI: 10.7883/yoken.jjid.2023.253] [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: 07/26/2024]
Abstract
The association between proton-pump inhibitor (PPI) use and systemic infections caused by bacterial translocation is unclear. This study aimed to investigate whether patients receiving PPI therapy have a higher risk of bloodstream infections (BSI) without an identifiable source of infection. We conducted a hospital-based case-control study which enrolled all patients aged 20 years and older who were hospitalized in Ichinomiya Nishi Hospital with BSI confirmed by two sets of positive blood cultures in 2019. Patient data were collected from medical records, and the bacterial translocation-type (BT-type) BSI group was defined as patients with BSI without an identifiable source of infection, whereas those with a BSI from an identifiable source were assigned to the control group based on the diagnostic criteria for each infectious disease. Data from 309 patients, including 66 cases and 243 controls, were analyzed. Compared with PPI non-users, PPI users had a 2.4-fold higher risk of developing BT-type BSI after controlling for potential confounders (adjusted odds ratio: 2.41, 95% confidence interval: 1.29-4.51, P = 0.006). In conclusion, PPI use is associated with a higher risk of BSI without an identifiable source; therefore, PPI use might increase the risk of BSI secondary to bacterial translocation.
Collapse
Affiliation(s)
- Shintaro Hayashi
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | | | - Yuichiro Ito
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Yuta Harada
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Hiroki Dodo
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Kana Kumahara
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Tatsuji Yogi
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Noritsugu Ohashi
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Reiji Higashi
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| | - Akihiro Mori
- Department of Gastroenterology, Ichinomiya Nishi Hospital, Japan
| |
Collapse
|
6
|
Chen H, Liu H, Sun Y, Su M, Lin J, Wang J, Lin J, Zhao X. Analysis of fecal microbiota and related clinical indicators in ICU patients with sepsis. Heliyon 2024; 10:e28480. [PMID: 38586361 PMCID: PMC10998127 DOI: 10.1016/j.heliyon.2024.e28480] [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: 12/04/2022] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 04/09/2024] Open
Abstract
Background To analyze the characteristics of fecal microbiota disturbance in the intensive care unit (ICU) patients with sepsis and the correlation with related clinical indicators. Methods This study included 31 patients with sepsis admitted to the emergency ICU ward between September 2019 and December 2021. They were divided into Group without septic shock (ND_NS group, 7 cases) and Group with septic shock (ND_S group, 24 cases) according to the presence or absence of septic shock. Furthermore, we divided these 31 sepsis patients into Clinical Improvement group (21 cases) and Death or DAMA group (10 cases) based on clinical outcome, 15 cases of Physical Examiner recruited in the same period were included as control group: ND_HC group (15 cases). The fecal samples of the patients with sepsis within 24 h of admission and random fecal samples of the control group were collected and analyzed by 16S rDNA gene sequencing used for the analysis of fecal microbiota. At the same time, the relevant clinical data of these patients with sepsis were also collected for analysis. Results There were 15 cases with drug-resistant bacteria in the ND_S group and only 2 cases in the ND_NS group (P = 0.015). There were significant differences in APACHE II score, length of ICU stay, lactate level, and oxygenation index of patients between the Death or DAMA group and Clinical Improvement group (all P < 0.05). For phylum level, the abundance of Firmicutes, Actinobacteria, and Bacteroidetes decreased in the ND group compared with the ND_HC group, while the abundance of Proteobacteria increased (P < 0.05). For genus level, the relative abundance of Escherichia-Shigella and Klebsiella were significantly increased in the ND group compared with the ND_HC group (P < 0.05). The top six genera in relative abundance in the ND_S group were Escherichia-Shigella, Enterococcus, Bifidobacterium, Lactobacillus, Akkermansia, and Klebsiella. Compared with the Clinical Improvement group, the relative abundance of Escherichia-Shigella and Klebsiella in the Death or DAMA group showed an increasing trend with no significant significance, while the relative abundance of Enterococcus and Faecalibacterium decreased in the Death or DAMA group (P < 0.05). Alpha diversity analysis showed that compared with the ND_HC group, the alpha diversity of the fecal microbiota in the ND group decreased. There were significant differences in the Observed_species index, Chao1 index, and ACE index of patients between the ND_HC group and ND group (all P < 0.05). Moreover, compared with the ND_NS group, the Alpha diversity of the ND_S group was more abundant. PCoA analysis showed significant differences in microbial community structure between the ND group and ND_HC group (P = 0.001). There also were significant differences in microbial community structure between the ND_S group and ND_NS group (P = 0.008). LEfSe analysis showed that compared with the ND_HC group, there were significant differences in the species of the ND group, including Enterobacteriaceae, Escherichia-Shigella, Enterococcus, Elizabethkingia, and Family_XIII_AD3011_group. Conclusions ICU patients with sepsis suffered intestinal microecological disturbances with significantly decreased abundance of fecal microbiota, diversity, and beneficial symbiotic bacteria. For these patients, the ratio of pathogenic bacteria, including Escherichia-Shigella and Klebsiella increased and became the main bacterial genus in some samples. Moreover, the increasing trend of these two pathogenic bacteria may be correlated with the development of septic shock and the risk of death in patients with sepsis.
Collapse
Affiliation(s)
- Huaying Chen
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Huiheng Liu
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Yujing Sun
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Meiqin Su
- Department of Pharmacy, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Jinzhou Lin
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Junsheng Wang
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Jueying Lin
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| | - Xiaoyan Zhao
- Emergency Intensive Care Unit, Zhongshan Hospital of Xiamen University, No.201, South Hubin Road, Xiamen, 361000, Fujian, China
| |
Collapse
|
7
|
Wu R, Xu J, Zeng H, Fan Y, Li H, Peng T, Xiao F. Golden bifid treatment regulates gut microbiota and serum metabolites to improve myocardial dysfunction in cecal ligation and puncture-induced sepsis mice. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167049. [PMID: 38301856 DOI: 10.1016/j.bbadis.2024.167049] [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: 07/27/2023] [Revised: 01/07/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
Myocardial damage is a major consequence and a significant contributor to death in cases of sepsis, a severe infection characterized by a distinct inflammatory response and a potential threat to the patient's life. Recently, the effects of intestinal microbiota and serum metabolites on sepsis have garnered increasing attention. Herein, the effects of golden bifid treatment upon cecal ligation and puncture (CLP)-induced sepsis in mice as a model for myocardial dysfunction were explored. Our results demonstrated that golden bifid treatment partially improved myocardial dysfunction and apoptosis, cardiac inflammation and oxidative stress, and intestinal mucosal permeability and barrier dysfunction in CLP-induced sepsis mice. The intestinal microbiota diversity and abundance were also altered within sepsis mice and improved by golden bifid treatment. Mucispirillum schaedleri, Acinetobacter baumannii and Lactobacullus intestinalis were significantly correlated with heart damage markers, inflammatory factors, or oxidative stress indicators. Serum differential metabolite levels were also significantly correlated with these parameters. Altogether, golden bifid treatment might be an underlying approach for treating sepsis-induced myocardial dysfunction and highlight the underlying effect of intestinal microbiota and serum metabolites on the pathogenesis and treatment of sepsis-triggered myocardial dysfunction.
Collapse
Affiliation(s)
- Rui Wu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Junmei Xu
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Hua Zeng
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yongmei Fan
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Hui Li
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Tian Peng
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Feng Xiao
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
8
|
Herrnreiter CJ, Luck ME, Cannon AR, Li X, Choudhry MA. Reduced Expression of miR-146a Potentiates Intestinal Inflammation following Alcohol and Burn Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:881-893. [PMID: 38189569 PMCID: PMC10922766 DOI: 10.4049/jimmunol.2300405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/13/2023] [Indexed: 01/09/2024]
Abstract
MicroRNAs (miRNAs) are small noncoding RNA molecules that negatively regulate gene expression. Within the intestinal epithelium, miRNAs play a critical role in gut homeostasis, and aberrant miRNA expression has been implicated in various disorders associated with intestinal inflammation and barrier disruption. In this study, we sought to profile changes in intestinal epithelial cell miRNA expression after alcohol and burn injury and elucidate their impact on inflammation and barrier integrity. Using a mouse model of acute ethanol intoxication and burn injury, we found that small intestinal epithelial cell expression of miR-146a is significantly decreased 1 d following injury. Using in vitro studies, we show that reduced miR-146a promotes intestinal epithelial cell inflammation by promoting p38 MAPK signaling via increased levels of its target TRAF6 (TNFR-associated factor 6). Furthermore, we demonstrate that in vivo miR-146a overexpression significantly inhibits intestinal inflammation 1 d following combined injury and potentially supports intestinal barrier homeostasis. Overall, this study highlights the important impact that miRNA expression can have on intestinal homeostasis and the valuable potential of harnessing aberrant miRNA expression as a therapeutic target to control intestinal inflammation.
Collapse
Affiliation(s)
- Caroline J. Herrnreiter
- Biochemistry, Molecular and Cancer Biology Program, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Marisa E. Luck
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Abigail R. Cannon
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Xiaoling Li
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Mashkoor A. Choudhry
- Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| |
Collapse
|
9
|
Xiao Y, Feng J, Jia J, Li J, Zhou Y, Song Z, Guan F, Li X, Liu L. Vitamin K1 ameliorates lipopolysaccharide-triggered skeletal muscle damage revealed by faecal bacteria transplantation. J Cachexia Sarcopenia Muscle 2024; 15:81-97. [PMID: 38018317 PMCID: PMC10834346 DOI: 10.1002/jcsm.13379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 08/03/2023] [Accepted: 09/25/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Sepsis-associated muscle weakness is common in patients of intensive care units (ICUs), and it is closely associated with poor outcomes. The mechanism of sepsis-induced muscle weakness is unclear. Recent studies have found that gut microbiota and metabolites are involved in the regulation of skeletal muscle mass and metabolism. This study aimed to investigate the effects of gut microbiota and metabolites on sepsis-associated muscle weakness. METHODS In a lipopolysaccharide (LPS)-induced inflammation mouse model, mice with different sensitivities to LPS-induced inflammation were considered as donor mice for the faecal microbiota transplantation (FMT) assay, and recipient mice were divided into sensitive (Sen) and resistant (Res) groups. Skeletal muscle mass and function, as well as colonic barrier integrity were tested and gut microbiota and metabolite composition were analysed in both groups of mice. The effect of intestinal differential metabolite vitamin K1 on LPS-triggered muscle damage was investigated, and the underlying mechanism was explored. RESULTS Recipients exhibited varying LPS-triggered muscle damage and intestinal barrier disruption. Tibialis anterior (TA) muscle of Sen exhibited upregulated expression levels of MuRF-1 (0.825 ± 0.063 vs. 0.304 ± 0.293, P = 0.0141) and MAFbx (1.055 ± 0.079 vs. 0.456 ± 0.3, P = 0.0092). Colonic tight junction proteins ZO-1 (0.550 ± 0.087 vs. 0.842 ± 0.094, P = 0.0492) and occludin (0.284 ± 0.057 vs. 0.664 ± 0.191, P = 0.0487) were significantly downregulated in the Sen group. Metabolomic analysis showed significantly higher vitamin K1 in the faeces (P = 0.0195) and serum of the Res group (P = 0.0079) than those of the Sen group. After vitamin K1 intervention, muscle atrophy-related protein expression downregulated (P < 0.05). Meanwhile SIRT1 protein expression were upregulated (0.320 ± 0.035 vs. 0.685 ± 0.081, P = 0.0281) and pNF-κB protein expression were downregulated (0.815 ± 0.295 vs. 0.258 ± 0.130, P = 0.0308). PI3K (0.365 ± 0.142 vs. 0.763 ± 0.013, P = 0.0475), pAKT (0.493 ± 0.159 vs. 1.183 ± 0.344, P = 0.0254) and pmTOR (0.509 ± 0.088 vs. 1.110 ± 0.190, P = 0.0368) protein expression levels were upregulated in TA muscle. Meanwhile, vitamin K1 attenuated serum inflammatory factor levels. CONCLUSIONS Vitamin K1 might ameliorate LPS-triggered skeletal muscle damage by antagonizing NF-κB-mediated inflammation through upregulation of SIRT1 and regulating the balance between protein synthesis and catabolism.
Collapse
Affiliation(s)
- Yuru Xiao
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, Southwest Medical University, Luzhou, China
| | - Jie Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yingshun Zhou
- Laboratory of Pathogen and Microbiology, Southwest Medical University, Luzhou, China
| | - Zhangyong Song
- Department of Pathogenic Biology, Southwest Medical University, Luzhou, China
| | - Fasheng Guan
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Xuexin Li
- Department of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Li Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
10
|
Deng Z, Ouyang Z, Mei S, Zhang X, Li Q, Meng F, Hu Y, Dai X, Zhou S, Mao K, Huang C, Dai J, Yi C, Tan N, Feng T, Long H, Tian X. Enhancing NKT cell-mediated immunity against hepatocellular carcinoma: Role of XYXD in promoting primary bile acid synthesis and improving gut microbiota. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116945. [PMID: 37490989 DOI: 10.1016/j.jep.2023.116945] [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: 05/03/2023] [Revised: 06/30/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 'Xiayuxue decoction' (XYXD) is a traditional Chinese medicine compound, composing of three natural medicines: Rheum officinale Baill., Prunus persica (L.) Batsch and Eupolyphaga sinensis Walker. It is derived from the famous traditional Chinese medical classics 'Jingui Yaolue' and has been used for thousands of years. In the Guidelines for the Diagnosis and Treatment of Primary liver Cancer issued by China's Health Commission, XYXD was applied in the treatment of primary liver cancer. AIM OF THE STUDY To clarify the pharmacodynamic material basis and mechanism of XYXD in the treatment of hepatocellular carcinoma (HCC). MATERIALS AND METHODS Firstly, the active components of XYXD and its distribution in vivo were identified by Ultraperformance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS). Then, the effective components and mechanism of XYXD against HCC were explored by network pharmacology combined with cell experiments in vitro. Furthermore, the anti-HCC effect of XYXD was determined by animal experiments in vivo. Metagenomic sequencing was used to detect its effect in gut microbiota, and targeted metabolism was used to detect the changes of bile acids in the liver. Finally, the related targets of NKT cell immune function activation were detected by RT-qPCR and Elisa. RESULTS A total of 113 active ingredients in XYXD were identified, and the distribution of active ingredients in blood, liver, tumor, cecum, intestinal contents and feces was clarified. The circulation process and active ingredient group of XYXD were preliminarily clarified. In addition, we found five anti-HCC active ingredients in XYXD through network pharmacology combined with cell experiments in vitro, among which aloe emodin had the most significant effect, and predicted the potential mechanism of XYXD against HCC through NKT cell pathway. Moreover, the inhibitory effect of XYXD on liver tumor growth was clarified by animal experiments in vivo. The mechanism was mainly to promote the production of bile salt hydrolase (BSH) by increasing the abundance of Bacteroides and Lactobacillus, BSH converts conjugated bile acids into primary bile acids, and reduces the conversion of primary bile acids to secondary bile acids by reducing the abundance of Eubacterium, thereby increasing the content of primary bile acids. Primary bile acids trigger NKT cells in the liver to produce interferon-γ to exert anti-HCC immune effects. CONCLUSION This study found that the traditional Chinese herbal formula XYXD can trigger the immune effect of NKT cells against HCC by regulating the interaction between gut microbiota and bile acids.
Collapse
Affiliation(s)
- Zhe Deng
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Zhaoguang Ouyang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, Tianjin Province, China; Department of Preventive Dentistry, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong, Guangzhou, China
| | - Si Mei
- Department of Physiology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Xue Zhang
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Qian Li
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Fanying Meng
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Yuxing Hu
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Xinjun Dai
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Siqian Zhou
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan Province, China
| | - Kexin Mao
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan Province, China
| | - Caizhi Huang
- Laboratory Department of Hunan Children's Hospital, Changsha, 410007, Hunan province, China
| | - Jingjing Dai
- College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Chun Yi
- Department of Pathology, Faculty of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Nianhua Tan
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Ting Feng
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Hongping Long
- The First Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan Province, China.
| | - Xuefei Tian
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China; Hunan Province University Key Laboratory of Oncology of Traditional Chinese Medicine Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China; Key Laboratory of Traditional Chinese Medicine for Mechanism of Tumor Prevention &Treatment, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
11
|
Ramachandran MS, Sandaradura I, Nayyar V. Prevalence of multidrug-resistant organisms in intensive care unit patients and rate of subsequent bacteraemia: a 5-year study. Anaesthesiol Intensive Ther 2024; 56:277-284. [PMID: 39917977 PMCID: PMC11781307 DOI: 10.5114/ait.2024.146641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 11/03/2024] [Indexed: 02/11/2025] Open
Abstract
INTRODUCTION Multidrug-resistant organism (MRO) bacteraemia is associated with significant mortality. A limited number of studies have examined the relationship between MRO colonisation and subsequent bacteraemia in critically ill patients. MATERIAL AND METHODS All patients with a positive surveillance swab result and a positive blood culture result for MROs admitted to the Westmead intensive care unit (ICU) between 1 January 2014 and 31 March 2019 had their results matched with ICU data extracted from the ICU database and analysed for the risk of bacteraemia among swab positive patients. RESULTS There were 3,878 (2,388 males, 1,490 females) assessable admissions during the period. The median APACHE II (Acute Physiology and Chronic Health Evaluation) score was 17. A total of 9,681 swab results were collected from 3,878 patients. Of the 3,878 patients, 818 were positive for MROs, and 3,060 were negative. Thirty-two swab positive patients (3.9%) tested positive for MROs in the blood culture, and 16 (0.52%) in the swab negative group had MROs in their blood cultures. This difference was statistically significant (adjusted [adj] OR 6.33; 95% CI: 3.40-11.76). The swab positive group was also associated with a significantly higher positive blood culture with orga-nisms other than MROs than the swab negatives (11.1% and 6.2%, respectively, adj OR 1.37; 95% CI: 1.04-1.82). The overall mortality was higher in swab positive compared to swab negative patients (20.7% vs. 13.1%, P < 0.001). The overall prevalence of MRO colonisation was 21% in our cohort. CONCLUSIONS The risk of MRO bacteraemia was higher among patients with a positive surveillance swab result for the organism compared to those with a negative swab, but this was not associated with higher mortality in that group.
Collapse
Affiliation(s)
| | - Indunil Sandaradura
- Centre for Infectious Diseases and Microbiology, Australia
- University of Sydney School of Medicine, Australia
| | - Vineet Nayyar
- University of Sydney, Australia
- Intensive Care Services, Westmead Hospital, Westmead, New South Wales, Australia
| |
Collapse
|
12
|
Luo Y, Zhou Y, Huang P, Zhang Q, Luan F, Peng Y, Wei J, Li N, Wang C, Wang X, Zhang J, Yu K, Zhao M, Wang C. Causal relationship between gut Prevotellaceae and risk of sepsis: a two-sample Mendelian randomization and clinical retrospective study in the framework of predictive, preventive, and personalized medicine. EPMA J 2023; 14:697-711. [PMID: 38094582 PMCID: PMC10713913 DOI: 10.1007/s13167-023-00340-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/28/2023] [Indexed: 10/25/2024]
Abstract
Objective Gut microbiota is closely related to sepsis. Recent studies have suggested that Prevotellaceae could be associated with intestinal inflammation; however, the causal relationship between Prevotellaceae and sepsis remains uncertain. From the perspective of predictive, preventive, and personalized medicine (PPPM), exploring the causal relationship between gut Prevotellaceae and sepsis could provide opportunity for targeted prevention and personalized treatment. Methods The genome-wide association study (GWAS) summary-level data of Prevotellaceae (N = 7738) and sepsis were obtained from the Dutch Microbiome Project and the UK Biobank (sepsis, 1380 cases; 429,985 controls). MR analysis was conducted to estimate the associations between Prevotellaceae and sepsis risk. The 16S rRNA sequencing analysis was conducted to calculate the relative abundance of Prevotellaceae in sepsis patients to explore the relationship between Prevotellaceae relative abundance and the 28-day mortality. Results Genetic liability to f__Prevotellaceae (OR, 1.91; CI, 1.35-2.71; p = 0.0003) was associated with a high risk of sepsis with inverse-variance weighted (IVW). The median Prevotellaceae relative abundance in non-survivors was significantly higher than in survivors (2.34% vs 0.17%, p < 0.001). Multivariate analysis confirmed that Prevotellaceae relative abundance (OR, 1.10; CI, 1.03-1.22; p = 0.027) was an independent factor of 28-day mortality in sepsis patients. ROC curve analysis indicated that Prevotellaceae relative abundance (AUC: 0.787, 95% CI: 0.671-0.902, p = 0.0003) could predict the prognosis of sepsis patients. Conclusion Our results revealed that Prevotellaceae was causally associated with sepsis and affected the prognosis of sepsis patients. These findings may provide insights to clinicians on developing improved sepsis PPPM strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00340-6.
Collapse
Affiliation(s)
- Yinghao Luo
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Yang Zhou
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Pengfei Huang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Qianqian Zhang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Feiyu Luan
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Yahui Peng
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Jieling Wei
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Nana Li
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Chunying Wang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Xibo Wang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Jiannan Zhang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Kaijiang Yu
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Mingyan Zhao
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| | - Changsong Wang
- Departments of Critical Care Medicine, the First Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang China
- Heilongjiang Provincial Key Laboratory of Critical Care Medicine, 23 Postal Street, Nangang District, Harbin, 150001 Heilongjiang China
| |
Collapse
|
13
|
Zhang L, Shi X, Qiu H, Liu S, Yang T, Li X, Liu X. Protein modification by short-chain fatty acid metabolites in sepsis: a comprehensive review. Front Immunol 2023; 14:1171834. [PMID: 37869005 PMCID: PMC10587562 DOI: 10.3389/fimmu.2023.1171834] [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: 02/22/2023] [Accepted: 09/15/2023] [Indexed: 10/24/2023] Open
Abstract
Sepsis is a major life-threatening syndrome of organ dysfunction caused by a dysregulated host response due to infection. Dysregulated immunometabolism is fundamental to the onset of sepsis. Particularly, short-chain fatty acids (SCFAs) are gut microbes derived metabolites serving to drive the communication between gut microbes and the immune system, thereby exerting a profound influence on the pathophysiology of sepsis. Protein post-translational modifications (PTMs) have emerged as key players in shaping protein function, offering novel insights into the intricate connections between metabolism and phenotype regulation that characterize sepsis. Accumulating evidence from recent studies suggests that SCFAs can mediate various PTM-dependent mechanisms, modulating protein activity and influencing cellular signaling events in sepsis. This comprehensive review discusses the roles of SCFAs metabolism in sepsis associated inflammatory and immunosuppressive disorders while highlights recent advancements in SCFAs-mediated lysine acylation modifications, such as substrate supplement and enzyme regulation, which may provide new pharmacological targets for the treatment of sepsis.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xinhui Shi
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Hongmei Qiu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Sijia Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Ting Yang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Drug Metabolism, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xin Liu
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
14
|
Hwang TZ, Wang YM, Jeng SF, Lee YC, Chen TS, Su SY, Huang CC, Lam CF. Intraoperative Enteral Nutrition Feeding in Free-Flap Healing after Reconstruction Surgery for Head and Neck Cancers. Otolaryngol Head Neck Surg 2023; 169:843-851. [PMID: 36960779 DOI: 10.1002/ohn.335] [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: 10/24/2022] [Revised: 01/31/2023] [Accepted: 02/26/2023] [Indexed: 03/25/2023]
Abstract
OBJECTIVE To investigate the beneficial outcomes of intraoperative enteral feeding in free-flap regeneration after extended head and neck cancer resection and flap reconstruction surgery. STUDY DESIGN A pilot randomized, double-blind, placebo-controlled clinical trial. SETTING Single tertiary care center. METHODS Patients with advanced head and neck cancers requiring radical tumor resections and free-flap reconstruction were randomly assigned to receive intraoperative enteral nutrition feeding (100 kcal/100 mL at 10-20 mL/h) via a nasogastric tube during free-flap reconstruction (n = 28) or continue fasting (n = 28). The primary outcome was impaired free-flap regeneration that required surgical reintervention within 90 days after the operation. Participants were enrolled between April 2020 and January 2022; the 90-day follow-up ended in April 2022. RESULTS The incidence of total or partial flap failure was similar between the 2 groups (14.2% or n = 4 in each group), but the rate of wound dehiscence or edge necrosis was significantly reduced in the feeding group (n = 6 vs 0 for fasting vs feeding; absolute risk reduction, 25.0% [95% confidence interval, 6.9-43.0]%; p = 0.022). Hospital stay length was shorter (p = 0.042) and hand grip strength was better preserved (p = 0.025) in the feeding group. Plasma concentrations of interleukin (IL)-6 and IL-8 after the operation increased significantly more in the fasting group. Perioperative adverse events did not differ between the 2 groups. CONCLUSION Perioperative enteral feeding is a simple, safe, and effective approach to improve perioperative systemic catabolism and proinflammatory reactions, thereby enhancing early wound regeneration after major operations.
Collapse
Affiliation(s)
- Tzer-Zen Hwang
- Department of Otolaryngology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Ming Wang
- Department of Critical Care Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Seng-Feng Jeng
- Department of Plastic Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yi-Chen Lee
- Department of Nutrition Therapy, E-Da Hospital and E-Da Cancer Hospital, Kaohsiung, Taiwan
- Department of Nutrition, I-Shou University, Kaohsiung, Taiwan
| | - Tzu-Shan Chen
- Department of Medical Research, E-Da Hospital and E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Shin-Ying Su
- Department of Anesthesiology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chien-Chi Huang
- Department of Medical Research, E-Da Hospital and E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Chen-Fuh Lam
- Department of Anesthesiology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
- Department of Anesthesiology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chia-Yi, Taiwan
| |
Collapse
|
15
|
Wen B, Farooqui A, Bourdon C, Tarafdar N, Ngari M, Chimwezi E, Thitiri J, Mwalekwa L, Walson JL, Voskuijl W, Berkley JA, Bandsma RHJ. Intestinal disturbances associated with mortality of children with complicated severe malnutrition. COMMUNICATIONS MEDICINE 2023; 3:128. [PMID: 37773543 PMCID: PMC10541881 DOI: 10.1038/s43856-023-00355-0] [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: 11/23/2022] [Accepted: 09/11/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND Children admitted to hospital with complicated severe malnutrition (CSM) have high mortality despite compliance with standard WHO management guidelines. Limited data suggests a relationship between intestinal dysfunction and poor prognosis in CSM, but this has not been explicitly studied. This study aimed to evaluate the role of intestinal disturbances in CSM mortality. METHODS A case-control study nested within a randomized control trial was conducted among children hospitalized with CSM in Kenya and Malawi. Children who died (cases, n = 68) were compared with those who were discharged, propensity matched to the cases on age, HIV and nutritional status (controls, n = 68) on fecal metabolomics that targeted about 70 commonly measured metabolites, and enteropathy markers: fecal myeloperoxidase (MPO), fecal calprotectin, and circulating intestinal fatty acid binding protein (I-FABP). RESULTS The fecal metabolomes of cases show specific reductions in amino acids, monosaccharides, and microbial fermentation products, when compared to controls. SCFA levels did not differ between groups. The overall fecal metabolomics signature moderately differentiates cases from controls (AUC = 0.72). Enteropathy markers do not differ between groups overall, although serum I-FABP is elevated in cases in a sensitivity analysis among non-edematous children. Integrative analysis with systemic data suggests an indirect role of intestinal inflammation in the causal path of mortality. CONCLUSIONS Intestinal disturbances appear to have an indirect association with acute mortality. Findings of the study improve our understanding of pathophysiological pathways underlying mortality of children with CSM.
Collapse
Affiliation(s)
- Bijun Wen
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - Amber Farooqui
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - Celine Bourdon
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
| | - Nawar Tarafdar
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada
| | - Moses Ngari
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | | | - Johnstone Thitiri
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
| | - Laura Mwalekwa
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Paediatrics, Coast General Hospital, Mombasa, Kenya
| | - Judd L Walson
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- Departments of Global Health, Medicine, Pediatrics and Epidemiology, University of Washington, Seattle, WA, USA
| | - Wieger Voskuijl
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- Amsterdam Institute for Global Health and Development, Department of Global Health, Amsterdam University Medical Centres, Amsterdam, The Netherlands
- Department of Paediatrics and Child Health, Kamuzu University of Health Sciences (formerly College of Medicine), Blantyre, Malawi
| | - James A Berkley
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya
- KEMRI/Wellcome Trust Research Programme, Kilifi, Kenya
- Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Robert H J Bandsma
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, Canada.
- Department of Translational medicine, Hospital for Sick Children, Toronto, Canada.
- The Childhood Acute Illness & Nutrition Network, Nairobi, Kenya.
- Department of Biomedical Sciences, Kamuzu University of Health Sciences (formerly College of Medicine), Blantyre, Malawi.
| |
Collapse
|
16
|
Huang R, Yao Y, Tong X, Wang L, Qian W, Lu J, Zhang W, Liu Y, Wang S, Xian S, Zhu Y, Huang J, Guo X, Gu M, Lv H, Bi W, Meng C, Chang Z, Zhang J, Xu D, Ji S. Tracing the evolving dynamics and research hotspots of microbiota and immune microenvironment from the past to the new era. Microbiol Spectr 2023; 11:e0013523. [PMID: 37768071 PMCID: PMC10581186 DOI: 10.1128/spectrum.00135-23] [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: 01/10/2023] [Accepted: 07/31/2023] [Indexed: 09/29/2023] Open
Abstract
Gut microbiota can regulate many physiological processes within gastrointestinal tract and other distal sites. Dysbiosis may not only influence chronic diseases like the inflammatory bowel disease (IBD), metabolic disease, tumor and its therapeutic efficacy, but also deteriorate acute injuries. This article aims to review the documents in this field and summarize the research hotspots as well as developing processes. Gut microbiota and immune microenvironment-related documents from 1976 to 2022 were obtained from the Web of Science Core Collection database. Bibliometrics was used to assess the core authors and journals, most contributive countries and affiliations together with hotspots in this field and keyword co-occurrence analysis. Data were visualized to help comprehension. Nine hundred and twelve documents about gut microbiota and immune microenvironment were retrieved, and the annual publications increased gradually. The most productive author, country, and affiliation were "Zitvogel L," USA and "UNIV TEXAS MD ANDERSON CANC CTR," respectively. FRONTIERS IN IMMUNOLOGY, CANCERS, and INTERNATIONAL JOURNAL OF MOLECULAR SCIENCE were the periodicals with most publications. Keyword co-occurrence analysis identified three clusters, including gut microbiota, inflammation, and IBD. Combined with the visualized analysis of documents and keyword co-occurrence as well as literature reading, we recognized three key topics of gut microbiota: cancer and therapy; immunity, inflammation and IBD; acute injuries and metabolic diseases. This article revealed researches on gut microbiota and immune microenvironment were growing. More attention should be given to the latest hotspots like gut microbiota, inflammation, IBD, cancer and immunotherapy, acute traumas, and metabolic diseases.IMPORTANCEGut microbiota can regulate many physiological processes within gastrointestinal tract and other distal sites. Dysbiosis may not only influence chronic diseases like inflammatory bowel disease (IBD), metabolic disease, tumor and its therapeutic efficacy, but also deteriorate acute injuries. While the application of bibliometrics in the field of gut microbiota and immune microenvironment still remains blank, which focused more on the regulation of the gut microbiota on the immune microenvironment of different kinds of diseases. Here, we intended to review and summarize the presented documents in gut microbiota and immune microenvironment field by bibliometrics. And we revealed researches on gut microbiota and immune microenvironment were growing. More attention should be given to the latest hotspots like gut microbiota, inflammation, IBD, cancer and immunotherapy, acute traumas, and metabolic diseases.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yuntao Yao
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xirui Tong
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Lei Wang
- Beijing Genomics Institute (BGI), Shenzhen, China
| | - Weijin Qian
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianyu Lu
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Wei Zhang
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Yifan Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siqiao Wang
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Shuyuan Xian
- Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Yushu Zhu
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Jie Huang
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xinya Guo
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Minyi Gu
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Hanlin Lv
- Beijing Genomics Institute (BGI), Shenzhen, China
| | - Wenshuai Bi
- Beijing Genomics Institute (BGI), Shenzhen, China
| | - Chenwei Meng
- Beijing Genomics Institute (BGI), Shenzhen, China
| | - Zhengyan Chang
- Department of Pathology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Zhang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dayuan Xu
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| | - Shizhao Ji
- Department of Burn Surgery, First Affiliated Hospital of Naval Medical University, and Research Unit of Key Techniques for Treatment of Burns and Combined Burns and Trauma Injury, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
17
|
Microbiome and Metabolomics in Liver Cancer: Scientific Technology. Int J Mol Sci 2022; 24:ijms24010537. [PMID: 36613980 PMCID: PMC9820585 DOI: 10.3390/ijms24010537] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/12/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022] Open
Abstract
Primary liver cancer is a heterogeneous disease. Liver cancer metabolism includes both the reprogramming of intracellular metabolism to enable cancer cells to proliferate inappropriately and adapt to the tumor microenvironment and fluctuations in regular tissue metabolism. Currently, metabolomics and metabolite profiling in liver cirrhosis, liver cancer, and hepatocellular carcinoma (HCC) have been in the spotlight in terms of cancer diagnosis, monitoring, and therapy. Metabolomics is the global analysis of small molecules, chemicals, and metabolites. Metabolomics technologies can provide critical information about the liver cancer state. Here, we review how liver cirrhosis, liver cancer, and HCC therapies interact with metabolism at the cellular and systemic levels. An overview of liver metabolomics is provided, with a focus on currently available technologies and how they have been used in clinical and translational research. We also list scalable methods, including chemometrics, followed by pathway processing in liver cancer. We conclude that important drivers of metabolomics science and scientific technologies are novel therapeutic tools and liver cancer biomarker analysis.
Collapse
|
18
|
Xiang C, Chen Y, Liu X, Zheng Z, Zhang H, Tan C. Prevention and Treatment of Grade C Postoperative Pancreatic Fistula. J Clin Med 2022; 11:7516. [PMID: 36556131 PMCID: PMC9784648 DOI: 10.3390/jcm11247516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Postoperative pancreatic fistula (POPF) is a troublesome complication after pancreatic surgeries, and grade C POPF is the most serious situation among pancreatic fistulas. At present, the incidence of grade C POPF varies from less than 1% to greater than 9%, with an extremely high postoperative mortality rate of 25.7%. The patients with grade C POPF finally undergo surgery with a poor prognosis after various failed conservative treatments. Although various surgical and perioperative attempts have been made to reduce the incidence of grade C POPF, the rates of this costly complication have not been significantly diminished. Hearteningly, several related studies have found that intra-abdominal infection from intestinal flora could promote the development of grade C POPF, which would help physicians to better prevent this complication. In this review, we briefly introduced the definition and relevant risk factors for grade C POPF. Moreover, this review discusses the two main pathways, direct intestinal juice spillover and bacterial translocation, by which intestinal microbes enter the abdominal cavity. Based on the abovementioned theory, we summarize the operation techniques and perioperative management of grade C POPF and discuss novel methods and surgical treatments to reverse this dilemma.
Collapse
Affiliation(s)
| | | | | | | | | | - Chunlu Tan
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
19
|
Mohamadian M, Parsamanesh N, Chiti H, Sathyapalan T, Sahebkar A. Protective effects of curcumin on ischemia/reperfusion injury. Phytother Res 2022; 36:4299-4324. [PMID: 36123613 DOI: 10.1002/ptr.7620] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/06/2022] [Accepted: 08/24/2022] [Indexed: 12/13/2022]
Abstract
Ischemia/reperfusion (I/R) injury is a term used to describe phenomena connected to the dysfunction of various tissue damage due to reperfusion after ischemic injury. While I/R may result in systemic inflammatory response syndrome or multiple organ dysfunction syndrome, there is still a long way to improve therapeutic outcomes. A number of cellular metabolic and ultrastructural alterations occur by prolonged ischemia. Ischemia increases the expression of proinflammatory gene products and bioactive substances within the endothelium, such as cytokines, leukocytes, and adhesion molecules, even as suppressing the expression of other "protective" gene products and substances, such as thrombomodulin and constitutive nitric oxide synthase (e.g., prostacyclin, nitric oxide [NO]). Curcumin is the primary phenolic pigment derived from turmeric, the powdered rhizome of Curcuma longa. Numerous studies have shown that curcumin has strong antiinflammatory and antioxidant characteristics. It also prevents lipid peroxidation and scavenges free radicals like superoxide anion, singlet oxygen, NO, and hydroxyl. In our study, we highlight the mechanisms of protective effects of curcumin against I/R injury in various organs.
Collapse
Affiliation(s)
- Malihe Mohamadian
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Negin Parsamanesh
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossein Chiti
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Huang Z, Huang Y, Chen J, Tang Z, Chen Y, Liu H, Huang M, Qing L, Li L, Wang Q, Jia B. The role and therapeutic potential of gut microbiome in severe burn. Front Cell Infect Microbiol 2022; 12:974259. [DOI: 10.3389/fcimb.2022.974259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/21/2022] [Indexed: 11/19/2022] Open
Abstract
Severe burn is a serious acute trauma that can lead to significant complications such as sepsis, multiple organ failure, and high mortality worldwide. The gut microbiome, the largest microbial reservoir in the human body, plays a significant role in this pathogenic process. Intestinal dysbiosis and disruption of the intestinal mucosal barrier are common after severe burn, leading to bacterial translocation to the bloodstream and other organs of the body, which is associated with many subsequent severe complications. The progression of some intestinal diseases can be improved by modulating the composition of gut microbiota and the levels of its metabolites, which also provides a promising direction for post-burn treatment. In this article, we summarised the studies describing changes in the gut microbiome after severe burn, as well as changes in the function of the intestinal mucosal barrier. Additionally, we presented the potential and challenges of microbial therapy, which may provide microbial therapy strategies for severe burn.
Collapse
|
21
|
Lv ZY, Shi YL, Bassi GS, Chen YJ, Yin LM, Wang Y, Ulloa L, Yang YQ, Xu YD. Electroacupuncture at ST36 ( Zusanli) Prevents T-Cell Lymphopenia and Improves Survival in Septic Mice. J Inflamm Res 2022; 15:2819-2833. [PMID: 35535053 PMCID: PMC9078867 DOI: 10.2147/jir.s361466] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Purpose Sepsis is the main cause of death in intensive care unit. Maladaptive cytokine storm and T-cell lymphopenia are critical prognosis predictors of sepsis. Electroacupuncture (EA) is expected to be an effective intervention to prevent sepsis. This study aims to determine the potential of EA at ST36 (Zusanli) to prevent experimental septic mice. Methods Mice were randomly assigned into PBS, LPS, or EA+LPS group. EA (0.1 mA, continuous wave, 10 Hz) was performed stimulating the ST36 for 30 min, once a day for 3 days. After the third day, all mice were challenged with PBS or LPS (4 mg/kg) simultaneously. Mice were evaluated for survival, ear temperature, and other clinical symptoms. Lung and small intestine tissue injuries were analyzed by hematoxylin and eosin staining. Bio-Plex cytokine assay was used to analyze the concentration of cytokines. T lymphocytes were analyzed by flow cytometry and Western blot assays. The role of T cells in preventing sepsis by EA was analyzed by using nude mice lacking T lymphocytes. Results EA at ST36 improved survival, symptom scores, and ear temperature of endotoxemic mice. EA also improved dramatically pulmonary and intestinal injury by over 50% as compared to untreated mice. EA blunted the inflammatory cytokine storm by inducing a lasting inhibition of the production of major inflammatory factors (TNF-α, IL-1β, IL-5, IL-6, IL-10, IL-17A, eotaxin, IFN-γ, MIP-1β and KC). Flow cytometry and Western blot analyses showed EA significantly reduced T-lymphocyte apoptosis and pyroptosis. Furthermore, T lymphocytes were critical for the effects of EA at ST36 stimulation blunted serum TNF-α levels in wild-type but not in nude mice. Conclusion EA halted systemic inflammation and improved survival in endotoxemic mice. These effects are associated with the protective effect of EA on T lymphocytes, and T cells are required in the anti-inflammatory effects of EA in sepsis.
Collapse
Affiliation(s)
- Zhi-Ying Lv
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yang-Lin Shi
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Gabriel Shimizu Bassi
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yan-Jiao Chen
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Lei-Miao Yin
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yu Wang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Luis Ulloa
- Department of Anesthesiology, Duke University, Durham, NC, USA
| | - Yong-Qing Yang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yu-Dong Xu
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
22
|
Oh BLZ, Fan L, Lee SHR, Foo KM, Chiew KH, Seeto ZZL, Chen ZW, Neoh CCC, Liew GSM, Eng JJ, Lam JCM, Quah TC, Tan AM, Chan YH, Yeoh AEJ. Life-threatening infections during treatment for acute lymphoblastic leukemia on the Malaysia-Singapore 2003 and 2010 clinical trials: A risk prediction model. Asia Pac J Clin Oncol 2022; 18:e456-e468. [PMID: 35134276 DOI: 10.1111/ajco.13756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022]
Abstract
AIM Life-threatening infections significantly impact the care of children undergoing therapy for acute lymphoblastic leukemia (ALL) who are at risk of severe sepsis due to both host and treatment factors. Our aim was to develop a life-threatening infection risk prediction model that would allow remote rapid triage of patients to reduce time to first dose of antibiotics and sepsis-related mortality. METHODS A retrospective analysis of 2068 fever episodes during ALL therapy was used for model building and subsequent internal validation. RESULTS Three hundred and seventy-seven patients were treated for ALL in two institutions with comparable critical and supportive care resources. A total of 55 patients accounted for 71 admissions to the critical care unit for sepsis that led to eight septic deaths during a 16-year study period. A retrospective analysis of risk factors for sepsis enabled us to build a model focused on 13 variables that discriminated admissions requiring critical care well: area under the receiver operating characteristic curve of .82; 95% CI .76-.87, p<.001, and Brier score of .033. Significant univariate predictors included neutropenia, presence of symptoms of abdominal pain, diarrhea, fever during induction or steroid-based phases, and the lack of any localizing source of infection at time of presentation. CONCLUSION We have developed a risk prediction model that can reliably identify ALL patients undergoing treatment who are at a higher risk of life-threatening sepsis. Clinical applicability can potentially be extended to low-middle income settings, and its utility should be further studied in real-world settings.
Collapse
Affiliation(s)
- Bernice L Z Oh
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Lijia Fan
- Division of Critical Care, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Shawn H R Lee
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Koon Mian Foo
- Department of Paediatric Subspecialties Haematology/Oncology Service, KK Women's and Children's Hospital, Singapore
| | - Kean Hui Chiew
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Zelia Z L Seeto
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Zhi Wei Chen
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Cheryl C C Neoh
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore
| | - Germaine S M Liew
- Department of Paediatric Subspecialties Haematology/Oncology Service, KK Women's and Children's Hospital, Singapore
| | - Jing Jia Eng
- Department of Paediatric Subspecialties Haematology/Oncology Service, KK Women's and Children's Hospital, Singapore
| | - Joyce C M Lam
- Department of Paediatric Subspecialties Haematology/Oncology Service, KK Women's and Children's Hospital, Singapore
| | - Thuan Chong Quah
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Ah Moy Tan
- Department of Paediatric Subspecialties Haematology/Oncology Service, KK Women's and Children's Hospital, Singapore
| | - Yiong Huak Chan
- Biostatistics Unit, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Allen E J Yeoh
- Viva-University Children's Cancer Centre, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, National University Health System, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| |
Collapse
|
23
|
Simvastatin Improves Outcomes of Endotoxin-induced Coagulopathy by Regulating Intestinal Microenvironment. Curr Med Sci 2022; 42:26-38. [PMID: 35041135 DOI: 10.1007/s11596-022-2526-3] [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: 04/09/2021] [Accepted: 07/04/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVE The systemic inflammatory response is regarded as the major cause of endotoxin-induced coagulopathy, which is a strong predictor of mortality in patients with severe sepsis. Simvastatin plays an important role in reducing inflammation. In addition, the gut has long been hypothesized to be the "motor" of critical illness, driving or aggravating sepsis by the increased intestinal permeability and bacterial translocation. Whether simvastatin plays a role in severe endotoxin-induced coagulopathy through the gut is unclear. METHODS In this study, mice were administered 20 mg/kg simvastatin by gavage for 2 weeks and then intraperitoneally injected with 50 mg/kg endotoxin. Twelve h later, cytokine release, coagulation dysfunction, organ damage, and survival were assessed. Besides, the intestinal barrier, permeability, bacteria abundance, and translocation were evaluated. RESULTS We found that the severity of endotoxin-induced coagulopathy was significantly improved in simvastatin-pretreated mice, who showed attenuated depletion of coagulation factors and platelets, decreased plasminogen activator inhibitor-1 (PAI-1) expression, reduced organ fibrin deposition, and improved survival time. Also, simvastatin reduced epithelial apoptosis and improved intestinal barrier function by upregulating antimicrobial peptides, lysozyme, and mucins. Simvastatin increased Lactobacillales counts, while the lipopolysaccharide group showed increased Desulfovibrio and Mucispirillum, which can produce harmful toxins. Finally, the decreased intestinal permeability in the simvastatin group caused reduced bacterial translocation in the organs and blood, both in terms of quantity and species. CONCLUSION Simvastatin improves the prognosis of severe endotoxemia, and the intestinal microenvironment participates in this process.
Collapse
|
24
|
Xu F, Yang C, Tang M, Wang M, Cheng Z, Chen D, Chen X, Liu K. The Role of Gut Microbiota and Genetic Susceptibility in the Pathogenesis of Pancreatitis. Gut Liver 2021; 16:686-696. [PMID: 34911043 PMCID: PMC9474482 DOI: 10.5009/gnl210362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022] Open
Abstract
Pancreatitis is one of the most common inflammatory diseases of the pancreas caused by autodigestion induced by excessive premature protease activation. However, recognition of novel pathophysiological mechanisms remains a still challenge. Both genetic and environmental factors contribute to the pathogenesis of pancreatitis, and the gut microbiota is a potential source of an environmental effect. In recent years, several new frontiers in gut microbiota and genetic risk assessment research have emerged and improved the understanding of the disease. These investigations showed that the disease progression of pancreatitis could be regulated by the gut microbiome, either through a translocation influence or in a host immune response manner. Meanwhile, the onset of the disease is also associated with the heritage of a pathogenic mutation, and the disease progression could be modified by genetic risk factors. In this review, we focused on the recent advances in the role of gut microbiota in the pathogenesis of pancreatitis, and the genetic susceptibility in pancreatitis.
Collapse
Affiliation(s)
- Fumin Xu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chunmei Yang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingcheng Tang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Ming Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhenhao Cheng
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongfeng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Kaijun Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
25
|
Doudakmanis C, Bouliaris K, Kolla C, Efthimiou M, Koukoulis GD. Bacterial translocation in patients undergoing major gastrointestinal surgery and its role in postoperative sepsis. World J Gastrointest Pathophysiol 2021; 12:106-114. [PMID: 34877025 PMCID: PMC8611185 DOI: 10.4291/wjgp.v12.i6.106] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/14/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Bacteria of the human intestinal microflora have a dual role. They promote digestion and are part of a defense mechanism against pathogens. These bacteria could become potential pathogens under certain circumstances. The term “bacterial translocation” describes the passage of bacteria of the gastrointestinal tract through the intestinal mucosa barrier to mesenteric lymph nodes and other organs. In some cases, the passage of bacteria and endotoxins could result in blood stream infections and in multiple organ failure. Open elective abdominal surgery more frequently results in malfunction of the intestinal barrier and subsequent bacterial translocation and blood stream infections than laparoscopic surgery. Postoperative sepsis is a common finding in patients who have undergone non-elective abdominal surgeries, including trauma patients treated with laparotomy. Postoperative sepsis is an emerging issue, as it changes the treatment plan in surgical patients and prolongs hospital stay. The association between bacterial translocation and postoperative sepsis could provide novel treatment options.
Collapse
Affiliation(s)
- Christos Doudakmanis
- Department of General Surgery, General Hospital of Larissa, Larisa 41221, Greece
| | | | - Christina Kolla
- Department of General Surgery, General Hospital of Larissa, Larisa 41221, Greece
| | - Matthaios Efthimiou
- Department of General Surgery, General Hospital of Larissa, Larisa 41221, Greece
| | - Georgios D Koukoulis
- Department of General Surgery, General Hospital of Larissa, Larisa 41221, Greece
| |
Collapse
|
26
|
Lentinan Attenuates Damage of the Small Intestinal Mucosa, Liver, and Lung in Mice with Gut-Origin Sepsis. J Immunol Res 2021; 2021:2052757. [PMID: 34790828 PMCID: PMC8592742 DOI: 10.1155/2021/2052757] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/15/2021] [Accepted: 10/26/2021] [Indexed: 01/08/2023] Open
Abstract
This study is aimed at exploring the effects of lentinan on small intestinal mucosa as well as lung and liver injury in mice with gut-origin sepsis. Cecal ligation and perforation (CLP) were used to construct a mouse model of gut-origin sepsis. The mice were randomly divided into six groups: sham operation group (sham), gut-origin sepsis model group (CLP), ulinastatin-positive drug control group (UTI), lentinan low concentration group (LTN-L, 5 mg/kg), lentinan medium concentration group (LTN-M, 10 mg/kg), and lentinan high concentration group (LTN-H, 20 mg/kg). H&E staining was used to detect the pathological damage of the small intestine, liver, and lung. The serum of mice in each group was collected to detect the expression changes of inflammatory cytokines, oxidative stress biomarkers, and liver function indexes. In vitro assessment of bacterial translocation was achieved through inoculated culture media. Western blot and RT-qPCR were used to detect the expression of molecules related to the NF-κB signaling pathway in the small intestine tissues of mice. The results showed that compared with the CLP group, the injury degree of the small intestine, liver, and lung in mice with gut-origin sepsis was improved with the increase of lentinan concentration. In addition, TNF-α, IL-1β, IL-6, and HMGB1 were decreased with the increase of lentinan concentration, but the expression of IL-10 was increased. Lentinan could also reduce the expression of oxidative stress injury indexes and liver function indexes and inhibit bacterial translocation to liver and lung tissues. Further mechanism investigation revealed that lentinan downregulated the expression of the NF-κB signaling pathway molecules (NF-κB, TLR4, and Bax) and upregulated the expression of occludin and Bcl-2. In conclusion, lentinan inhibits the activity of the NF-κB signaling pathway, thus attenuating injuries of small intestinal mucosa and liver and lung in mice with gut-origin sepsis and reducing the inflammatory response in the process of sepsis.
Collapse
|
27
|
Integrated analysis of dysregulated microRNA and mRNA expression in intestinal epithelial cells following ethanol intoxication and burn injury. Sci Rep 2021; 11:20213. [PMID: 34642361 PMCID: PMC8510995 DOI: 10.1038/s41598-021-99281-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023] Open
Abstract
Gut barrier dysfunction is often implicated in pathology following alcohol intoxication and burn injury. MicroRNAs (miRNAs) are negative regulators of gene expression that play a central role in gut homeostasis, although their role after alcohol and burn injury is poorly understood. We performed an integrated analysis of miRNA and RNA sequencing data to identify a network of interactions within small intestinal epithelial cells (IECs) which could promote gut barrier disruption. Mice were gavaged with ~ 2.9 g/kg ethanol and four hours later given a ~ 12.5% TBSA full thickness scald injury. One day later, IECs were harvested and total RNA extracted for RNA-seq and miRNA-seq. RNA sequencing showed 712 differentially expressed genes (DEGs) (padj < 0.05) in IECs following alcohol and burn injury. Furthermore, miRNA sequencing revealed 17 differentially expressed miRNAs (DEMs) (padj < 0.1). Utilizing the miRNet, miRDB and TargetScan databases, we identified both validated and predicted miRNA gene targets. Integration of small RNA sequencing data with mRNA sequencing results identified correlated changes in miRNA and target expression. Upregulated miRNAs were associated with decreased proliferation (miR-98-3p and miR-381-3p) and cellular adhesion (miR-29a-3p, miR-429-3p and miR3535), while downregulated miRNAs were connected to upregulation of apoptosis (Let-7d-5p and miR-130b-5p) and metabolism (miR-674-3p and miR-185-5p). Overall, these findings suggest that alcohol and burn injury significantly alters the mRNA and miRNA expression profile of IECs and reveals numerous miRNA–mRNA interactions that regulate critical pathways for gut barrier function after alcohol and burn injury.
Collapse
|
28
|
Fan L, Lee JH. Enteral feeding and the microbiome in critically ill children: a narrative review. Transl Pediatr 2021; 10:2778-2791. [PMID: 34765500 PMCID: PMC8578772 DOI: 10.21037/tp-20-349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 04/09/2021] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE This narrative review summarizes our current knowledge on the interplay between enteral nutrition (EN) and gut microbiota in critically ill children, using examples from two commonly encountered diagnoses in the pediatric intensive care unit (PICU): severe sepsis and acute respiratory distress syndrome (ARDS). This review will also highlight potential areas of therapeutic interventions that should be explored in future studies. BACKGROUND Critically ill children display extreme dysbiosis in their gut microbiome. Factors within the PICU that are often associated with dysbiosis include the use of broad-spectrum antibiotics, proton-pump inhibitors (PPIs), intravenous morphine, and fasting. Dysbiosis can potentially lead to adverse clinical outcomes (e.g., nosocomial infection, and prolonged hospitalization). EN may modulate dysbiosis. The gut microbiota is involved in the breaking down of macronutrients, mainly carbohydrates and proteins. Fermentation of undigestible carbohydrate (e.g., inulin and oligosaccharides), and amino acids by large intestine microbiota produces short chain fatty acids (SCFAs). SCFAs serve as the main fuel source for enterocytes and help to maintain healthy gut lining. Changes to selected components of macronutrients can result in alterations in gut microbiome and have potentially beneficial effects in patients in the PICU. METHODS A comprehensive search of the MEDLINE, Cochrane Library and Google Scholar databases was conducted using appropriate MESH terms and keywords. In this narrative review, we provide a summary of current knowledge on effect of EN on gut microbiota in pediatric studies, but also describes animal- and lab-based, as well as adult studies where relevant. CONCLUSIONS The gut microbiome can be altered by dietary modifications and common PICU practices and treatment. Although there are strong associations in restoring eubiosis and improvement in clinical outcomes, proving causality remains challenging. Further microbiome research is needed to provide mechanistic insights into the impact of the ever changing gut microbiome. In the future, new microbiota targeted therapies could potentially be the treatment of challenging PICU conditions and restore homeostasis in these children.
Collapse
Affiliation(s)
- Lijia Fan
- Division of Paediatric Critical Care, Department of Paediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Jan Hau Lee
- Children's Intensive Care Unit, KK Women's and Children's Hospital, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
29
|
Abstract
ABSTRACT Burn injuries are a common form of traumatic injury that leads to significant morbidity and mortality worldwide. Burn injuries are characterized by inflammatory processes and alterations in numerous organ systems and functions. Recently, it has become apparent that the gastrointestinal bacterial microbiome is a key component of regulating the immune response and recovery from burn and can also contribute to significant detrimental sequelae after injury, such as sepsis and multiple organ failure. Microbial dysbiosis has been linked to multiple disease states; however, its role in exacerbating acute traumatic injuries, such as burn, is poorly understood. In this article, we review studies that document changes in the intestinal microbiome after burn injury, assess the implications in post-burn pathogenesis, and the potential for further discovery and research.
Collapse
Affiliation(s)
- Marisa E. Luck
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Caroline J. Herrnreiter
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Biochemistry and Molecular Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Mashkoor A. Choudhry
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Biochemistry and Molecular Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| |
Collapse
|
30
|
Ardehali SH, Eslamian G, Malek S. Associations between systemic inflammation and intestinal permeability with Onodera's prognostic nutritional index in critically ill patients. Acute Crit Care 2021; 36:361-368. [PMID: 34404119 PMCID: PMC8907464 DOI: 10.4266/acc.2021.00178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/06/2021] [Indexed: 11/30/2022] Open
Abstract
Background Malnutrition is a serious condition in critically ill patients. The aim of this study is to evaluate the relationships between the Onodera’s prognostic nutritional index (OPNI) and intestinal permeability and between OPNI and systemic inflammation in critically ill patients. Methods This was a cross-sectional study conducted in the general intensive care unit (ICU) of a university-affiliated hospital. A total of 162 patients admitted between May 2018 and December 2019, was included in the study. The OPNI was calculated at admission and categorized as ≤40 or >40. We assessed plasma endotoxin and zonulin concentrations as markers of intestinal permeability as well as serum interleukin-6 (IL-6) and high-sensitivity C-reactive protein (hs-CRP) as markers of systemic inflammation upon admission under stringent conditions. The relationships between these markers and OPNI were assessed after adjusting for potential confounders through estimation of a binary logistic regression model. Results Median (interquartile range) hs-CRP, IL-6 zonulin, and endotoxin were significantly greater in the low OPNI subgroup than in the high OPNI subgroup (all P<0.05). Multivariate analyses showed significant association between serum IL-6 (odds ratio [OR], 0.88; 95% confidence interval [CI], 0.64–0.96), serum hs-CRP (OR, 0.77; 95% CI, 0.53–0.92), plasma endotoxin (OR, 0.81; 95% CI, 0.72–0.93), and plasma zonulin (OR, 0.83; 95% CI, 0.75–0.98) levels with OPNI in the overall population. Conclusions Our results provide evidence that higher plasma endotoxin, zonulin, IL-6, and hs-CRP levels are associated with progressively lower OPNI in mixed ICU populations, particularly in surgical ICU patients.
Collapse
Affiliation(s)
- Seyed Hossein Ardehali
- Department of Anesthesiology and Critical Care, Shohadaye Tajrish Hospital, Tehran,Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Eslamian
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Malek
- Department of Nutrition and Food Science, California State University, Chico, CA, USA
| |
Collapse
|
31
|
Probiotic Bacterial Application in Pediatric Critical Illness as Coadjuvants of Therapy. ACTA ACUST UNITED AC 2021; 57:medicina57080781. [PMID: 34440989 PMCID: PMC8399162 DOI: 10.3390/medicina57080781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022]
Abstract
The use of probiotics in critically ill adult and children patients has been growing exponentially over the last 20 years. Numerous factors in pediatriac intensive care unit (PICU) patients may contribute to intestinal dysbiosis, which subsequently promotes the pathobiota's growth. Currently, lactobacillus and bifidobacterium species are mainly used to prevent the development of systemic diseases due to the subverted microbiome, followed by streptococcus, enterococcus, propionibacterium, bacillus and Escherichia coli, Lactobacillus rhamnosus GG, and Lactobacillus reuteri DSM 17938. The aim of this article is to review the scientific literature for further confirmation of the importance of the usage of probiotics in intensive care unit (ICU) patients, especially in the pediatric population. A progressive increase in nosocomial infections, especially nosocomial bloodstream infections, has been observed over the last 30 years. The World Health Organization (WHO) reported that the incidence of nosocomial infections in PICUs was still high and ranged between 5% and 10%. Petrof et al. was one of the first to demonstrate the efficacy of probiotics for preventing systemic diseases in ICU patients. Recently, however, the use of probiotics with different lactobacillus spp. has been shown to cause a decrease of pro-inflammatory cytokines and an increase in anti-inflammatory cytokines. In addition, in some studies, the use of probiotics, in particular the mix of Lactobacillus and Bifidobacterium reduces the incidence of ventilator-associated pneumonia (VAP) in PICU patients requiring mechanical ventilation. In abdominal infections, there is no doubt at all about the usefulness of using Lactobacillus spp probiotics, which help to treat ICU-acquired diarrhoea episodes as well as in positive blood culture for candida spp. Despite the importance of using probiotics being supported by various studies, their use is not yet part of the standard protocols to which all doctors must adhere. In the meantime, while waiting for protocols to be drawn up as soon as possible for use in PICUs, routine use could certainly stimulate the intestine's immune defences. Though it is still too early to say, they could be considered the drugs of the future.
Collapse
|
32
|
Wang C, Li Q, Tang C, Zhao X, He Q, Tang X, Ren J. Characterization of the blood and neutrophil-specific microbiomes and exploration of potential bacterial biomarkers for sepsis in surgical patients. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1343-1357. [PMID: 34288545 PMCID: PMC8589375 DOI: 10.1002/iid3.483] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/12/2021] [Accepted: 06/21/2021] [Indexed: 12/29/2022]
Abstract
Introduction Recent studies have demonstrated the presence of a circulating microbiome in the blood of healthy subjects and chronic inflammatory patients. However, our knowledge regarding the blood microbiome and its potential roles in surgical patients remains very limited. The objective of this study was to determine the blood microbial landscape in surgical patients and to explore its potential associations with postoperative sepsis. Materials and Methods 2825 patients who underwent surgical treatments were screened for enrollment and 204 cases were recruited in this study. The patients were sub‐grouped into noninfected, infected, sepsis, and septic shock according to postoperative clinical manifestations. A total of 222 blood samples were obtained for neutrophil isolation, DNA extraction and high‐throughput sequencing, quantitative proteomics analysis, and flow cytometric analyses. Results Blood and neutrophils in surgical patients and healthy controls contained highly diverse microbiomes, mainly comprising Proteobacteria, Actinobacteria, Firmicutes and Bacteroidetes. The majority (80.7%–91.5%) of the microbiomes were composed of gut‐associated bacteria. The microbiomes in septic patients were significantly distinct from those of healthy controls, and marked differences in microbiome composition were observed between sepsis and septic shock groups. Several specific bacterial genera, including Flavobacterium, Agrococcus, Polynucleobacter, and Acidovorax, could distinguish patients with septic shock from those with sepsis, with higher area under curve values. Moreover, Agrococcus, Polynucleobacter, and Acidovorax were positively associated with the sequential (sepsis‐related) organ failure assessment scores and/or acute physiology and chronic health examination scores in septic shock patients. The proteins involved in bactericidal activities of neutrophils were downregulated in septic patients. Conclusions We present evidence identifying significant changes of blood and neutrophil‐specific microbiomes across various stages of sepsis, which might be associated with the progression of sepsis after surgical treatments. Several certain bacterial genera in blood microbiome could have potential as microbial markers for early detection of sepsis.
Collapse
Affiliation(s)
- Chenyang Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chun Tang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaofan Zhao
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qin He
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingming Tang
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
33
|
Qiang W, Xuan H, Yu S, Hailun P, Yueli Z, Zhiguo P, Lei S. Impact of the gut microbiota on heat stroke rat mediated by Xuebijing metabolism. Microb Pathog 2021; 155:104861. [PMID: 33864878 DOI: 10.1016/j.micpath.2021.104861] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/17/2022]
Abstract
The goal of the present study was to evaluate the fecal microbiome and serum metabolites in Xuebijing (XBJ)-injected rats after heat stroke using 16S rRNA gene sequencing and gas chromatography-mass spectrometry (GC-MS) metabolomics. Eighteen rats were divided into the control group (CON), heat stroke group (HS), and XBJ group. The 16S rRNA gene sequencing results revealed that the abundance of Bacteroidetes was overrepresented in the XBJ group compared to the HS group, while Actinobacteria was underrepresented. Metabolomic profiling showed that the pyrimidine metabolism pathway, pentose phosphate pathway, and glycerophospholipid metabolism pathway were upregulated in the XBJ group compared to the HS group. Taken together, these results demonstrated that heat stroke not only altered the gut microbiome community structure of rats but also greatly affected metabolic functions, leading to gut microbiome toxicity.
Collapse
Affiliation(s)
- Wen Qiang
- The First Clinical Medical College, Southern Medical University, Guangzhou, China; Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - He Xuan
- Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China
| | - Shao Yu
- Second Department of Internal Medicine for Cadres, General Hospital of Guangzhou Military Command, China
| | - Peng Hailun
- The First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zhao Yueli
- Graduate School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Pan Zhiguo
- The First Clinical Medical College, Southern Medical University, Guangzhou, China; Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China.
| | - Su Lei
- The First Clinical Medical College, Southern Medical University, Guangzhou, China; Department of Critical Care Medicine, General Hospital of Guangzhou Military Command, Guangzhou, China.
| |
Collapse
|
34
|
Zhuang X, Chen F, Zhou Q, Zhu Y, Yang X. A rapid preliminary prediction model for intestinal necrosis in acute mesenteric ischemia: a retrospective study. BMC Gastroenterol 2021; 21:154. [PMID: 33827660 PMCID: PMC8028195 DOI: 10.1186/s12876-021-01746-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Acute mesenteric ischemia (AMI) is a life-threatening condition. However, there is no accurate method to predict intestinal necrosis in AMI patients that may facilitate early surgical intervention. This study thus aimed to explore a simple and accurate model to predict intestinal necrosis in patients with AMI. METHODS A single-center retrospective study was performed on the data of 132 AMI patients treated between October 2011 and June 2020. The patients were divided into the intestinal necrosis and non-intestinal necrosis groups. The clinical characteristics and laboratory data were analyzed by univariate analysis, and the variables with statistical significance were further analyzed by multivariate logistic regression analysis. The independent predictors of intestinal necrosis were determined and a logistic prediction model was established. Finally, the accuracy, sensitivity, and specificity of the model in predicting intestinal necrosis were evaluated. RESULTS Univariate analysis showed that white blood cell (WBC) count, blood urea nitrogen (BUN) level, neutrophil ratio, prothrombin time (PT), and LnD-dimer were associated with intestinal necrosis. According to logistic regression multivariate analysis, WBC count, BUN level and LnD-dimer were independent predictors of intestinal necrosis. These parameters were used to establish a clinical prediction model of intestinal necrosis (CPMIN) as follows: model score = 0.349 × BUN (mmol/L) + 0.109 × WBC × 109 (109/L) + 0.394 × LnD - Dimer (ug/L) - 7.883. The area under the receiver operating characteristics (ROC) curve of the model was 0.889 (95% confidence interval: 0.833-0.944). Model scores greater than - 0.1992 predicted the onset of intestinal necrosis. The accuracy, specificity, and sensitivity of the model were 82.6%, 78.2%, and 88.3%, respectively. The proportion of intestinal necrosis in the high-risk patient group (CPMIN score ≥ - 0.1992) was much greater than that in the low-risk patient group (CPMIN score < - 0.1992; 82.7% vs. 15.0%, p < 0.001). CONCLUSION The CPMIN can effectively predict intestinal necrosis and guide early surgical intervention to improve patient prognosis. Patients with AMI who are classified as high-risk should be promptly treated with surgery to avoid the potential complications caused by delayed operation. Patients classified as low-risk group can receive non-surgical treatment. This model may help to lower the morbidity and mortality from AMI. However, this model's accuracy should be validated by larger sample size studies in the future.
Collapse
Affiliation(s)
- Xinsuo Zhuang
- Emergency and Trauma Center, The International Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 1367 West Wenyi Rd.Zhejiang Province, Hangzhou, 310058 China
| | - Fumei Chen
- Emergency and Trauma Center, The International Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 1367 West Wenyi Rd.Zhejiang Province, Hangzhou, 310058 China
| | - Qian Zhou
- Emergency and Trauma Center, The International Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 1367 West Wenyi Rd.Zhejiang Province, Hangzhou, 310058 China
| | - Yuanrun Zhu
- Emergency and Trauma Center, The International Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 1367 West Wenyi Rd.Zhejiang Province, Hangzhou, 310058 China
| | - Xiaofeng Yang
- Emergency and Trauma Center, The International Medical Center, The First Affiliated Hospital, Zhejiang University School of Medicine, 1367 West Wenyi Rd.Zhejiang Province, Hangzhou, 310058 China
| |
Collapse
|
35
|
Mustansir Dawoodbhoy F, Patel BK, Patel K, Bhatia M, Lee CN, Moochhala SM. Gut Microbiota Dysbiosis as a Target for Improved Post-Surgical Outcomes and Improved Patient Care: A Review of Current Literature. Shock 2021; 55:441-454. [PMID: 32881759 DOI: 10.1097/shk.0000000000001654] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Critical illness results in significant changes in the human gut microbiota, leading to the breakdown of the intestinal barrier function, which plays a role in the pathogenesis of multiple organ dysfunction. Patients with sepsis/acute respiratory distress syndrome (ARDS) have a profoundly distorted intestinal microbiota rhythm, which plays a considerable role in the development of gut-derived infections and intestinal dysbiosis. Despite recent medical developments, postsurgical complications are associated with a high morbidity and mortality rate. Bacterial translocation, which is the movement of bacteria and bacterial products across the intestinal barrier, was shown to be a mechanism behind sepsis. Current research is focusing on a solution by addressing significant factors that contribute to intestinal dysbiosis, which subsequently leads to multiple organ failure and, thus, mortality. It may, however, be challenging to manipulate the microbiota in critically ill patients for enhanced therapeutic gain. Probiotic manipulation is advantageous for maintaining the gut-barrier defense and for modulating the immune response. Based on available published research, this review aims to address the application of potential strategies in the intensive care unit, supplemented with current therapeutics by the administration of probiotics, prebiotics, and fecal microbiota transplant, to reduce post-surgical complications of sepsis/ARDS in critically ill patients.
Collapse
Affiliation(s)
| | | | - Kadamb Patel
- School of Applied Sciences, Temasek Polytechnic, Singapore
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago Christchurch, Christchurch, New Zealand
| | - Chuen Neng Lee
- Department of Surgery, National University of Singapore, Singapore
| | | |
Collapse
|
36
|
Ala-Houhala M, Anttila VJ. Characteristics of late recurrent candidemia in adult patients. Mycoses 2021; 64:503-510. [PMID: 33377571 DOI: 10.1111/myc.13236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES Candida species are one of the most common causes of health care-associated bloodstream infections. However, recurrent candidemia is rare, and the characteristics of late recurrent (LR) candidemia are partly unclear. Our aim was to evaluate the characteristics of LR candidemia in adult patients. PATIENTS AND METHODS A retrospective cohort study was performed in the hospital district of Helsinki and Uusimaa in Finland (2007-2016). All candidemia cases were searched in an electronic database during the study period. Patients with LR candidemia were compared with patients with a single candidemia episode to evaluate the characteristics of LR candidemia. LR candidemia was defined as having at least two episodes of candidemia more than 30 days apart. RESULTS We identified 24 episodes of LR candidemia in 20 patients. Patients with LR candidemia represented 6% of all patients with candidemia during the study period, and most of these cases were nosocomial. The median time between the first and the recurrent episode was 5.1 months. One-year mortality in LR candidemia was 45%. Underlying gastrointestinal disease (OR 7.21, 95% CI 2.52-20.61) and history of intra-venous drug use (IVDU) (OR 3.62, 95% CI 1.03-12.69) were independent risk factors for LR candidemia in the multivariable analysis. CONCLUSION Our study indicates that the gastrointestinal tract may be a continuous source of infection in patients with chronic gastrointestinal diseases. Gastrointestinal diseases and IVDU should be regarded as risk factors for LR candidemia.
Collapse
Affiliation(s)
- Mari Ala-Houhala
- Inflammation Center, Division of Infectious Diseases, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Veli-Jukka Anttila
- Inflammation Center, Division of Infectious Diseases, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
37
|
Angurana S, Mehta A. Probiotics in critically ill children: An updated review. JOURNAL OF PEDIATRIC CRITICAL CARE 2021. [DOI: 10.4103/jpcc.jpcc_73_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
38
|
Lee DU, Fan GH, Hastie DJ, Prakasam VN, Addonizio EA, Ahern RR, Seog KJ, Karagozian R. The Clinical Impact of Cirrhosis on the Hospital Outcomes of Patients Admitted With Influenza Infection: Propensity Score Matched Analysis of 2011-2017 US Hospital Data. J Clin Exp Hepatol 2021; 11:531-543. [PMID: 34511813 PMCID: PMC8414330 DOI: 10.1016/j.jceh.2021.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND/OBJECTIVES Patients with cirrhosis have liver-related immune dysfunction that potentially predisposes the patients to increased influenza infection risk. Our study evaluates this cross-sectional relationship using a national registry of hospital patients. METHODS This study included the 2011-2017 National Inpatient Sample database. From this, respiratory influenza cases were isolated and stratified using the presence of cirrhosis into a cirrhosis-present study cohort and cirrhosis-absent controls; propensity score matching method was used to match the controls to the study cohort (cirrhosis-present) using a 1:1 matching ratio. The endpoints included mortality, length of stay, hospitalization costs, and influenza-related complications. RESULTS Following the match, there were 2,040 with cirrhosis and matched 2,040 without cirrhosis admitted with respiratory influenza infection. Compared to the controls, cirrhosis patients had higher in-hospital mortality (7.79 vs 3.43% p < 0.001, OR 2.38 95% CI 1.78-3.17), longer length of stay (7.25 vs 6.52 d p < 0.001), higher hospitalization costs ($70,009 vs $65,035 p < 0.001), and were more likely be discharged to a skilled nursing facility and home healthcare (vs routine home discharges). In terms of influenza-related complications, the cirrhosis cohort had higher rates of sepsis (29.8 vs 22% p < 0.001, OR 1.51 95% CI 1.31-1.74). In the multivariate regression analysis, cirrhosis was associated with higher mortality (p < 0.001, aOR 2.31 95% CI 1.59-3.35) and length of stay (p = 0.018, aOR 1.03 95% CI 1.01-1.06). In subgroup analysis of patients with decompensated (n = 597) versus compensated cirrhosis (n = 1443), those with decompensated cirrhosis had higher rates of in-hospital mortality (12.7 vs 5.75% p < 0.001, OR 2.39 95% CI 1.72-3.32), length of stay (8.85 vs 6.59 d p < 0.001), and hospitalization costs ($92,858 vs $60,556 p < 0.001). In the multivariate analysis, decompensated cirrhosis was associated with increased mortality (p < 0.001, aOR 2.86 95% CI 1.90-4.32). CONCLUSION This study shows the presence of cirrhosis to result in higher hospital mortality and postinfluenza complications in patients with influenza infection.
Collapse
Key Words
- AHRQ, agency for healthcare research and quality
- DRG, diagnosis-related group
- HCUP, healthcare cost and utilization project
- ICD-10, international classification of diseases, tenth edition
- ICD-9, international classification of diseases, ninth edition
- NIS, nationwide inpatient sample
- SBP, spontaneous bacterial peritonitis
- SID, state inpatient database
- VIF, variation inflation factor
- ascites
- common cold
- flu
- influenza-related complications
- portal hypertension
Collapse
Affiliation(s)
- David U. Lee
- Address for correspondence: David Uihwan Lee MD, Liver Center, Division of Gastroenterology, Tufts Medical Center, 800 Washington Street, Boston, MA, 02111, USA. T: 617-636-4168, F: 617-636-9292.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Scott RA, Williams HG, Hoad CL, Alyami A, Ortori CA, Grove JI, Marciani L, Moran GW, Spiller RC, Menys A, Aithal GP, Gowland PA. MR Measures of Small Bowel Wall
T2
Are Associated With Increased Permeability. J Magn Reson Imaging 2020; 53:1422-1431. [DOI: 10.1002/jmri.27463] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Robert A. Scott
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Nottingham Digestive Diseases Centre University of Nottingham Nottingham UK
| | - Hannah G. Williams
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy University of Nottingham Nottingham UK
| | - Caroline L. Hoad
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy University of Nottingham Nottingham UK
| | - Ali Alyami
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
| | - Catherine A. Ortori
- Centre for Analytical Bioscience, School of Pharmacy University of Nottingham Nottingham UK
| | - Jane I. Grove
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Nottingham Digestive Diseases Centre University of Nottingham Nottingham UK
| | - Luca Marciani
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Nottingham Digestive Diseases Centre University of Nottingham Nottingham UK
| | - Gordon W. Moran
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Nottingham Digestive Diseases Centre University of Nottingham Nottingham UK
| | - Robin C. Spiller
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Nottingham Digestive Diseases Centre University of Nottingham Nottingham UK
| | | | - Guruprasad P. Aithal
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Nottingham Digestive Diseases Centre University of Nottingham Nottingham UK
| | - Penny A. Gowland
- National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre at the Nottingham University Hospitals NHS Trust and University of Nottingham Nottingham UK
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy University of Nottingham Nottingham UK
| |
Collapse
|
40
|
Niu X, Shang H, Chen S, Chen R, Huang J, Miao Y, Cui W, Wang H, Sha Z, Peng D, Zhu R. Effects of Pinus massoniana pollen polysaccharides on intestinal microenvironment and colitis in mice. Food Funct 2020; 12:252-266. [PMID: 33295902 DOI: 10.1039/d0fo02190c] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The stability of the intestinal microenvironment is the basis for maintaining the normal physiological activities of the intestine. On the contrary, disordered dynamic processes lead to chronic inflammation and disease pathology. Pinus massoniana pollen polysaccharide (PPPS), isolated from Taishan Pinus massoniana pollen, has been reported with extensive biological activities, including immune regulation. However, the role of PPPS in the intestinal microenvironment and intestinal diseases is still unknown. In this work, we initiated our investigation by using 16S rRNA high-throughput sequencing technology to assess the effect of PPPS on gut microbiota in mice. The result showed that PPPS regulated the composition of gut microbiota in mice and increased the proportion of probiotics. Subsequently, we established immunosuppressive mice using cyclophosphamide (CTX) and found that PPPS regulated the immunosuppressive state of lymphocytes in Peyer's patches (PPs). Moreover, PPPS also regulated systemic immunity by acting on intestinal PPs. PPPS alleviated lipopolysaccharide (LPS) -induced Caco2 cell damage, indicating that PPPS has the ability to reduce the damage and effectively improve the barrier dysfunction in Caco2 cells. In addition, PPPS alleviated colonic injury and relieved colitis symptoms in dextran sodium sulfate (DSS)-induced colitis mice. Overall, our findings indicate that PPPS shows a practical regulatory effect in the intestinal microenvironment, which provides an essential theoretical basis for us to develop the potential application value of PPPS further.
Collapse
Affiliation(s)
- Xiangyun Niu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Hongqi Shang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Siyan Chen
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Ruichang Chen
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Jin Huang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Yongqiang Miao
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Wenping Cui
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Huan Wang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Zhou Sha
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| | - Duo Peng
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| | - Ruiliang Zhu
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an, China. and Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, Tai'an, China
| |
Collapse
|
41
|
Desvaux M, Dalmasso G, Beyrouthy R, Barnich N, Delmas J, Bonnet R. Pathogenicity Factors of Genomic Islands in Intestinal and Extraintestinal Escherichia coli. Front Microbiol 2020; 11:2065. [PMID: 33101219 PMCID: PMC7545054 DOI: 10.3389/fmicb.2020.02065] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022] Open
Abstract
Escherichia coli is a versatile bacterial species that includes both harmless commensal strains and pathogenic strains found in the gastrointestinal tract in humans and warm-blooded animals. The growing amount of DNA sequence information generated in the era of "genomics" has helped to increase our understanding of the factors and mechanisms involved in the diversification of this bacterial species. The pathogenic side of E. coli that is afforded through horizontal transfers of genes encoding virulence factors enables this bacterium to become a highly diverse and adapted pathogen that is responsible for intestinal or extraintestinal diseases in humans and animals. Many of the accessory genes acquired by horizontal transfers form syntenic blocks and are recognized as genomic islands (GIs). These genomic regions contribute to the rapid evolution, diversification and adaptation of E. coli variants because they are frequently subject to rearrangements, excision and transfer, as well as to further acquisition of additional DNA. Here, we review a subgroup of GIs from E. coli termed pathogenicity islands (PAIs), a concept defined in the late 1980s by Jörg Hacker and colleagues in Werner Goebel's group at the University of Würzburg, Würzburg, Germany. As with other GIs, the PAIs comprise large genomic regions that differ from the rest of the genome by their G + C content, by their typical insertion within transfer RNA genes, and by their harboring of direct repeats (at their ends), integrase determinants, or other mobility loci. The hallmark of PAIs is their contribution to the emergence of virulent bacteria and to the development of intestinal and extraintestinal diseases. This review summarizes the current knowledge on the structure and functional features of PAIs, on PAI-encoded E. coli pathogenicity factors and on the role of PAIs in host-pathogen interactions.
Collapse
Affiliation(s)
- Mickaël Desvaux
- Université Clermont Auvergne, INRAE, MEDiS, Clermont-Ferrand, France
| | - Guillaume Dalmasso
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Racha Beyrouthy
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Nicolas Barnich
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Julien Delmas
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Richard Bonnet
- UMR Inserm 1071, USC-INRAE 2018, M2iSH, Université Clermont Auvergne, Clermont-Ferrand, France
- Laboratoire de Bactériologie, CHU Clermont-Ferrand, Clermont-Ferrand, France
| |
Collapse
|
42
|
Nakahori Y, Shimizu K, Ogura H, Asahara T, Osuka A, Yamano S, Tasaki O, Kuwagata Y, Shimazu T. Impact of fecal short-chain fatty acids on prognosis in critically ill patients. Acute Med Surg 2020; 7:e558. [PMID: 32864147 PMCID: PMC7447560 DOI: 10.1002/ams2.558] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 11/24/2022] Open
Abstract
Aim This study aimed to evaluate the relationship between fecal organic acids and mortality in critically ill patients. Methods This retrospective study included 128 patients who fulfilled the criteria of systemic inflammatory response syndrome and had a serum C‐reactive protein level of greater than 10 mg/dL. Patients were treated in the intensive care unit for more than 2 days. Patients were divided into two groups: survivors and non‐survivors. We measured and compared eight kinds of fecal organic acids between the two groups. We focused on the minimum and maximum value of each fecal organic acid and evaluated prognostic factors by using classification and regression tree (CART) and multivariate logistic regression analyses. Results We included 90 patients as survivors and 38 as non‐survivors. The CART analysis revealed that the dominant factors for mortality were the minimum values of propionate and acetate and the maximum values of lactate and formic acid. In the evaluation of the minimum values of fecal organic acids, propionate was significantly associated with increased mortality (odds ratio, 0.11 [95% confidence interval, 0.024–0.51]; P = 0.005), acetate (0.047 [0.005–0.49]; P = 0.01), and age (1.048 [1.015–1.083]; P = 0.004). In the evaluation of the maximum values, lactate was significantly associated with increased mortality (5.21 [2.024–13.42], P = 0.001) and age (1.050 [1.017–1.084]; P = 0.003). Conclusion An altered balance of fecal organic acids was significantly associated with mortality in critically ill patients.
Collapse
Affiliation(s)
- Yasutaka Nakahori
- Division of Trauma and Surgical Critical Care Osaka General Medical Center Osaka Japan.,Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Osaka Japan
| | - Kentaro Shimizu
- Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Osaka Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Osaka Japan
| | | | - Akinori Osuka
- Department of Trauma Critical Care Medicine and Burn Center Chukyo Hospital Aichi Japan
| | - Shuhei Yamano
- Acute and Critical Care Center Nagasaki University Hospital Nagasaki Japan
| | - Osamu Tasaki
- Acute and Critical Care Center Nagasaki University Hospital Nagasaki Japan
| | - Yasuyuki Kuwagata
- Department of Emergency and Critical Care Medicine Kansai Medical University Osaka Japan
| | - Takeshi Shimazu
- Department of Traumatology and Acute Critical Medicine Osaka University Graduate School of Medicine Osaka Japan
| |
Collapse
|
43
|
Nallanchakravarthula S, Amruta N, Ramamurthy C. Cancer Microbiome; Opportunities and Challenges. Endocr Metab Immune Disord Drug Targets 2020; 21:215-229. [PMID: 32819239 DOI: 10.2174/1871530320999200818134942] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/03/2020] [Accepted: 06/11/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Microbe-host association has emerged as a modulator in modern medicine. Cancer and its associated host microbes are collectively referred to as the cancer microbiome. The cancer microbiome is complex, and many aspects remain unclear including metabolic plasticity, microenvironment remodeling, cellular communications, and unique signatures within the host, all of which have a vital role in homeostasis and pathogenesis of host physiology. However, the role of the microbiome in cancer initiation, progression, and therapy is still poorly understood and remains to be explored. OBJECTIVE The objective of this review is to elucidate the role of the microbiome in cancer metabolism and the tumor microenvironment. It also focuses on the importance of therapeutic opportunities and challenges in the manipulation of the cancer microbiome. METHODS A literature search was conducted on the role of the microbiome in cancer initiation, progression, and therapy. CONCLUSION The tumor microenvironment and cancer metabolism are significant in host-microbiome interactions. The microbiome can modulate standard cancer therapies like chemotherapy and immunotherapy. Microbiome transplantation has also been demonstrated as an effective therapy against cancer. Furthermore, the modulation of the microbiome also has potential clinical outcomes in modern medicine.
Collapse
Affiliation(s)
| | - Narayanappa Amruta
- Department of Neurosurgery, Tulane University, New Orleans, Louisiana, United States
| | - Chitteti Ramamurthy
- C.G. Bhakta Institute of Biotechnology, UkaTarsadia University, Maliba campus, Bardoli Surat (Dist), Gujarat, India
| |
Collapse
|
44
|
Alagna L, Palomba E, Mangioni D, Bozzi G, Lombardi A, Ungaro R, Castelli V, Prati D, Vecchi M, Muscatello A, Bandera A, Gori A. Multidrug-Resistant Gram-Negative Bacteria Decolonization in Immunocompromised Patients: A Focus on Fecal Microbiota Transplantation. Int J Mol Sci 2020; 21:ijms21165619. [PMID: 32764526 PMCID: PMC7460658 DOI: 10.3390/ijms21165619] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Antimicrobial resistance is an important issue for global health; in immunocompromised patients, such as solid organ and hematological transplant recipients, it poses an even bigger threat. Colonization by multidrug-resistant (MDR) bacteria was acknowledged as a strong risk factor to subsequent infections, especially in individuals with a compromised immune system. A growing pile of studies has linked the imbalance caused by the dominance of certain taxa populating the gut, also known as intestinal microbiota dysbiosis, to an increased risk of MDR bacteria colonization. Several attempts were proposed to modulate the gut microbiota. Particularly, fecal microbiota transplantation (FMT) was successfully applied to treat conditions like Clostridioides difficile infection and other diseases linked to gut microbiota dysbiosis. In this review we aimed to provide a look at the data gathered so far on FMT, focusing on its possible role in treating MDR colonization in the setting of immunocompromised patients and analyzing its efficacy and safety.
Collapse
Affiliation(s)
- Laura Alagna
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Emanuele Palomba
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-3494073517
| | - Davide Mangioni
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20122 Milan, Italy
| | - Giorgio Bozzi
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Andrea Lombardi
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Riccardo Ungaro
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Valeria Castelli
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Maurizio Vecchi
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Antonio Muscatello
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
| | - Alessandra Bandera
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
| | - Andrea Gori
- Infectious Disease Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (L.A.); (D.M.); (G.B.); (A.L.); (R.U.); (V.C.); (A.M.); (A.B.); (A.G.)
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Centre for Multidisciplinary Research in Health Science, University of Milan, 20122 Milan, Italy
| |
Collapse
|
45
|
Adelman MW, Woodworth MH, Langelier C, Busch LM, Kempker JA, Kraft CS, Martin GS. The gut microbiome's role in the development, maintenance, and outcomes of sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:278. [PMID: 32487252 PMCID: PMC7266132 DOI: 10.1186/s13054-020-02989-1] [Citation(s) in RCA: 181] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/12/2020] [Indexed: 12/15/2022]
Abstract
The gut microbiome regulates a number of homeostatic mechanisms in the healthy host including immune function and gut barrier protection. Loss of normal gut microbial structure and function has been associated with diseases as diverse as Clostridioides difficile infection, asthma, and epilepsy. Recent evidence has also demonstrated a link between the gut microbiome and sepsis. In this review, we focus on three key areas of the interaction between the gut microbiome and sepsis. First, prior to sepsis onset, gut microbiome alteration increases sepsis susceptibility through several mechanisms, including (a) allowing for expansion of pathogenic intestinal bacteria, (b) priming the immune system for a robust pro-inflammatory response, and (c) decreasing production of beneficial microbial products such as short-chain fatty acids. Second, once sepsis is established, gut microbiome disruption worsens and increases susceptibility to end-organ dysfunction. Third, there is limited evidence that microbiome-based therapeutics, including probiotics and selective digestive decontamination, may decrease sepsis risk and improve sepsis outcomes in select patient populations, but concerns about safety have limited uptake. Case reports of a different microbiome-based therapy, fecal microbiota transplantation, have shown correlation with gut microbial structure restoration and decreased inflammatory response, but these results require further validation. While much of the evidence linking the gut microbiome and sepsis has been established in pre-clinical studies, clinical evidence is lacking in many areas. To address this, we outline a potential research agenda for further investigating the interaction between the gut microbiome and sepsis.
Collapse
Affiliation(s)
- Max W Adelman
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, GA, 30303, USA.
| | - Michael H Woodworth
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, GA, 30303, USA
| | - Charles Langelier
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Lindsay M Busch
- Critical Care Medicine Department, National Institutes of Health Clinical Center, Bethesda, MD, USA
| | - Jordan A Kempker
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Colleen S Kraft
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 49 Jesse Hill Jr. Drive, Atlanta, GA, 30303, USA.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Greg S Martin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Emory Critical Care Center, Emory Healthcare, Atlanta, GA, USA
| |
Collapse
|
46
|
Yang B, Zhang LY, Chen Y, Bai YP, Jia J, Feng JG, Liu KX, Zhou J. Melatonin alleviates intestinal injury, neuroinflammation and cognitive dysfunction caused by intestinal ischemia/reperfusion. Int Immunopharmacol 2020; 85:106596. [PMID: 32442902 DOI: 10.1016/j.intimp.2020.106596] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/29/2020] [Accepted: 05/10/2020] [Indexed: 12/31/2022]
Abstract
Intestinal ischemia/reperfusion (I/R) can cause multiple organ damage with extremely high morbidity and mortality. Melatonin has anti-inflammatory, anti-oxidative and anti-apoptotic effects against various diseases. This study aimed to explore whether melatonin had a protective effect against intestinal I/R-induced neuroinflammation and cognitive dysfunction, and investigate its potential mechanisms. In this study, melatonin was administered to the rats with intestinal I/R, then histological changes in intestine and brain (frontal cortex and hippocampal CA1 area) tissues and cognitive function were detected, respectively. The encephaledema and blood-brain barrier (BBB) permeability were observed. Moreover, the alterations of proinflammatory factors (tumor necrosis factor-α, interleukin-6 and interleukin-1β), oxidative response (malondialdehyde, superoxide dismutase, and reactive oxygen species), apoptosis and proteins associated with inflammation,including Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (Myd88) and phosphorylated nuclear factor kappa beta (NF-κB), and apoptosis (cleaved caspase-3) in brain tissues were examined. Furthermore, the expressions of TLR4, Myd88, and microglial activity were observed by multiple immunofluorescence staining. The results showed that intestinal I/R-induced abnormal neurobehavior and cerebral damage were ameliorated after melatonin treatment, which were demonstrated by improved cognitive dysfunction and aggravated histology. Furthermore, melatonin decreased the levels of proinflammatory factors and oxidative stress in plasma, intestine and brain tissues, attenuated apoptotic cell, and inhibited the expressions of related proteins and the immunoreactivity of TLR4 or Myd88 in microglia in brain tissues. These findings showed that melatonin might relieve neuroinflammation and cognitive dysfunction caused by intestinal I/R, which could be, at least partially, related to the inhibition of the TLR4/Myd88 signaling in microglia.
Collapse
Affiliation(s)
- Bo Yang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Li-Yin Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Yi-Ping Bai
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Jian-Guo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China
| | - Ke-Xuan Liu
- Department of Anesthesiology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, PR China.
| |
Collapse
|
47
|
Mangieri CW, Ling JA, Modlin DM, Rose ED, Burgess PL. Utilization of combination bowel preparation (CBP) is protective against the development of post-operative Clostridium difficile infection (CDI), decreases septic complications, and provides a survival benefit. Surg Endosc 2020; 35:928-933. [DOI: 10.1007/s00464-020-07563-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
|
48
|
Wang H, Zhang S, Zhao H, Qin H, Zhang J, Dong J, Zhang H, Liu X, Zhao Z, Zhao Y, Shao M, Wu F, Zhang W. Carbon Monoxide Inhibits the Expression of Proteins Associated with Intestinal Mucosal Pyroptosis in a Rat Model of Sepsis Induced by Cecal Ligation and Puncture. Med Sci Monit 2020; 26:e920668. [PMID: 32351244 PMCID: PMC7207005 DOI: 10.12659/msm.920668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 01/20/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Carbon monoxide (CO) has anti-inflammatory effects and protects the intestinal mucosal barrier in sepsis. Pyroptosis, or cell death associated with sepsis, is mediated by caspase-1 activation. This study aimed to investigate the role of CO on the expression of proteins associated with intestinal mucosal pyroptosis in a rat model of sepsis induced by cecal ligation and puncture (CLP). MATERIAL AND METHODS The rat model of sepsis was developed using CLP. Male Sprague-Dawley rats (n=120) were divided into six study groups: the sham group (n=20); the CLP group (n=20); the hemin group (treated with ferric chloride and heme) (n=20); the zinc protoporphyrin IX (ZnPPIX) group (n=20); the CO-releasing molecule 2 (CORM-2) group (n=20); and the inactive CORM-2 (iCORM-2) group (n=20). Hemin and CORM-2 were CO donors, and ZnPPIX was a CO inhibitor. In the six groups, the seven-day survival curves, the fluorescein isothiocyanate (FITC)-labeled dextran 4000 Da (FD-4) permeability assay, levels of intestinal pyroptosis proteins caspase-1, caspase-11, and gasdermin D (GSDMD) were measured by confocal fluorescence microscopy. Proinflammatory cytokines interleukin (IL)-18, IL-1ß, and high mobility group box protein 1 (HMGB1) were measured by Western blot and enzyme-linked immunosorbent assay (ELISA). RESULTS CO reduced the mortality rate in rats with sepsis and reduced intestinal mucosal permeability and mucosal damage. CO also reduced the expression levels of IL-18, IL-1ß, and HMGB1, and reduced pyroptosis by preventing the cleavage of caspase-1 and caspase-11. CONCLUSIONS In a rat model of sepsis induced by CLP, CO had a protective role by inhibiting intestinal mucosal pyroptosis.
Collapse
Affiliation(s)
- Hongzhou Wang
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| | - Shunwen Zhang
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Haijun Zhao
- The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Huiyuan Qin
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jie Zhang
- The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Jiangtao Dong
- The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Hui Zhang
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| | - Xiaoling Liu
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| | - Zhengyong Zhao
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| | - Yanheng Zhao
- The First Affiliated Hospital of the Medical College, Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Meng Shao
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| | - Fang Wu
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| | - Wanjiang Zhang
- Department of Pathophysiology, Shihezi University School of Medicine and The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi, Xinjiang, P.R. China
| |
Collapse
|
49
|
Lipocalin2 Induced by Bacterial Flagellin Protects Mice against Cyclophosphamide Mediated Neutropenic Sepsis. Microorganisms 2020; 8:microorganisms8050646. [PMID: 32365611 PMCID: PMC7284693 DOI: 10.3390/microorganisms8050646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022] Open
Abstract
Neutropenic sepsis is a fatal consequence of chemotherapy, and septic complications are the principal cause of mortality. Chemotherapy-induced neutropenia leads to the formation of microscopic ulcers in the gastrointestinal epithelium that function as a portal of entry for intraluminal bacteria, which translocate across the intestinal mucosal barrier and gain access to systemic sites, causing septicemia. A cyclophosphamide-induced mouse model was developed to mimic the pathophysiologic sequence of events that occurs in patients with neutropenic sepsis. The TLR5 agonist bacterial flagellin derived from Vibrio vulnificus extended the survival of cyclophosphamide-treated mice by reducing the bacterial load in internal organs. The protective effect of flagellin was mediated by the antimicrobial protein lipocalin 2 (Lcn2), which is induced by TLR5-NF-κB activation in hepatocytes. Lcn2 sequestered iron from infecting bacteria, particularly siderophore enterobactin-dependent members of the Enterobacteriaceae family, thereby limiting their proliferation. Lcn2 should be considered for the treatment of neutropenic sepsis and gastrointestinal damage during chemotherapy to prevent or minimize the adverse effects of cancer chemotherapy.
Collapse
|
50
|
Probiotics Used for Postoperative Infections in Patients Undergoing Colorectal Cancer Surgery. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5734718. [PMID: 32076609 PMCID: PMC7019203 DOI: 10.1155/2020/5734718] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/31/2019] [Indexed: 01/09/2023]
Abstract
Objective The objective of this study was to conduct a systematic review and meta-analysis about probiotics to improve postoperative infections in patients undergoing colorectal cancer surgery. Methods The PubMed and the Web of Science were used to search for appropriate randomized clinical trials (RCTs) comparing probiotics with placebo for the patients undergoing colorectal cancer surgery. The RevMan 5.3 was performed for meta-analysis to evaluate the postoperative infection, including the total infection, surgical site infection, central line infection, pneumonia, urinary tract infection, septicemia, and postoperative leakage. Results Our meta-analysis included 6 studies involving a total of 803 patients. For the incidence of total postoperative infection (odd ratios (OR) 0.31, 95% confidence interval (CI) 0.15–0.64, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, I2 = 0%), surgical site infection (OR 0.62, 95% CI 0.39–0.99, Conclusions Probiotics is beneficial to prevent postoperative infections (including total postoperative infection, surgical site infection, pneumonia, urinary tract infection, and septicemia) in patients with colorectal cancer.
Collapse
|