1
|
Zhao J, Guo Y, Jiang Q, Lan H, Hung WL, Lynch B. Bifidobacterium longum subsp. infantis YLGB-1496-Toxicological evaluation. J Appl Toxicol 2025; 45:230-244. [PMID: 39252460 DOI: 10.1002/jat.4688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/11/2024]
Abstract
Bifidobacterium infantis YLGB-1496, originally isolated from breast milk from a Taiwanese mother, is under study for use as a probiotic. As part of safety assessment, an Ames, in vivo mouse micronucleus, and in vivo mouse spermatocyte chromosome aberration assay were conducted along with a 13-week oral rat toxicity study. B. infantis YLGB-1496 had no activity in any of the genotoxicity assays. Administration of the bacteria to Sprague-Dawley rats at doses ranging from 0 to 1.5 g/kg bw/day had no treatment-related effects on any of the endpoints measured. There appear to be no concerns for translocation or pathogenicity of B. infantis YLGB-1496 based on extensive experience with the species in general. The results of the current investigations support potential use of B. infantis YLGB-1496 as a probiotic in infant formula.
Collapse
Affiliation(s)
- Jian Zhao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yueyi Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qiuyue Jiang
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Wei-Lian Hung
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Barry Lynch
- Intertek Health Sciences Inc., Mississauga, ON, Canada
| |
Collapse
|
2
|
Scanlan PD, Baquero F, Levin BR. Short-sighted evolution of virulence for invasive gut microbes: From hypothesis to tests. Proc Natl Acad Sci U S A 2024; 121:e2409905121. [PMID: 39570365 PMCID: PMC11626195 DOI: 10.1073/pnas.2409905121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024] Open
Abstract
Why microbes harm their hosts is a fundamental question in evolutionary biology with broad relevance to our understanding of infectious diseases. Several hypotheses have been proposed to explain this "evolution of virulence." In this perspective, we reexamine one of these hypotheses in the specific context of the human gut microbiome, namely short-sighted evolution. According to the short-sighted evolution hypothesis, virulence is a product of niche expansion within a colonized host, whereby variants of commensal microbes establish populations in tissues and sites where the infection causes morbidity or mortality. This evolution is short-sighted in that the evolved variants that infect those tissues and sites are not transmitted to other hosts. The specific hypothesis that we propose is that some bacteria responsible for invasive infections and disease are the products of the short-sighted evolution of commensal bacteria residing in the gut microbiota. We present observations in support of this hypothesis and discuss the challenges inherent in assessing its general application to infections and diseases associated with specific members of the gut microbiota. We then describe how this hypothesis can be tested using genomic data and animal model experiments and outline how such studies will serve to provide fundamental information about both the evolution and genetic basis of virulence, and the bacteria of intensively studied yet poorly understood habitats including the gut microbiomes of humans and other mammals.
Collapse
Affiliation(s)
- Pauline D. Scanlan
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- School of Microbiology, University College Cork, CorkT12 Y337, Ireland
| | - Fernando Baquero
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid28034, Spain
- Centro de Investigación Médica en Red, Epidemiología y Salud Pública, Madrid28007, Spain
| | - Bruce R. Levin
- Department of Biology, Emory University, Atlanta, GA30322
| |
Collapse
|
3
|
Kubota Y, Hiu H, Masuda Y, Nakamichi C. Concurrent Bacteremia and Severe Fever With Thrombocytopenia Syndrome: A Report of Two Cases. Cureus 2024; 16:e75408. [PMID: 39781123 PMCID: PMC11710864 DOI: 10.7759/cureus.75408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2024] [Indexed: 01/12/2025] Open
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) is a tick-borne viral hemorrhagic fever caused by the severe fever with thrombocytopenia syndrome virus (SFTSV). This virus, which is transmitted through ticks, is prevalent in Asian countries, including Japan. This report describes two rare cases of SFTS with concurrent bacteremia. In the first case, a 73-year-old Japanese man presented with fever and coagulopathy and subsequently developed oral mucosal hemorrhage and bloody stools. Initial tests showed thrombocytopenia and leukopenia. Blood cultures on admission revealed Enterococcus faecalis bacteremia, and SFTSV co-infection was subsequently confirmed by polymerase chain reaction (PCR). He was treated with vancomycin, followed by ampicillin, and recovered. In the second case, a 70-year-old Japanese man presented with fever and diarrhea and subsequently developed renal impairment and thrombocytopenia. Blood cultures on admission revealed Escherichia coli bacteremia, and SFTSV co-infection was subsequently confirmed by PCR. He was treated with ceftriaxone, followed by ampicillin, and recovered. We conducted a literature search to look for evidence of a link between SFTS and bacteremia. Further epidemiological studies on the association between SFTS and bacteremia are warranted to provide an understanding of the pathogenic mechanisms.
Collapse
Affiliation(s)
- Yoshifumi Kubota
- Department of Emergency and Critical Care Medicine, National Hospital Organization Nagasaki Medical Center, Nagasaki, JPN
| | - Hiroyuki Hiu
- Department of Emergency and Critical Care Medicine, National Hospital Organization Nagasaki Medical Center, Nagasaki, JPN
| | - Yukiko Masuda
- Department of Emergency and Critical Care Medicine, National Hospital Organization Nagasaki Medical Center, Nagasaki, JPN
| | - Chikaaki Nakamichi
- Department of Emergency and Critical Care Medicine, National Hospital Organization Nagasaki Medical Center, Nagasaki, JPN
| |
Collapse
|
4
|
Asgari B, Burke JR, Quigley BL, Bradford G, Hatje E, Kuballa A, Katouli M. Identification of Virulence Genes Associated with Pathogenicity of Translocating Escherichia coli with Special Reference to the Type 6 Secretion System. Microorganisms 2024; 12:1851. [PMID: 39338525 PMCID: PMC11433802 DOI: 10.3390/microorganisms12091851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Recent genomic characterisation of translocating Escherichia coli HMLN-1 isolated from mesenteric lymph nodes (MLNs) and blood of a patient with a fatal case of pancreatitis revealed the presence of a type 6 secretion system (T6SS) that was not present in non-translocating E. coli strains. This strain was also genomically similar to adherent-invasive E. coli (AIEC) LF82 pathotype. We aimed to identify the role of T6SS-1 in the pathogenesis of this strain and other pathogenic E. coli. The HMLN-1 strain was initially tested for the presence of six virulence genes (VGs) associated with AIEC strains and an iron sequestering system. Additionally, HMLN-1's interaction with a co-culture of Caco-2:HT29-MTX cells and its intra-macrophagic survival was evaluated. We subsequently screened a collection of 319 pathogenic E. coli strains isolated from patients with urinary tract infection (UTI), diarrhoea, inflammatory bowel disease (IBD) and septicaemia for the presence of T6SS-1 and its expression related to adhesion, invasion and translocation via the above co-culture of the intestinal cell lines. The results showed that HMLN-1 harboured four of the AIEC-associated VGs (dsbA, htrA, ompC and afaC). Screening of the pathogenic E. coli collection detected the presence of the T6SS-1 genes in septicaemic and UTI E. coli strains at a significantly higher level than diarrhoea and IBD strains (p < 0.0001). The high expression of T6SS-1 in E. coli HMLN-1 upon adhesion and invasion, as well as its high prevalence among extra-intestinal E. coli strains, suggests a role for T6SS-1 in the pathogenesis of translocating E. coli.
Collapse
Affiliation(s)
- Behnoush Asgari
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
| | - Jarred R. Burke
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
- Servatus Biopharmaceuticals, Coolum Beach, QLD 4573, Australia
| | - Bonnie L. Quigley
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
- Thompson Institute, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia
| | - Georgia Bradford
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
| | - Eva Hatje
- Centre for Immunology and Infection Control, Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Anna Kuballa
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
- School of Health, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia
| | - Mohammad Katouli
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore DC, QLD 4558, Australia; (B.A.); (J.R.B.); (B.L.Q.); (G.B.); (A.K.)
| |
Collapse
|
5
|
Lee CC, Chiu CH. Link between gut microbiota and neonatal sepsis. J Formos Med Assoc 2024; 123:638-646. [PMID: 37821302 DOI: 10.1016/j.jfma.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/15/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023] Open
Abstract
In neonates, the gastrointestinal tract is rapidly colonized by bacteria after birth. Gut microbiota development is critical during the first few years of life. However, disruption of gut microbiota development in neonates can lead to gut dysbiosis, characterized by overcolonization by pathogenic bacteria and delayed or failed maturation toward increasing microbial diversity and Fermicutes dominance. Gut dysbiosis can predispose infants to sepsis. Pathogenic bacteria can colonize the gut prior to sepsis and cause sepsis through translocation. This review explores gut microbiota development in neonates, the evidence linking gut dysbiosis to neonatal sepsis, and the potential role of probiotics in gut microbiota modulation and sepsis prevention.
Collapse
Affiliation(s)
- Chien-Chung Lee
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan; Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
6
|
Xu H, You J, He W, Pei L, Han Y, Wang X, Tian Z, Zheng X, Wu E, Ling Y. Dynamic changes in the migratory microbial components of colon tissue during different periods of sepsis in an LPS-induced rat model. Front Cell Infect Microbiol 2024; 13:1330087. [PMID: 38287976 PMCID: PMC10822926 DOI: 10.3389/fcimb.2023.1330087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/26/2023] [Indexed: 01/31/2024] Open
Abstract
Previous studies have shown that bacterial translocation may play an important role in worsening gastrointestinal injury during sepsis. However, the dynamics of specific microbiota components in intestinal tissues at different sepsis stages remain unclear. Rats receiving intraperitoneal lipopolysaccharide (LPS) were sacrificed at 12 h and 48 h post-injection. Routine blood, serum cytokines, and microbiota in colon tissue, colonic contents, and lung tissue at different time points were assessed. Migratory microbial components in colonic tissue at 12 h and 48 h post-LPS were identified using source tracking, characteristic component identification, and abundance difference analyses. Colonic tissue microbiota changed dynamically over time after LPS injection, involving translocation of microbial components from colon contents and lung tissue at different time points. Bacteria migrating to colon tissue at 12 h sepsis were mainly from colonic contents, while those at 48 h were predominantly from the lung tissue. The migratory microbial components in colon tissue were widely associated with blood indicators and colonizing genus abundance and microbiota functionality in colon tissue. In this study, the temporal dynamics of bacterial translocation from various sources into colon tissues at different sepsis progression stages were characterized for the first time, and the species composition of these migrating microbes was delineated. These bacterial migrants may contribute to the pathophysiological processes in sepsis through direct interactions or indirectly by modulating colonic microbiota community structure and function.
Collapse
Affiliation(s)
- Hao Xu
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Jia You
- Department of Obstetrics and Gynecology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wenqin He
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Lingpeng Pei
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Yue Han
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Xueer Wang
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Zhigang Tian
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiwei Zheng
- Department of Respiratory and Critical Care Medicine, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Enqi Wu
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| | - Yaqin Ling
- School of Pharmacy, Minzu University of China, Beijing, China
- Key Laboratory of Ethnomedicine (Minzu University of China), Ministry of Education, Beijing, China
| |
Collapse
|
7
|
Madhav A, Bousfield R, Pereira-Dias J, Cormie C, Forrest S, Keane J, Kermack L, Higginson E, Dougan G, Spiers H, Massey D, Sharkey L, Rutter C, Woodward J, Russell N, Amin I, Butler A, Atkinson K, Dymond T, Bartholdson Scott J, Baker S, Gkrania-Klotsas E. A metagenomic prospective cohort study on gut microbiome composition and clinical infection in small bowel transplantation. Gut Microbes 2024; 16:2323232. [PMID: 38439546 PMCID: PMC10936650 DOI: 10.1080/19490976.2024.2323232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
Two-thirds of small-bowel transplantation (SBT) recipients develop bacteremia, with the majority of infections occurring within 3 months post-transplant. Sepsis-related mortality occurs in 31% of patients and is commonly caused by bacteria of gut origin, which are thought to translocate across the implanted organ. Serial post-transplant surveillance endoscopies provide an opportunity to study whether the composition of the ileal and colonic microbiota can predict the emergence as well as the pathogen of subsequent clinical infections in the SBT patient population. Five participants serially underwent aspiration of ileal and colonic bowel effluents at transplantation and during follow-up endoscopy either until death or for up to 3 months post-SBT. We performed whole-metagenome sequencing (WMS) of 40 bowel effluent samples and compared the results with clinical infection episodes. Microbiome composition was concordant between participants and timepoint-matched ileal and colonic samples. Four out of five (4/5) participants had clinically significant infections thought to be of gut origin. Bacterial translocation from the gut was observed in 3/5 patients with bacterial infectious etiologies. In all three cases, the pathogens had demonstrably colonized the gut between 1-10 days prior to invasive clinical infection. Recipients with better outcomes received donor grafts with higher alpha diversity. There was an increase in the number of antimicrobial resistance genes associated with longer hospital stay for all participants. This metagenomic study provides preliminary evidence to support the pathogen translocation hypothesis of gut-origin sepsis in the SBT cohort. Ileal and colonic microbiome compositions were concordant; therefore, fecal metagenomic analysis could be a useful surveillance tool for impeding infection with specific gut-residing pathogens.
Collapse
Affiliation(s)
- Archana Madhav
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rachel Bousfield
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Joana Pereira-Dias
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Claire Cormie
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Sally Forrest
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jacqueline Keane
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Leanne Kermack
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ellen Higginson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Gordon Dougan
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Harry Spiers
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Dunecan Massey
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lisa Sharkey
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Charlotte Rutter
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jeremy Woodward
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Neil Russell
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Irum Amin
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Andrew Butler
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Kayleigh Atkinson
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Tom Dymond
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Josefin Bartholdson Scott
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Effrossyni Gkrania-Klotsas
- Department of Medicine / Department of Surgery, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
8
|
Suen KFK, Low JXY, Charalambous CP. Urinary tract infection is associated with 2.4-fold increased risk of surgical site infection in hip fracture surgery: systematic review and meta-analysis. J Hosp Infect 2023; 139:56-66. [PMID: 37343771 DOI: 10.1016/j.jhin.2023.06.016] [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: 02/24/2023] [Revised: 06/15/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND There is no consensus regarding whether urinary tract infection (UTI) should be screened for or treated in hip fracture patients. AIM To assess the relationship between perioperative UTI and surgical site infection (SSI) in hip fracture patients, and the relationship between urinary catheterization and SSI in these patients. METHODS PubMed, Embase, CINAHL and Cochrane Library were searched to identify studies that evaluated the relationship between perioperative UTI and SSI and/or between urinary catheterization and SSI. Articles were included if they used the term UTI or specified UTI as symptomatic bacteriuria. FINDINGS A total of 4139 records were identified, with eight studies included. Meta-analysis of seven studies which evaluated perioperative UTI and SSI showed an SSI rate of 7.1% (95% confidence interval (CI): 3.8-13.2) among 1217 patients with UTI vs 2.4% (95% CI: 1.0-5.7) in 36,514 patients without UTI (OR: 2.41; 95% CI: 1.67-3.46; P < 0.001). In three studies which specifically defined UTI as symptomatic bacteriuria, the SSI rate among UTI patients was 5.7% (95% CI: 4.0-8.1) vs 1.1% (95% CI: 0.2-5.2) in those without UTI (OR: 3.00; 95% CI: 0.55-16.26; P = 0.20). One study evaluated urinary catheterization and SSI. CONCLUSION Perioperative UTI is associated with a higher risk of SSI among hip fracture patients but the evidence is limited by the heterogeneity in the definition of UTI. We recommend considering the possibility of perioperative UTI in hip fracture patients, with treatment administered as necessary to reduce SSI rates.
Collapse
Affiliation(s)
- K F K Suen
- Department of Orthopaedics, Blackpool Victoria Hospital, Blackpool Teaching Hospitals NHS Foundation Trust, Blackpool, Lancashire, UK
| | - J X Y Low
- Department of Orthopaedics, Blackpool Victoria Hospital, Blackpool Teaching Hospitals NHS Foundation Trust, Blackpool, Lancashire, UK
| | - C P Charalambous
- Department of Orthopaedics, Blackpool Victoria Hospital, Blackpool Teaching Hospitals NHS Foundation Trust, Blackpool, Lancashire, UK; School of Medicine, University of Central Lancashire, Preston, Lancashire, UK.
| |
Collapse
|
9
|
Wang Y, Wang Y, Ma J, Li Y, Cao L, Zhu T, Hu H, Liu H. YuPingFengSan ameliorates LPS-induced acute lung injury and gut barrier dysfunction in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116452. [PMID: 37019161 DOI: 10.1016/j.jep.2023.116452] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yupingfengsan (YPFS) is a traditional Chinese medicine decoction. YPFS comprises Astragalus mongholicus Bunge (Huangqi), Atractylodes rubra Dekker (Baizhu), and Saposhnikovia divaricata (Turcz.ex Ledeb.) Schischk (Fangfeng). YPFS is commonly used to treat chronic obstructive pulmonary disease, asthma, respiratory infections, and pneumonia, but the mechanism of action remains unclear. AIM OF THE STUDY Acute lung injury (ALI) and its severe form of acute respiratory distress syndrome (ARDS) cause morbidity and mortality in critical patients. YPFS is a commonly used herbal soup to treat respiratory and immune system diseases. Nevertheless, the effect of YPFS on ALI remains unclear. This study aimed to investigate the effect of YPFS on lipopolysaccharide (LPS)-induced ALI in mice and elucidate its potential molecular mechanisms. MATERIALS AND METHODS The major components of YPFS were detected by High-performance liquid chromatography (HPLC). C57BL/6J mice were given YPFS for seven days and then treated with LPS. IL-1β, IL-6, TNF-α, IL-8, iNOS, NLRP3, PPARγ, HO-1, ZO-1, Occludin, Claudin-1, AQP3, AQP4, AQP5, ENaCα, ENaCβ, EnaCγ mRNA in lung and ZO-1, Occludin, Claudin-1, AQP3, AQP4, AQP5, ENaCα, ENaCβ, and EnaCγ mRNA in colon tissues were measured by Real-Time Quantitative PCR (RT-qPCR). The expressions of TLR4, MyD88, NOD-like receptor thermal protein domain associated protein 3 (NLRP3), ASC, MAPK signaling pathway, Nrf2, and HO-1 in the lung were detected by Western blot. Plasma inflammatory factors Interleukin (IL)-1β, IL-6, and Tumor Necrosis Factor-α (TNF-α) were determined by Enzyme-linked Immunosorbent Assay (ELISA). Lung tissues were processed for H & E staining, and colon tissues for HE, WGA-FITC, and Alcian Blue staining. RESULTS The results showed that YPFS administration alleviated lung injury and suppressed the production of inflammatory factors, including IL-1β, IL-6, and TNF-α. Additionally, YPFS reduced pulmonary edema by promoting the expressions of aquaporin and sodium channel-related genes (AQP3, AQP4, AQP5, ENaCα, ENaCβ, and EnaCγ). Further, YPFS intervention exhibited a therapeutic effect on ALI by inhibiting the activation of the NLRP3 inflammasome and MAPK signaling pathways. Finally, YPFS improved gut barrier integrity and suppressed intestinal inflammation in LPS-challenged mice. CONCLUSIONS YPFS protected mice against LPS-induced ALI by attenuating lung and intestinal tissue damage. This study sheds light on the potential application of YPFS to treat ALI/ARDS.
Collapse
Affiliation(s)
- Yao Wang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China; College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China
| | - Yanchun Wang
- Clinical College of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China
| | - Jun Ma
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China
| | - Yanan Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China
| | - Lu Cao
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China
| | - Tianxiang Zhu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China
| | - Haiming Hu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan, 430065, PR China.
| |
Collapse
|
10
|
Grigorean VT, Erchid A, Coman IS, Liţescu M. Colorectal Cancer-The "Parent" of Low Bowel Obstruction. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050875. [PMID: 37241107 DOI: 10.3390/medicina59050875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/29/2023] [Accepted: 04/30/2023] [Indexed: 05/28/2023]
Abstract
Introduction: Despite the improvement of early diagnosis methods for multiple pathological entities belonging to the digestive tract, bowel obstruction determined by multiple etiologies represents an important percentage of surgical emergencies. General data: Although sometimes obstructive episodes are possible in the early stages of colorectal cancer, the most commonly installed intestinal obstruction has the significance of an advanced evolutionary stage of neoplastic disease. Development of Obstructive Mechanism: The spontaneous evolution of colorectal cancer is always burdened by complications. The most common complication is low bowel obstruction, found in approximately 20% of the cases of colorectal cancer, and it can occur either relatively abruptly, or is preceded by initially discrete premonitory symptoms, non-specific (until advanced evolutionary stages) and generally neglected or incorrectly interpreted. Success in the complex treatment of a low neoplastic obstruction is conditioned by a complete diagnosis, adequate pre-operative preparation, a surgical act adapted to the case (in one, two or three successive stages), and dynamic postoperative care. The moment of surgery should be chosen with great care and is the result of the experience of the anesthetic-surgical team. The operative act must be adapted to the case and has as its main objective the resolution of intestinal obstruction and only in a secondary way the resolution of the generating disease. Conclusions: The therapeutic measures adopted (medical-surgical) must have a dynamic character in accordance with the particular situation of the patient. Except for certain or probably benign etiologies, the possibility of colorectal neoplasia should always be considered, in low obstructions, regardless of the patient's age.
Collapse
Affiliation(s)
- Valentin Titus Grigorean
- General Surgery Department, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
- General Surgery Department, "Bagdasar-Arseni" Clinical Emergency Hospital, 12 Berceni Road, 041915 Bucharest, Romania
| | - Anwar Erchid
- General Surgery Department, "Bagdasar-Arseni" Clinical Emergency Hospital, 12 Berceni Road, 041915 Bucharest, Romania
| | - Ionuţ Simion Coman
- General Surgery Department, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
- General Surgery Department, "Bagdasar-Arseni" Clinical Emergency Hospital, 12 Berceni Road, 041915 Bucharest, Romania
| | - Mircea Liţescu
- General Surgery Department, "Carol Davila" University of Medicine and Pharmacy, 37 Dionisie Lupu Street, 020021 Bucharest, Romania
- General Surgery Department, "Sf. Ioan" Clinical Emergency Hospital, 13 Vitan-Bârzeşti Road, 042122 Bucharest, Romania
| |
Collapse
|
11
|
Chen J, Zou Y, Zheng T, Huang S, Guo L, Lin J, Zheng Q. The in Vitro Fermentation of Cordyceps militaris Polysaccharides Changed the Simulated Gut Condition and Influenced Gut Bacterial Motility and Translocation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14193-14204. [PMID: 36305603 DOI: 10.1021/acs.jafc.2c05785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The motility ability of intestinal lipopolysaccharide (LPS)-producing bacteria determines their translocation to the enterohepatic circulation and works as an infectious complication. In this study, the health effects of Cordyceps militaris polysaccharides (CMPs) were re-evaluated based on whether these polysaccharides could affect the motility of gut commensal LPS-producing bacteria and impede their translocation. The results showed that CMP-m fermentation in the gut could change the chemical environment, leading to a decrease in velocity and a shift in the motility pattern. Further study suggested that detachment/fragmentation of flagella, decreased motor forces, and changed chemical conditions might account for this weakened motility. The adhesion and invasion abilities of gut bacteria were also reduced, with lower expression of virulence-related genes. These results indicated that the health regulation effects of CMP-m might be through decreasing the motility of LPS-producing bacteria, hindering their translocation and therefore reducing the LPS level in the enterohepatic circulation.
Collapse
Affiliation(s)
- Jieming Chen
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| | - Yuan Zou
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| | - Taotao Zheng
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| | - Shishi Huang
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| | - Liqiong Guo
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| | - Junfang Lin
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| | - Qianwang Zheng
- Institute of Food Biotechnology and College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510640, China
- Research Center for Micro-Ecological Agent Engineering and Technology of Guangdong Province, Guangzhou 510640, China
| |
Collapse
|
12
|
Effects of Different Routes and Forms of Vitamin D Administration on Mesenteric Lymph Node CD4+ T Cell Polarization and Intestinal Injury in Obese Mice Complicated with Polymicrobial Sepsis. Nutrients 2022; 14:nu14173557. [PMID: 36079813 PMCID: PMC9460651 DOI: 10.3390/nu14173557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 12/02/2022] Open
Abstract
This study compared the efficacies of enteral cholecalciferol and/or intravenous (IV) calcitriol administration on mesenteric lymph node (MLN) cluster-of-differentiation-4-positive (CD4+) T cell distribution and intestinal barrier damage in obese mice complicated with sepsis. Mice were fed a high-fat diet for 16 weeks and then sepsis was induced by cecal ligation and puncture (CLP). Mice were divided into the following sepsis groups: without vitamin D (VD) (S); with oral cholecalciferol 1 day before CLP (G); with IV calcitriol 1 h after CLP (V); and with both cholecalciferol before and IV calcitriol after CLP (GV). All mice were sacrificed at 12 or 24 h after CLP. The findings show that the S group had a higher T helper (Th)17 percentage than the VD-treated groups at 12 h after CLP. The V group exhibited a higher Th1 percentage and Th1/Th2 ratio than the other groups at 24 h, whereas the V and GV groups had a lower Th17/regulatory T (Treg) ratio 12 h post-CLP in MLNs. In ileum tissues, the VD-treated groups had higher tight junction protein and cathelicidin levels, and higher mucin gene expression than the S group at 24 h post-CLP. Also, aryl hydrocarbon receptor (AhR) and its associated cytochrome P450 1A1 and interleukin 22 gene expressions were upregulated. In contrast, levels of lipid peroxides and inflammatory mediators in ileum tissues were lower in the groups with VD treatment after CLP. These results suggest that IV calcitriol seemed to have a more-pronounced effect on modulating the homeostasis of Th/Treg subsets in MLNs. Both oral cholecalciferol before and IV calcitriol after CLP promoted cathelicidin secretion, alleviated intestinal inflammation, and ameliorated the epithelial integrity in obese mice complicated with sepsis possibly via VD receptor and AhR signaling pathways.
Collapse
|
13
|
Sanidad KZ, Amir M, Ananthanarayanan A, Singaraju A, Shiland NB, Hong HS, Kamada N, Inohara N, Núñez G, Zeng MY. Maternal gut microbiome-induced IgG regulates neonatal gut microbiome and immunity. Sci Immunol 2022; 7:eabh3816. [PMID: 35687695 DOI: 10.1126/sciimmunol.abh3816] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The gut microbiome elicits antigen-specific immunoglobulin G (IgG) at steady state that cross-reacts to pathogens to confer protection against systemic infection. The role of gut microbiome-specific IgG antibodies in the development of the gut microbiome and immunity against enteric pathogens in early life, however, remains largely undefined. In this study, we show that gut microbiome-induced maternal IgG is transferred to the neonatal intestine through maternal milk via the neonatal Fc receptor and directly inhibits Citrobacter rodentium colonization and attachment to the mucosa. Enhanced neonatal immunity against oral C. rodentium infection was observed after maternal immunization with a gut microbiome-derived IgG antigen, outer membrane protein A, or induction of IgG-inducing gut bacteria. Furthermore, by generating a gene-targeted mouse model with complete IgG deficiency, we demonstrate that IgG knockout neonates are more susceptible to C. rodentium infection and exhibit alterations of the gut microbiome that promote differentiation of interleukin-17A-producing γδ T cells in the intestine, which persist into adulthood and contribute to increased disease severity in a dextran sulfate sodium-induced mouse model of colitis. Together, our studies have defined a critical role for maternal gut microbiome-specific IgG antibodies in promoting immunity against enteric pathogens and shaping the development of the gut microbiome and immune cells in early life.
Collapse
Affiliation(s)
- Katherine Z Sanidad
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.,Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Mohammed Amir
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.,Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Aparna Ananthanarayanan
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.,Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Anvita Singaraju
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Nicholas B Shiland
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA
| | - Hanna S Hong
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.,WPI Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Naohiro Inohara
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Melody Y Zeng
- Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY, USA.,Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.,Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
14
|
Chen Y, Qi A, Teng D, Li S, Yan Y, Hu S, Du X. Probiotics and synbiotics for preventing postoperative infectious complications in colorectal cancer patients: a systematic review and meta-analysis. Tech Coloproctol 2022; 26:425-436. [PMID: 35348943 DOI: 10.1007/s10151-022-02585-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/22/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND The health benefits of probiotics and synbiotics in healthy adults are well established, but their role in preventing infectious complications after surgery for colorectal cancer remains controversial. The aim of this meta-analysis was to assess the impact of probiotics/synbiotics on the incidence of infectious complications in patients who had surgery for colorectal cancer. METHODS A comprehensive literature search of all randomized control trials (RCTs) was conducted using PubMed, Embase, World Health Organization (WHO) Global Index Medicus, WHO clinical trial registry, and Clinicaltrials.gov. Inclusion criteria included RCTs comparing the use of any strain or dose of a specified probiotic/synbiotic with placebo or a "standard care" control group. The incidence of postoperative infectious complications was analyzed. RESULTS Fourteen RCTs involving 1566 patients (502 receiving probiotics, 273 receiving synbiotics, and 791 receiving placebo) were analyzed. Overall, probiotic or synbiotic administration significantly reduced the risk of developing postoperative infectious complications by 37% (relative risk (RR) = 0.63, 95% confidence interval (CI) 0.54-0.74, p < 0.001). Furthermore, when considering the six different types of postoperative infectious complications (septicemia, incision infection, central line infection, pneumonia infection, urinary infection, and incidence of diarrhea), probiotic or synbiotic administration was beneficial in reducing the incidence of each one of them. The quality of evidence was listed below: incidence of diarrhea (high), septicemia (moderate), incision infection (moderate), pneumonia infection (moderate), urinary infection (moderate), and central line infection (low). However, for the main outcome of infectious complications, we found evidence of possible publication bias, although estimates still showed a reduction following trim-and-fill analysis (RR = 0.72, 95% CI 0.62-0.84, p < 0.001). CONCLUSIONS The use of probiotic/synbiotic supplementation is associated with a significant reduction in the risk of developing postoperative infectious complications in patients who had surgery for colorectal cancer. Additional studies are needed to confirm the findings due to publication bias and low quality of evidence.
Collapse
Affiliation(s)
- Y Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - A Qi
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - D Teng
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - S Li
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - Y Yan
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - S Hu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China
| | - X Du
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Haidian, Beijing, 100853, China.
| |
Collapse
|
15
|
Potruch A, Schwartz A, Ilan Y. The role of bacterial translocation in sepsis: a new target for therapy. Therap Adv Gastroenterol 2022; 15:17562848221094214. [PMID: 35574428 PMCID: PMC9092582 DOI: 10.1177/17562848221094214] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Sepsis is a leading cause of death in critically ill patients, primarily due to multiple organ failures. It is associated with a systemic inflammatory response that plays a role in the pathogenesis of the disease. Intestinal barrier dysfunction and bacterial translocation (BT) play pivotal roles in the pathogenesis of sepsis and associated organ failure. In this review, we describe recent advances in understanding the mechanisms by which the gut microbiome and BT contribute to the pathogenesis of sepsis. We also discuss several potential treatment modalities that target the microbiome as therapeutic tools for patients with sepsis.
Collapse
|
16
|
Martel J, Chang SH, Ko YF, Hwang TL, Young JD, Ojcius DM. Gut barrier disruption and chronic disease. Trends Endocrinol Metab 2022; 33:247-265. [PMID: 35151560 DOI: 10.1016/j.tem.2022.01.002] [Citation(s) in RCA: 219] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 02/06/2023]
Abstract
The intestinal barrier protects the host against gut microbes, food antigens, and toxins present in the gastrointestinal tract. However, gut barrier integrity can be affected by intrinsic and extrinsic factors, including genetic predisposition, the Western diet, antibiotics, alcohol, circadian rhythm disruption, psychological stress, and aging. Chronic disruption of the gut barrier can lead to translocation of microbial components into the body, producing systemic, low-grade inflammation. While the association between gut barrier integrity and inflammation in intestinal diseases is well established, we review here recent studies indicating that the gut barrier and microbiota dysbiosis may contribute to the development of metabolic, autoimmune, and aging-related disorders. Emerging interventions to improve gut barrier integrity and microbiota composition are also described.
Collapse
Affiliation(s)
- Jan Martel
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Shih-Hsin Chang
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Yun-Fei Ko
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Chang Gung Biotechnology Corporation, Taipei, Taiwan; Biochemical Engineering Research Center, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - John D Young
- Chang Gung Biotechnology Corporation, Taipei, Taiwan.
| | - David M Ojcius
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, CA, USA.
| |
Collapse
|
17
|
Kaushal K, Gupta V, Goswami P, Agarwal S, Sharma S, Das P, Yadav R, Anand A, Sonika U, Gunjan D, Saraya A. Acute Variceal Bleed in Cirrhosis is Associated With Reversible Changes in Tight Junction Protein Expression in the Intestine: A Proof-of-Concept Study. J Clin Exp Hepatol 2022; 12:89-100. [PMID: 35068789 PMCID: PMC8766542 DOI: 10.1016/j.jceh.2021.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/23/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Tight junction proteins (TJPs) play an important role in gut-barrier dysfunction in cirrhosis and its complications such as acute variceal bleed (AVB). However, the dynamics of TJPs expression after AVB, its relation to bacterial translocation, and impact on clinical outcome is largely unknown. AIMS The aim of this study was to study the expression of TJPs in cirrhosis and assess its dynamic changes in AVB. In addition, the relation of TJP expression to endotoxemia and clinical outcomes was assessed. METHODS In this prospective pilot study, 17 patients of cirrhosis with AVB, 59 patients of cirrhosis without AVB (non-AVB cirrhosis), and 20 controls were assessed for claudin-2 and claudin-4 expression in the duodenal biopsy. In the AVB-cirrhosis group, additional biopsies were obtained after 3 weeks. Endotoxemia was assessed by measuring IgG anti-endotoxin antibody levels. Claudin expression was correlated with a 6-month survival. RESULTS Claudin-2 expression was downregulated in patients with AVB and non-AVB cirrhosis in villi (P < 0.001 and 0.013) and crypts (P < 0.001 and 0.012), respectively, compared with the controls. Claudin-4 expression was similar in villi (P = 0.079), but lower in crypts (P = 0.007) in patients with cirrhosis. Claudin-2 expression was upregulated on serial biopsies in both villi and crypts (P = 0.003 and 0.001, respectively) in AVB-cirrhosis with postbleed expression comparable with those with non-AVB cirrhosis. IgG anti-endotoxin antibody levels were elevated in cirrhosis with no correlation with claudin-2/4 expression. Claudin-2 expression independently predicted survival at 6 months. CONCLUSION Both claudin-2 and claudin-4 expression are downregulated in cirrhosis. AVB is associated with dynamic changes in TJPs expression. Gut-barrier dysfunction might predict outcomes independent of bacterial endotoxemia in cirrhosis.
Collapse
Key Words
- AVB, Acute Variceal Bleed
- DAB, 3,3′-Diaminobenzidine
- EBL, Endoscopic Band Ligation
- EGD, Esophagogastroduodenoscopy
- HRV, high-risk varices
- IHC, Immunohistochemistry
- NSBB, Non-selective Beta Blockers
- PAMP, Pathogen associated molecular patterns
- TJP, Tight Junction Protein
- TMB, 3, 3′, 5, 5′-tetramethylbenzidine
- acute variceal bleed
- claudin-2
- endotoxemia
- tight junction proteins
Collapse
Affiliation(s)
- Kanav Kaushal
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Vipin Gupta
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Pooja Goswami
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Samagra Agarwal
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Sanchit Sharma
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Abhinav Anand
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Ujjwal Sonika
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Deepak Gunjan
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Anoop Saraya
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029, India,Address for correspondence: Anoop Saraya, Professor and Head, Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi, 110029, India. Tel.: +91 9868397203.
| |
Collapse
|
18
|
Kaushal K, Agarwal S, Sharma S, Goswami P, Singh N, Sachdev V, Poudel S, Das P, Yadav R, Kumar D, Pandey G, Gunjan D, Saraya A. Demonstration of Gut-Barrier Dysfunction in Early Stages of Non-alcoholic Fatty Liver Disease: A Proof-Of-Concept Study. J Clin Exp Hepatol 2022; 12:1102-1113. [PMID: 35814507 PMCID: PMC9257921 DOI: 10.1016/j.jceh.2022.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND/AIMS Gut-barrier dysfunction is well recognized in pathogenesis of both non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). However, comparison of components of this dysfunction between the two etiologies remains unexplored especially in early stages of NAFLD. METHODS Components of gut-barrier dysfunction like alterations in intestinal permeability (IP) by lactulose mannitol ratio (LMR) in urine, systemic endotoxemia (IgG and IgM anti-endotoxin antibodies), systemic inflammation (serum tumor necrosis factor alpha [TNF-α] and interleukin-1 [IL-1] levels), tight junction (TJ) proteins expression in duodenal biopsy and stool microbiota composition using Oxford Nanopore MinION device were prospectively evaluated in patients with NAFLD (n = 34) with no cirrhosis, ALD (n = 28) and were compared with disease free controls (n = 20). RESULTS Patients with ALD had more advanced disease than those with NAFLD (median liver stiffness -NAFLD:7.1 kPa [5.9-8.9] vs. ALD:14.3 kPa [9.6-24], P < 0.001]. Median LMR was significantly higher in NAFLD and ALD group when compared to controls (NAFLD 0.054 [0.037-0.17] vs. controls 0.027 [0.021-0.045] (P = 0.001)) and ALD 0.043 [0.03-0.068] vs. controls 0.027 [0.021-0.045] (P = 0.019)]. Anti-endotoxin antibody titer (IgM) (MMU/mL) was lowest in NAFLD 72.9 [3.2-1089.5] compared to ALD 120.6 [20.1-728]) (P = 0.042) and controls 155.3 [23.8-442.9]) (P = 0.021). Median TNF-α (pg/mL) levels were elevated in patients with NAFLD (53.3 [24.5-115]) compared to controls (16.1 [10.8-33.3]) (P < 0.001) and ALD (12.3 [10.1-42.7]) (P < 0.001). Expression of zonulin-1 and claudin-3 in duodenal mucosa was lowest in NAFLD. On principal co-ordinate analysis (PCoA), the global bacterial composition was significantly different across the three groups (PERMANOVA test, P < 0.001). CONCLUSION While remaining activated in both etiologies, gut-barrier dysfunction abnormalities were more pronounced in NAFLD at early stages compared to ALD despite more advanced disease in the latter.
Collapse
Key Words
- ALD, alcoholic liver disease
- ALT, alanine transaminase
- AST, aspartate transaminase
- IL-1, interleukin-1
- IP, intestinal permeability
- KFT, kidney function test
- LFT, liver function test
- LMR, lactulose mannitol ratio
- NAFLD, non-alcoholic fatty liver disease
- TNF, tumor necrosis factor
- alcoholic liver disease
- endotoxemia
- intestinal permeability
- non-alcoholic fatty liver disease
- tight junction protein
Collapse
Affiliation(s)
- Kanav Kaushal
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Samagra Agarwal
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Sanchit Sharma
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Pooja Goswami
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Namrata Singh
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Vikas Sachdev
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Shekhar Poudel
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Rajni Yadav
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Dinesh Kumar
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Gaurav Pandey
- Department of Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, 226014, India
| | - Deepak Gunjan
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India
| | - Anoop Saraya
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India,Address for correspondence: Anoop Saraya, Professor and Head, Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, 110029 , India.
| |
Collapse
|
19
|
Yeh CL, Wu JM, Yang PJ, Lee PC, Chen KY, Huang CC, Yeh SL, Lin MT. Intravenous calcitriol administration modulates mesenteric lymph node CD4 + T-cell polarization and attenuates intestinal inflammation in obese mice complicated with polymicrobial sepsis. JPEN J Parenter Enteral Nutr 2021; 46:1371-1383. [PMID: 34882304 DOI: 10.1002/jpen.2313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Sepsis is a lethal syndrome with T-cell dysregulation, imbalanced inflammatory reactions, and gastrointestinal dysfunction. Obesity coexistent with sepsis can cause more-deleterious disease outcomes. Vitamin D is a nutrient with immunomodulatory ability and helps maintain intestinal homeostasis. This study investigated treatment with calcitriol on mesenteric lymph node (MLN) CD4+ T-cell polarization and intestinal injury in obese mice with sepsis. METHODS Mice received a high-fat diet for 10 weeks; then, mice were separated into an obese control group without sepsis and sepsis groups that underwent cecal ligation and puncture (CLP). Septic mice were subdivided into a group that was injected with saline (SS group) or a group that was injected with calcitriol (SD group) via a tail vein 1 h after CLP. Obese mice with sepsis were euthanized at 12 or 24 h post CLP. RESULTS Sepsis resulted in increased percentages of type 2 T helper (Th2), Th17, and regulatory T (Treg) cells in MLNs. Also, inflammation-associated genes were upregulated and tight junction genes downregulated in the intestines after CLP. Compared with the SS group, the SD group exhibited reduced Th2, Th17, and Treg percentages in MLNs. Also, intestinal inflammatory chemokine expressions were reduced, whereas MUC2, ZO-1, and occludin had increased after CLP. Lower inflammatory cytokine levels in peritoneal lavage fluid in the ileum were also noted in the SD group. CONCLUSIONS Intravenous calcitriol treatment after sepsis can elicit more-balanced CD4 T-cell subsets in lymph nodes near the intestines and alleviate intestinal inflammation and injury in obese mice complicated with sepsis.
Collapse
Affiliation(s)
- Chiu-Li Yeh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei, Taiwan.,Nutrition Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Jin-Ming Wu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu City, Taiwan
| | - Po-Jen Yang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Chu Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuen-Yuan Chen
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Chieh Huang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu City, Taiwan
| | - Sung-Ling Yeh
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Tsan Lin
- Department of Surgery, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Intravenous calcitriol administration regulates the renin-angiotensin system and attenuates acute lung injury in obese mice complicated with polymicrobial sepsis. Biomed Pharmacother 2021; 141:111856. [PMID: 34217099 DOI: 10.1016/j.biopha.2021.111856] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/13/2021] [Accepted: 06/24/2021] [Indexed: 01/26/2023] Open
Abstract
Calcitriol, an active form of vitamin D, has immunomodulatory and anti-inflammatory properties. Vitamin D levels have inverse correlation with sepsis outcomes and obesity may aggravate the severity of the diseases. This study administered calcitriol to investigate its impact on sepsis-induced acute lung injury (ALI) in obese mice. Mice were fed a high-fat diet to induce obesity and were randomly assigned to control or sepsis groups, which were intravenously administered either saline (SS) or calcitriol (SD). Sepsis was induced by cecal ligation and puncture (CLP). Saline or calcitriol was injected 1 h after CLP via tail vein. Mice were sacrificed at either 12 or 24 h post-CLP and survival rates were observed. The results demonstrated that sepsis caused upregulation of inflammatory mediators and downregulation of renin-angiotensin system (RAS)-associated gene expressions in the lungs of obese mice. Cluster of differentiation 68 (CD68) expression and myeloperoxidase (MPO) activities also increased. Calcitriol treatment lowered expressions of blood and lung inflammatory mediators at 12 and/or 24 h after CLP. The RAS-proinflammatory-associated angiotensin type 1 receptor (AT1R) was lower while anti-inflammatory Mas receptor and AT2R expressions were higher at 12 h after CLP than those in the SS group. In addition, the SD group exhibited lower CD68 expression and MPO activity. Lower lung injury scores and higher survival rates were also noted in the SD group. The findings suggest that calcitriol treatment after sepsis induction upregulated RAS-associated anti-inflammatory pathway and decreased immune cell infiltration, which may have alleviated the severity of ALI of obese mice.
Collapse
|
23
|
Risk Factors Associated With Postendoscopic Mucosal Resection Bleeding in Patients With Cirrhosis: A Retrospective Multicenter Cohort Study. J Clin Gastroenterol 2021; 55:355-360. [PMID: 32796193 DOI: 10.1097/mcg.0000000000001407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM There is limited data regarding the safety of endoscopic mucosal resection (EMR) in the cirrhotic population. Our study aimed to evaluate the safety of colonoscopic EMR in cirrhosis. MATERIALS AND METHODS This was a retrospective review of cirrhotics who underwent colonic EMR at 8 Cleveland Clinic Centers between January 1, 2006, and December 31, 2018. Patient data including polyp details and complications occurring within 30 days of the procedure were noted. Univariable and multivariable logistic regression analyses were conducted to find risk factors for post-EMR bleeding. RESULTS A total of 238 patients who underwent EMR were included. There were 145 males (60.9%) and the mean age was 61.9±8.6 years. Immediate and delayed bleeding, and postpolypectomy syndrome rates were 9.2%, 5.8%, and 1.3%, respectively. Significant risk factors for postpolypectomy bleeding were: increased age (P=0.001), procedure duration >37 minutes (P=0.001), antiplatelet use within 5 days (P=0.023), and lesion diameter >15 mm (P=0.004). Multivariable analysis revealed independent predictors of procedure-related bleeding: age above 65 years [odds ratio (OR) 2.14, P=0.044], antiplatelet use within 5 days (OR 2.42, P=0.047), right colon polyp (OR 3.51, P=0.001), and lesion diameter >15 mm (OR 3.22, P=0.003). CONCLUSIONS EMR in cirrhotics has an acceptable bleeding risk. Age above 65 years, right colon polyp, polyp size >15 mm, and use of antiplatelets within 5 days are independent risk factors for bleeding.
Collapse
|
24
|
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
|
25
|
Tsaroucha A, Kaldis V, Vailas M, Schizas D, Lambropoulou M, Papalois A, Tsigalou C, Gaitanidis A, Pitiakoudis M, Simopoulos C. The positive effect of eugenol on acute pancreatic tissue injury: a rat experimental model. Pan Afr Med J 2021; 38:132. [PMID: 33912302 PMCID: PMC8052617 DOI: 10.11604/pamj.2021.38.132.20202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/20/2021] [Indexed: 11/18/2022] Open
Abstract
Introduction we present a rat experimental model used to evaluate the possible reduction in the extent of pancreatic tissue injury in acute pancreatitis cases, after administration of eugenol. Methods one hundred and twenty Wistar rats were used, which were randomly assigned in 3 groups: sham (n=20), control (n=50) and eugenol (n=50). Acute pancreatitis was induced by biliopancreatic ligation in the control and eugenol groups, but not in the Sham group. In the eugenol group, eugenol was administered per-os. Five histopathological parameters, such as edema, inflammatory infiltration, duct dilatation, hemorrhage and acinar necrosis were evaluated. Results at 72 h from acute pancreatitis induction, the total histological score was diminished in the eugenol group (p<0.0005) and duct dilatation and inflammatory infiltration were reduced compared to the control group (p<0.05). In addition, at 72 h, eugenol reduced pancreatic myeloperoxidase activity (p<0.0005). Conclusion eugenol, a highly free radical scavenger agent, may have a preventive role in acute pancreatic injury, as it was evident in our rat experimental model.
Collapse
Affiliation(s)
- Alexandra Tsaroucha
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,2nd Department of Surgery and Laboratory of Experimental Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Vasileios Kaldis
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michail Vailas
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,First Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Schizas
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,First Department of Surgery, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Lambropoulou
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,Laboratory of Histology-Embryology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Apostolos Papalois
- Experimental-Research Department, ELPEN Pharmaceuticals, Pikermi, Attica, Greece
| | - Christina Tsigalou
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,Laboratory of Microbiology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Apostolos Gaitanidis
- 2nd Department of Surgery and Laboratory of Experimental Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Michael Pitiakoudis
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,2nd Department of Surgery and Laboratory of Experimental Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Constantinos Simopoulos
- Postgraduate Program in Hepatobiliary/Pancreatic Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.,2nd Department of Surgery and Laboratory of Experimental Surgery, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
26
|
Barroso FAL, de Jesus LCL, de Castro CP, Batista VL, Ferreira Ê, Fernandes RS, de Barros ALB, Leclerq SY, Azevedo V, Mancha-Agresti P, Drumond MM. Intake of Lactobacillus delbrueckii (pExu: hsp65) Prevents the Inflammation and the Disorganization of the Intestinal Mucosa in a Mouse Model of Mucositis. Microorganisms 2021; 9:microorganisms9010107. [PMID: 33466324 PMCID: PMC7824804 DOI: 10.3390/microorganisms9010107] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023] Open
Abstract
5-Fluorouracil (5-FU) is an antineoplastic drug that causes, as a side effect, intestinal mucositis, acute inflammation in the small bowel. The Heat Shock Protein (Hsp) are highly expressed in inflammatory conditions, developing an important role in immune modulation. Thus, they are potential candidates for the treatment of inflammatory diseases. In the mucositis mouse model, the present study aimed to evaluate the beneficial effect of oral administration of milk fermented by Lactobacillus delbrueckii CIDCA 133 (pExu:hsp65), a recombinant strain. This approach showed increased levels of sIgA in the intestinal fluid, reducing inflammatory infiltrate and intestinal permeability. Additionally, the histological score was improved. Protection was associated with a reduction in the gene expression of pro-inflammatory cytokines such as Tnf, Il6, Il12, and Il1b, and an increase in Il10, Muc2, and claudin 1 (Cldn1) and 2 (Cldn2) gene expression in ileum tissue. These findings are corroborated with the increased number of goblet cells, the electronic microscopy images, and the reduction of intestinal permeability. The administration of milk fermented by this recombinant probiotic strain was also able to reverse the high levels of gene expression of Tlrs caused by the 5-FU. Thus, the rCIDCA 133:Hsp65 strain was revealed to be a promising preventive strategy for small bowel inflammation.
Collapse
Affiliation(s)
- Fernanda Alvarenga Lima Barroso
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Luís Cláudio Lima de Jesus
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Camila Prosperi de Castro
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Viviane Lima Batista
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Ênio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Renata Salgado Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus da UFMG, Universidade Federal de Minas Gerais, Cidade Universitária, Belo Horizonte 31270-901, Brazil; (R.S.F.); (A.L.B.d.B.)
| | - André Luís Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Campus da UFMG, Universidade Federal de Minas Gerais, Cidade Universitária, Belo Horizonte 31270-901, Brazil; (R.S.F.); (A.L.B.d.B.)
| | - Sophie Yvette Leclerq
- Laboratório de Inovação Biotecnológica, Fundação Ezequiel Dias (FUNED), Belo Horizonte 30510-010, Brazil;
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
| | - Pamela Mancha-Agresti
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
- Faculdade de Minas-Faminas-BH, Medicina, Belo Horizonte 31744-007, Brazil
- Correspondence: (P.M.-A.); (M.M.D.); Tel.: +55-31-99817-5004 (P.M.-A.); +55-31-99222-2761 (M.M.D.)
| | - Mariana Martins Drumond
- Laboratório de Genética Celular e Molecular (LGCM), Departamento de—Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, Brazil; (F.A.L.B.); (L.C.L.d.J.); (C.P.d.C.); (V.L.B.); (V.A.)
- Centro Federal de Educação Tecnológica de Minas Gerais (CEFET/MG), Departamento de Ciências Biológicas, Belo Horizonte 31421-169, Brazil
- Correspondence: (P.M.-A.); (M.M.D.); Tel.: +55-31-99817-5004 (P.M.-A.); +55-31-99222-2761 (M.M.D.)
| |
Collapse
|
27
|
Su SY, Tang QQ. Altered intestinal microflora and barrier injury in severe acute pancreatitis can be changed by zinc. Int J Med Sci 2021; 18:3050-3058. [PMID: 34400875 PMCID: PMC8364456 DOI: 10.7150/ijms.45980] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/04/2021] [Indexed: 01/01/2023] Open
Abstract
To investigate the effect of zinc (Zn) supplementation on intestinal microflora changes and bacterial translocation in rats with severe acute pancreatitis (SAP), the rats were divided into the sham surgery (SS), SAP, SS + Zn, and SAP + Zn groups. Saline (0.1 mL/100g) and 5% sodium taurocholate were injected into the pancreaticobiliary duct of the rats in the SS and SAP + Zn groups, respectively. Intraperitoneal injection of 5 mg/kg Zn was performed immediately after injecting saline or 5% sodium taurocholate into the rats in both groups. Serum amylase and Zn levels, plasma endogenous endotoxin, intestinal permeability, and the positive rate of intestinal bacterial translocation were detected, haematoxylin and eosin (H&E) staining was performed, and the pancreatic tissue scores were calculated for each group. In addition, immunohistochemical (IHC) staining was performed to evaluate the expression of IL-1β and TNF-α. Real-time fluorescence quantitative PCR was used to quantify the gene copy numbers of Escherichia, Bifidobacterium, and Lactobacillus in the cecum. The levels of amylase and plasma endotoxin in the SAP group were significantly higher than those in the SS and SS + Zn groups. Intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were increased in the SAP group. However, the levels of amylase and plasma endotoxin were decreased as a result of zinc supplementation in the SAP group. The expression of IL-1β and TNF-α was also reduced to a greater degree in the SAP + Zn group than in the SAP group. Moreover, alleviated intestinal mucosal permeability and intestinal bacterial translocation in the liver, pancreas, and mesenteric lymph nodes were found in the SAP + Zn group. The results of real-time quantitative PCR showed that the gene copy number of Escherichia increased with time, and the gene copy numbers of Lactobacillus and Bifidobacterium decreased over time. Zn supplementation prevented the release of TNF-α and IL-1β, alleviated intestinal permeability and endotoxemia, reduced bacterial translocation, and inhibited changes in pathogenic intestinal flora in rats with SAP.
Collapse
Affiliation(s)
- Shi-Yue Su
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei 230022, Anhui, China
| | - Qin-Qing Tang
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei 230022, Anhui, China
| |
Collapse
|
28
|
The Role of the Microbiome in Oral Squamous Cell Carcinoma with Insight into the Microbiome-Treatment Axis. Int J Mol Sci 2020; 21:ijms21218061. [PMID: 33137960 PMCID: PMC7662318 DOI: 10.3390/ijms21218061] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the leading presentations of head and neck cancer (HNC). The first part of this review will describe the highlights of the oral microbiome in health and normal development while demonstrating how both the oral and gut microbiome can map OSCC development, progression, treatment and the potential side effects associated with its management. We then scope the dynamics of the various microorganisms of the oral cavity, including bacteria, mycoplasma, fungi, archaea and viruses, and describe the characteristic roles they may play in OSCC development. We also highlight how the human immunodeficiency viruses (HIV) may impinge on the host microbiome and increase the burden of oral premalignant lesions and OSCC in patients with HIV. Finally, we summarise current insights into the microbiome–treatment axis pertaining to OSCC, and show how the microbiome is affected by radiotherapy, chemotherapy, immunotherapy and also how these therapies are affected by the state of the microbiome, potentially determining the success or failure of some of these treatments.
Collapse
|
29
|
Kumar N, Lata K, Ray MD. Drain site enterocutaneous fistula after hyperthermic intraperitoneal chemotherapy. JGH OPEN 2020; 5:172-174. [PMID: 33490634 PMCID: PMC7812455 DOI: 10.1002/jgh3.12446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 11/29/2022]
Abstract
Spontaneous sigmoid colon perforation after cytoreduction surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) is a rare complication. It is more commonly seen with mitomycin‐based HIPEC. This case study's patient presented with pus discharge at the drain site after 4 weeks of surgery. The symptoms persisted after conservative treatment. High suspicion after the feculent smell of the discharge fluidled to the prompt diagnosis of enterocutaneous fistula. There was limitedperforation with abscess formation, followed by fistula formation. The patient was treated successfully with surgery.
Collapse
Affiliation(s)
- Navin Kumar
- Department of surgical oncology All India Institute of Medical Sciences New Delhi India
| | - Kanak Lata
- Department of nuclear medicine All India Institute of Medical Sciences New Delhi India
| | - Mukur Dipi Ray
- Department of surgical oncology All India Institute of Medical Sciences New Delhi India
| |
Collapse
|
30
|
Ribeiro Hudson AS, Nascimento Soares AD, Coelho Horta NA, Fuscaldi LL, Machado-Moreira CA, Soares DD, Coimbra CC, de Oliveira Poletini M, Cardoso VN, Wanner SP. The magnitude of physical exercise-induced hyperthermia is associated with changes in the intestinal permeability and expression of tight junction genes in rats. J Therm Biol 2020; 91:102610. [PMID: 32716860 DOI: 10.1016/j.jtherbio.2020.102610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 04/01/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022]
Abstract
We investigated whether the magnitude of exercise-induced hyperthermia influences intestinal permeability and tight junction gene expression. Twenty-nine male Wistar rats were divided into four groups: rest at 24 °C and exercise at 13 °C, 24 °C or 31 °C. The exercise consisted of a 90-min treadmill run at 15 m/min, and different ambient temperatures were used to produce distinct levels of exercise-induced hyperthermia. Before the experimental trials, the rats were treated by gavage with diethylenetriaminepentaacetic acid labeled with technetium-99 metastable as a radioactive probe. The rats' core body temperature (TCORE) was measured by telemetry. Immediately after the trials, the rats were euthanized, and the intestinal permeability was assessed by measuring the radioactivity of blood samples. The mRNA levels of occludin and zonula occludens-1 (ZO-1) genes were determined in duodenum samples. Exercise at 24 °C increased TCORE to values close to 39 °C, without changing permeability compared with the resting trial at the same environment. Meanwhile, rats' TCORE exceeded 40 °C during exercise at 31 °C, leading to greater permeability relative to those observed after exercise in the other ambient temperatures (e.g., 0.0037%/g at 31 °C vs. 0.0005%/g at 13 °C; data expressed as medians; p < 0.05). Likewise, the rats exercised at 31 °C exhibited higher mRNA levels of ZO-1 and occludin genes than the rats exercised at 24 °C or 13 °C. The changes in permeability and gene expression were positively and significantly associated with the magnitude of hyperthermia. We conclude that marked hyperthermia caused by exercise in the warmer environment increases intestinal permeability and mRNA levels of tight junction genes.
Collapse
Affiliation(s)
- Alexandre Sérvulo Ribeiro Hudson
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Anne Danieli Nascimento Soares
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Nayara Abreu Coelho Horta
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leonardo Lima Fuscaldi
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Christiano Antônio Machado-Moreira
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danusa Dias Soares
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cândido Celso Coimbra
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maristela de Oliveira Poletini
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert Nascimento Cardoso
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Samuel Penna Wanner
- Exercise Physiology Laboratory, School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
31
|
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]
|
32
|
Spaziante M, Oliva A, Ceccarelli G, Alessandri F, Pugliese F, Venditti M. Follow-up blood cultures in Gram-negative bacilli bacteremia: are they needed for critically ill patients? Minerva Anestesiol 2020; 86:498-506. [PMID: 32100514 DOI: 10.23736/s0375-9393.20.14040-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Gram-negative bacilli bacteremias (GNB-Bs) represent a major cause of morbidity and mortality in Intensive Care Unit (ICU) patients. Aim of this study was to investigate the role of follow-up blood cultures (FUBCs) and the clinical significance of persistent bacteremia (PB) in these settings. METHODS We retrospectively analyzed clinical data and outcome of GNB-Bs that occurred in ICU patients over a span of 1 year. In particular we sought information on development and clinical details of PB, defined as repeatedly positive FUBCs after ≥96 hours of appropriate antibiotic treatment and ≥48 hours after removal of endovascular devices. RESULTS Among 307 ICU patients, 69 (22.4%) developed 107 GNB-Bs. Of these, 78 (73%) could be eventually analyzed: 50 of 78 (64.1%) were non-PBs from 26 patients and 28 of 78 (35.9%) were PBs from 23 patients. Duration of fever and bacteremia, time to procalcitonin normalization and weaning from vasopressors were longer in episodes of PBs than non-PBs (P=0.04, P<0.001, P=0.02 and P=0.004, respectively). Primary bacteremia was more frequent in non-PBs than in PBs (29 of 50, 58% vs. 3/28, 10.7%, P=0.0001), whereas septic thrombus infection (STI) was the source of infection in 14 of 28 (50%). Finally, clinical features and 30-day mortality did not differ between patients with PB and those who developed only non-PB episodes. CONCLUSIONS Among our ICU patients, more than one third of GNB-Bs for which FUBCs were performed resulted PB. This condition is often associated with the presence of STI; therefore, FUBCs seem useful for the optimal management of GNB in this clinical setting.
Collapse
Affiliation(s)
- Martina Spaziante
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Alessandra Oliva
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy.,IRCCS INM Neuromed, Pozzilli, Isernia, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Francesco Alessandri
- Department of Anesthesia and Intensive Care Medicine, Sapienza University, Rome, Italy
| | - Francesco Pugliese
- Department of Anesthesia and Intensive Care Medicine, Sapienza University, Rome, Italy
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy -
| |
Collapse
|
33
|
Ho J, Chan H, Liang Y, Liu X, Zhang L, Li Q, Zhang Y, Zeng J, Ugwu FN, Ho IHT, Hu W, Yau JCW, Wong SH, Wong WT, Ling L, Cho CH, Gallo RL, Gin T, Tse G, Yu J, Chan MTV, Leung CCH, Wu WKK. Cathelicidin preserves intestinal barrier function in polymicrobial sepsis. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2020; 24:47. [PMID: 32041659 PMCID: PMC7011568 DOI: 10.1186/s13054-020-2754-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/27/2020] [Indexed: 02/06/2023]
Abstract
Objectives The intestinal epithelium compartmentalizes the sterile bloodstream and the commensal bacteria in the gut. Accumulating evidence suggests that this barrier is impaired in sepsis, aggravating systemic inflammation. Previous studies reported that cathelicidin is differentially expressed in various tissues in sepsis. However, its role in sepsis-induced intestinal barrier dysfunction has not been investigated. Design To examine the role of cathelicidin in polymicrobial sepsis, cathelicidin wild-(Cnlp+/+) and knockout (Cnlp−/−) mice underwent cecal-ligation and puncture (CLP) followed by the assessment of septic mortality and morbidity as well as histological, biochemical, immunological, and transcriptomic analyses in the ileal tissues. We also evaluated the prophylactic and therapeutic efficacies of vitamin D3 (an inducer of endogenous cathelicidin) in the CLP-induced murine polymicrobial sepsis model. Results The ileal expression of cathelicidin was increased by three-fold after CLP, peaking at 4 h. Knockout of Cnlp significantly increased 7-day mortality and was associated with a higher murine sepsis score. Alcian-blue staining revealed a reduced number of mucin-positive goblet cells, accompanied by reduced mucin expression. Increased number of apoptotic cells and cleavage of caspase-3 were observed. Cnlp deletion increased intestinal permeability to 4kD fluorescein-labeled dextran and reduced the expression of tight junction proteins claudin-1 and occludin. Notably, circulating bacterial DNA load increased more than two-fold. Transcriptome analysis revealed upregulation of cytokine/inflammatory pathway. Depletion of Cnlp induced more M1 macrophages and neutrophils compared with the wild-type mice after CLP. Mice pre-treated with cholecalciferol (an inactive form of vitamin D3) or treated with 1alpha, 25-dihydroxyvitamin D3 (an active form of VD3) had decreased 7-day mortality and significantly less severe symptoms. Intriguingly, the administration of cholecalciferol after CLP led to worsened 7-day mortality and the associated symptoms. Conclusions Endogenous cathelicidin promotes intestinal barrier integrity accompanied by modulating the infiltration of neutrophils and macrophages in polymicrobial sepsis. Our data suggested that 1alpha, 25-dihydroxyvitamin D3 but not cholecalciferol is a potential therapeutic agent for treating sepsis.
Collapse
Affiliation(s)
- Jeffery Ho
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Hung Chan
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Yonghao Liang
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China.,State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Qing Li
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Yuchen Zhang
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Judeng Zeng
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Felix N Ugwu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Idy H T Ho
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Wei Hu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Johnny C W Yau
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Sunny H Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Shatin, China
| | - Wai Tat Wong
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Lowell Ling
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Chi H Cho
- Laboratory of Molecular Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Richard L Gallo
- Department of Dermatology, The University of California, San Diego, USA
| | - Tony Gin
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China
| | - Gary Tse
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Shatin, China
| | - Jun Yu
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China.,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Shatin, China
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China.
| | - Czarina C H Leung
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China.
| | - William K K Wu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China. .,State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region, China.
| |
Collapse
|
34
|
Wong YJ, Lum HM, Fook-Chong S, Lim ST, Salazar E. Do total parenteral nutrition and bowel rest reduce the risk for perforation in patients with gastrointestinal tract lymphoma receiving chemotherapy? Nutrition 2019; 67-68:110515. [PMID: 31476571 DOI: 10.1016/j.nut.2019.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/21/2019] [Accepted: 03/01/2019] [Indexed: 02/08/2023]
|
35
|
Hanslin K, Sjölin J, Skorup P, Wilske F, Frithiof R, Larsson A, Castegren M, Tano E, Lipcsey M. The impact of the systemic inflammatory response on hepatic bacterial elimination in experimental abdominal sepsis. Intensive Care Med Exp 2019; 7:52. [PMID: 31456116 PMCID: PMC6712186 DOI: 10.1186/s40635-019-0266-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 08/19/2019] [Indexed: 01/29/2023] Open
Abstract
Background Bacterial translocation from the gut has been suggested to induce a systemic inflammatory response syndrome (SIRS) and organ dysfunction. The liver has a pivotal role in eliminating circulating bacteria entering from the gut. We investigated whether pre-existing inflammation affects hepatic bacterial elimination. Methods Fifteen anaesthetised piglets were infused with E. coli in the portal vein for 3 h. The naive group (n = 6) received the bacterial infusion without endotoxin exposure. SIRS (SIRS group, n = 6) was induced by endotoxin infusion 24 h before the bacterial infusion. For effects of anaesthesia, controls (n = 3) received saline instead of endotoxin for 24 h. Bacterial counts and endotoxin levels in the portal and hepatic veins were analysed during bacterial infusion. Results The bacterial killing rate was higher in the naive group compared with the SIRS group (p = 0.001). The ratio of hepatic to portal venous bacterial counts, i.e. the median bacterial influx from the splanchnic circulation, was 0.06 (IQR 0.01–0.11) in the naive group and 0.71 (0.03–1.77) in the SIRS group at 3 h, and a magnitude lower in the naive group during bacteraemia (p = 0.03). Similar results were seen for hepatic endotoxin elimination. Peak log tumour necrosis factor alpha was higher in the naive 4.84 (4.77–4.89) vs. the SIRS group 3.27 (3.26–3.32) mg/L (p < 0.001). Conclusions Our results suggest that hepatic bacterial and endotoxin elimination is impaired in pigs with pre-existing SIRS while the inflammatory response to bacterial infusion is diminished. If similar mechanisms operate in human critical illness, the hepatic elimination of bacteria from the gut could be impaired by SIRS. Electronic supplementary material The online version of this article (10.1186/s40635-019-0266-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katja Hanslin
- Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan Sjölin
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Paul Skorup
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Frida Wilske
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Robert Frithiof
- Hedenstierna Laboratory, CIRRUS, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Anders Larsson
- Section of Clinical Chemistry, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Markus Castegren
- Section of Infectious Diseases, Department of Medical Sciences, Uppsala University, Uppsala, Sweden.,Perioperative Medicine and Intensive Care, Karolinska University Hospital and CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Eva Tano
- Section of Clinical Bacteriology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Miklos Lipcsey
- Hedenstierna Laboratory, CIRRUS, Anesthesiology and Intensive Care, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
36
|
Emphysematous Aortitis due to Clostridium septicum in an 89-Year-Old Female with Ileus. Case Rep Infect Dis 2019; 2019:1094837. [PMID: 31534806 PMCID: PMC6732609 DOI: 10.1155/2019/1094837] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/22/2019] [Accepted: 07/21/2019] [Indexed: 11/18/2022] Open
Abstract
Emphysematous aortitis is a rare but lethal form of infectious vasculitis. This condition was found incidentally on computed tomography of the chest during the evaluation of a patient presenting with pneumonia coincident with adynamic ileus. The patient did not have a history of malignancy. While colon cancer could not be ruled out, it is possible that ileus may have contributed to or resulted in bacterial translocation in this case. Appropriate investigations and empirical therapy against Clostridium septicum should be initiated in the presence of clinical and radiological findings suggestive of emphysematous aortitis.
Collapse
|
37
|
Antonini M, Lo Conte M, Sorini C, Falcone M. How the Interplay Between the Commensal Microbiota, Gut Barrier Integrity, and Mucosal Immunity Regulates Brain Autoimmunity. Front Immunol 2019; 10:1937. [PMID: 31475000 PMCID: PMC6706873 DOI: 10.3389/fimmu.2019.01937] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/30/2019] [Indexed: 12/21/2022] Open
Abstract
The intestinal barrier provides the host with a strong defense line against the external environment playing also a pivotal role in the crosstalk between the gut microbiota and the immune system. Notably, increasing lines of evidence concerning autoimmune disorders such as Multiple Sclerosis (MS) report an imbalance in both intestinal microbiota composition and mucosal immunity activation, along with an alteration of gut barrier permeability, suggesting this complex network plays a crucial role in modulating the course of autoimmune responses occurring in tissues outside the gut such as the central nervous system (CNS). Here, we review current knowledge on how gut inflammation and breakage of gut barrier integrity modulates the interplay between the commensal gut microbiota and the immune system and its role in shaping brain immunity.
Collapse
Affiliation(s)
- Martina Antonini
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Lo Conte
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Sorini
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Marika Falcone
- Experimental Diabetes Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
38
|
Wang C, Li Q, Ren J. Microbiota-Immune Interaction in the Pathogenesis of Gut-Derived Infection. Front Immunol 2019; 10:1873. [PMID: 31456801 PMCID: PMC6698791 DOI: 10.3389/fimmu.2019.01873] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Gut-derived infection is among the most common complications in patients who underwent severe trauma, serious burn, major surgery, hemorrhagic shock or severe acute pancreatitis (SAP). It could cause sepsis and multiple organ dysfunction syndrome (MODS), which are regarded as a leading cause of mortality in these cases. Gut-derived infection is commonly caused by pathological translocation of intestinal bacteria or endotoxins, resulting from the dysfunction of the gut barrier. In the last decades, the studies regarding to the pathogenesis of gut-derived infection mainly focused on the breakdown of intestinal epithelial tight junction and increased permeability. Limited information is available on the roles of intestinal microbial barrier in the development of gut-derived infection. Recently, advances of next-generation DNA sequencing techniques and its utilization has revolutionized the gut microecology, leading to novel views into the composition of the intestinal microbiota and its connections with multiple diseases. Here, we reviewed the recent progress in the research field of intestinal barrier disruption and gut-derived infection, mainly through the perspectives of the dysbiosis of intestinal microbiota and its interaction with intestinal mucosal immune cells. This review presents novel insights into how the gut microbiota collaborates with mucosal immune cells to involve the development of pathological bacterial translocation. The data might have important implication to better understand the mechanism underlying pathological bacterial translocation, contributing us to develop new strategies for prevention and treatment of gut-derived sepsis.
Collapse
Affiliation(s)
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
39
|
Post-surgical immune suppression: another target to improve postoperative outcomes. J Anesth 2019; 33:625-627. [PMID: 31062095 PMCID: PMC6874620 DOI: 10.1007/s00540-019-02651-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 11/30/2022]
|
40
|
Subramanya SH, Amberpet R, Chaudhary D, Nayak N, Padukone S, Bairy I, Gokhale S. Neonatal sepsis due to glycopeptide resistant Enterococcus faecium from colonized maternal gut- rare case evidence. Antimicrob Resist Infect Control 2019; 8:29. [PMID: 30774945 PMCID: PMC6368750 DOI: 10.1186/s13756-019-0490-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 02/03/2019] [Indexed: 11/13/2022] Open
Abstract
Background Vancomycin-resistant enterococcal infections in the neonatal ICU are growing global problems. We report a case of neonatal septicemia by multidrug-resistant vancomycin-resistant Enterococcus faecium (VRE), the source of infection being the mother's gut. Case presentation A newborn male child admitted to the neonatal intensive care unit (NICU) was diagnosed to have mild meconium aspiration syndrome, early onset neonatal septicemia, and bacteremia by multidrug and vancomycin-resistant Enterococcus faecium. Screening of gut flora of the baby and the mother were carried out to trace the source of infection. Stool cultures of the mother and the baby yielded Vancomycin-Resistant Enterococcus faecium. All three isolates of Enterococcus faecium had similar antibiogram, harbored the vanA gene and similar pulsed-field gel electrophoresis pattern. Baby responded to the 1 week therapy with oral linezolid suspension 20 mg/kg/day, 1 ml/t.d.s. No VRE was isolated from baby on a repeat stool culture 1 week after the linezolid therapy. He was discharged with the advice for the continuance of linezolid for seven more days. Conclusion Isolation of MDR-VRE from the blood culture of the baby and stool specimens of the mother and the baby with the same antibiogram profile and clonal similarities reveals that maternal gut colonization was responsible for neonatal sepsis. Optimal infection control measures and the development of guidelines for monitoring VRE colonization in pregnant women might be useful in reducing the occurrence of neonatal sepsis.
Collapse
Affiliation(s)
| | - Rajesh Amberpet
- Melaka Manipal Medical College, Manipal, India
- Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, India
| | - Dinesh Chaudhary
- Department of Medical Microbiology, Manipal College of Medical Sciences, Pokhara, Nepal
| | - Niranjan Nayak
- Department of Medical Microbiology, Manipal College of Medical Sciences, Pokhara, Nepal
| | - Shashiraja Padukone
- Melaka Manipal Medical College, Manipal, India
- Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, India
| | | | - Shishir Gokhale
- Department of Medical Microbiology, Manipal College of Medical Sciences, Pokhara, Nepal
| |
Collapse
|
41
|
Pathophysiology of Acute Illness and Injury. OPERATIVE TECHNIQUES AND RECENT ADVANCES IN ACUTE CARE AND EMERGENCY SURGERY 2019. [PMCID: PMC7122041 DOI: 10.1007/978-3-319-95114-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pathophysiology of acute illness and injury recognizes three main effectors: infection, trauma, and ischemia-reperfusion injury. Each of them can act by itself or in combination with the other two in developing a systemic inflammatory reaction syndrome (SIRS) that is a generalized reaction to the morbid event. The time course of SIRS is variable and influenced by the number and severity of subsequent insults (e.g., reparative surgery, acquired hospital infections). It occurs simultaneously with a complex of counter-regulatory mechanisms (compensatory anti-inflammatory response syndrome, CARS) that limit the aggressive effects of SIRS. In adjunct, a progressive dysfunction of the acquired (lymphocytes) immune system develops with increased risk for immunoparalysis and associated infectious complications. Both humoral and cellular effectors participate to the development of SIRS and CARS. The most important humoral mediators are pro-inflammatory (IL-1β, IL-6, IL-8, IL-12) and anti-inflammatory (IL-4, IL-10) cytokines and chemokines, complement, leukotrienes, and PAF. Effector cells include neutrophils, monocytes, macrophages, lymphocytes, and endothelial cells. The endothelium is a key factor for production of remote organ damage as it exerts potent chemo-attracting effects on inflammatory cells, allows for leukocyte trafficking into tissues and organs, and promotes further inflammation by cytokines release. Moreover, the loss of vasoregulatory properties and the increased permeability contribute to the development of hypotension and tissue edema. Finally, the disseminated activation of the coagulation cascade causes the widespread deposition of microthrombi with resulting maldistribution of capillary blood flow and ultimately hypoxic cellular damage. This mechanism together with increased vascular permeability and vasodilation is responsible for the development of the multiple organ dysfunction syndrome (MODS).
Collapse
|
42
|
Fadlallah J, Sterlin D, Fieschi C, Parizot C, Dorgham K, El Kafsi H, Autaa G, Ghillani-Dalbin P, Juste C, Lepage P, Malphettes M, Galicier L, Boutboul D, Clément K, André S, Marquet F, Tresallet C, Mathian A, Miyara M, Oksenhendler E, Amoura Z, Yssel H, Larsen M, Gorochov G. Synergistic convergence of microbiota-specific systemic IgG and secretory IgA. J Allergy Clin Immunol 2018; 143:1575-1585.e4. [PMID: 30554723 DOI: 10.1016/j.jaci.2018.09.036] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 08/24/2018] [Accepted: 09/04/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND Commensals induce local IgA responses essential to the induction of tolerance to gut microbiota, but it remains unclear whether antimicrobiota responses remain confined to the gut. OBJECTIVE The aim of this study was to investigate systemic and intestinal responses against the whole microbiota under homeostatic conditions and in the absence of IgA. METHODS We analyzed blood and feces from healthy donors, patients with selective IgA deficiency (SIgAd), and patients with common variable immunodeficiency (CVID). Immunoglobulin-coated bacterial repertoires were analyzed by using combined bacterial fluorescence-activated cell sorting and 16S rRNA sequencing. Bacterial lysates were probed by using Western blot analysis with healthy donor sera. RESULTS Although absent from the healthy gut, serum antimicrobiota IgG are present in healthy subjects and increased in patients with SIgAd. IgG converges with nonoverlapping secretory IgA specificities to target the same bacteria. Each individual subject targets a diverse microbiota repertoire with a proportion that correlates inversely with systemic inflammation. Finally, intravenous immunoglobulin preparations target CVID gut microbiota much less efficiently than healthy microbiota. CONCLUSION Secretory IgA and systemic IgG converge to target gut microbiota at the cellular level. SIgAd-associated inflammation is inversely correlated with systemic anticommensal IgG responses, which might serve as a second line of defense. We speculate that patients with SIgAd could benefit from oral IgA supplementation. Our data also suggest that intravenous immunoglobulin preparations can be supplemented with IgG from IgA-deficient patient pools to offer better protection against gut bacterial translocations in patients with CVID.
Collapse
Affiliation(s)
- Jehane Fadlallah
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France; Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Delphine Sterlin
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Claire Fieschi
- Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Christophe Parizot
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Karim Dorgham
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Hela El Kafsi
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Gaëlle Autaa
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Pascale Ghillani-Dalbin
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Catherine Juste
- Micalis Institute, INRA, AgroParisTech, Universite Paris-Saclay, 78350 Jouy-en-Josas, Paris, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, Universite Paris-Saclay, 78350 Jouy-en-Josas, Paris, France
| | - Marion Malphettes
- Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Lionel Galicier
- Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - David Boutboul
- Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Karine Clément
- Sorbonne Universite, INSERM, UMRS1166, NutriOmics team, ICAN, Paris, France; Assistance Publique Hôpitaux de Paris, Nutrition department, Pitié-Salpêtrière Hospital, Paris, France
| | - Sébastien André
- Sorbonne Universite, INSERM, UMRS1166, NutriOmics team, ICAN, Paris, France
| | - Florian Marquet
- Sorbonne Universite, INSERM, UMRS1166, NutriOmics team, ICAN, Paris, France
| | - Christophe Tresallet
- Department of Surgery, Assistance Publique Hôpitaux de Paris, Pitiè-Salpêtrière Hospital, Paris, France
| | - Alexis Mathian
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Makoto Miyara
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Eric Oksenhendler
- Université Paris Diderot Paris 7, Department of Clinical Immunology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), Paris, France
| | - Zahir Amoura
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Hans Yssel
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Martin Larsen
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France.
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Assistance Publique-Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France.
| |
Collapse
|
43
|
Hurni Y, Bonollo M, Ferrero L, Taraschi G, Canonica C, Venturelli Reyes Lozano S. Pyosalpinx complicating chronic hydrosalpinx in a 50-year old virgo woman: a case report. BMC WOMENS HEALTH 2018; 18:90. [PMID: 29890968 PMCID: PMC5996547 DOI: 10.1186/s12905-018-0583-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/05/2018] [Indexed: 11/23/2022]
Abstract
Background Pelvic inflammatory disease is an infection of the upper genital tract, including the uterus, ovaries, uterine tubes, and pelvic peritoneum. Tubo-ovarian abscess and pyosalpinx are common complications associated with pelvic inflammatory disease. They are usually encountered in sexually active women, but rare cases in Virgos have also been described. Case presentation Here, we report the case of a 50-year-old Virgo woman presenting with pyosalpinx secondary to previous laparotomic sigmoidectomy for acute diverticulitis. Inflammation caused by the woman’s diverticulitis and laparotomic surgery could have been the origin of her left uterine tube occlusion and consequent hydrosalpinx development. The contact between the rectum and left uterine tube observed in our patient suggests that superinfection of the hydrosalpinx could have occurred secondary to bacterial translocation. The patient’s condition was managed with laparoscopic left salpingectomy and antibiotic therapy, which resulted in complete resolution. Conclusions Regardless of sexual history, pelvic inflammatory disease should be considered in all women with abdominal pain. Diagnosing pelvic inflammatory disease in Virgos could be very challenging, but its recognition and appropriate treatment are indispensable because of the potential long-term complications.
Collapse
Affiliation(s)
- Yannick Hurni
- Department of Obstetric and Gynecology, Ospedale Regionale Bellinzona e Valli, 6500, Bellinzona, Switzerland.
| | - Marta Bonollo
- Department of Obstetric and Gynecology, Ospedale Regionale Bellinzona e Valli, 6500, Bellinzona, Switzerland
| | - Ludovica Ferrero
- Department of Obstetric and Gynecology, Ospedale Regionale Bellinzona e Valli, 6500, Bellinzona, Switzerland
| | - Gianmarco Taraschi
- Department of Obstetric and Gynecology, Ospedale Regionale Bellinzona e Valli, 6500, Bellinzona, Switzerland
| | - Claudia Canonica
- Department of Obstetric and Gynecology, Ospedale Regionale Bellinzona e Valli, 6500, Bellinzona, Switzerland
| | | |
Collapse
|
44
|
Autologous white blood cell infusion for trauma, brain trauma, stroke and select immune dysfunction co-morbidities: A promising and timely proposal? Med Hypotheses 2018; 117:7-15. [PMID: 30077201 DOI: 10.1016/j.mehy.2018.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/23/2018] [Accepted: 05/14/2018] [Indexed: 11/21/2022]
Abstract
All traumas suppress the immune system, resulting in higher morbidity and mortality. Infections, poor nutritional status, chronic illness, fatigue, therapies or procedures performed during and after transport also negatively affect the immune system. Large populations are impacted by trauma worldwide and suffer enormous costs in both direct and indirect expenditures from physical, psychological and functional losses. Most therapies and studies of trauma, brain trauma, stroke, immune suppression and their co-morbidities do not address nor discuss methods that promote immune system resuscitation or efficacy to support its role in post-trauma healing and rehabilitation. These omissions present an opportunity for using autologous stored naïve (unexposed to the current trauma and co-morbidities) white blood cell infusions (autologous white blood cell infusion) (AWBCI) to supplement treatment of most traumas, trauma-associated infections, other co-morbidities and immune suppression derived problems in order to improve the global standard of trauma care. We hypothesize to give the traumatized patients back their own immune system that has been 'stored' in some fashion, either cryogenically or just after or during the trauma event [surgery, etc for example]. We emphasize that other treatments should not be replaced - rather we suggest AWBCI as concurrent therapy. We present focused select animal and human studies as proofs of concept to arrive at and support our therapeutic suggestion and hypotheses, flowing historically from donor white blood cell therapy [DLI] to close cohort white blood cell therapy to autologous white blood cell infusion [AWBCI]. We integrate the concept of personalized medicine from an evidence-based framework while maintaining scientific rigor and statistical proof as a basis of our hypotheses.
Collapse
|
45
|
Does bacterial translocation influence the postoperative infections in splenectomized patients after abdominal trauma? Surgeon 2018; 16:94-100. [DOI: 10.1016/j.surge.2016.09.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 09/05/2016] [Accepted: 09/10/2016] [Indexed: 01/14/2023]
|
46
|
Wan MLY, Ling KH, El-Nezami H, Wang MF. Influence of functional food components on gut health. Crit Rev Food Sci Nutr 2018; 59:1927-1936. [DOI: 10.1080/10408398.2018.1433629] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Murphy L. Y. Wan
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences Building, The University of Hong Kong, Pokfulam, Hong Kong
| | - K. H. Ling
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences Building, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hani El-Nezami
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences Building, The University of Hong Kong, Pokfulam, Hong Kong
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - M. F. Wang
- School of Biological Sciences, Faculty of Science, Kadoorie Biological Sciences Building, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
47
|
Li Q, Wang C, Tang C, Zhao X, He Q, Li J. Identification and Characterization of Blood and Neutrophil-Associated Microbiomes in Patients with Severe Acute Pancreatitis Using Next-Generation Sequencing. Front Cell Infect Microbiol 2018; 8:5. [PMID: 29423379 PMCID: PMC5790034 DOI: 10.3389/fcimb.2018.00005] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 01/09/2018] [Indexed: 01/28/2023] Open
Abstract
Infectious complications are a leading cause of death for patients with severe acute pancreatitis (SAP). Yet, our knowledge about details of the blood microbial landscape in SAP patients remains limited. Recently, some studies have reported that the peripheral circulation harbors a diverse bacterial community in healthy and septic subjects. The objective of this study was to examine the presence of the blood bacterial microbiome in SAP patients and its potential role in the development of infectious complications. Here we conducted a prospective observational study on a cohort of 50 SAP patients and 12 healthy subjects to profile the bacterial composition in the blood. The patients were subgrouped into uninfected (n = 17), infected (n = 16), and septic (n = 17) cases. Applying 16S rDNA-based next-generation sequencing technique, we investigated blood and neutrophil-associated microbiomes in SAP patients, and assessed their connections with immunological alterations. Based on the sequencing data, a diverse bacterial microbiota was found in peripheral blood and neutrophils from the healthy and SAP subjects. As compared to healthy controls, the blood and neutrophil-associated microbiomes in the patients were significantly altered, with an expansion in Bacteroidetes and Firmicutes as well as a decrease in Actinobacteria. Variations in the microbiome composition in patients were associated with immunological disorders, including altered lymphocyte subgroups, elevated levels of serum cytokines and altered proteomic profiles of neutrophils. However, no significant compositional difference was observed between the patient subgroups, implying that the microbiota alterations might not be linked to presence/absence of infectious complications in SAP. Together, we present an initial description of the blood and neutrophil-associated bacterial profiles in SAP patients, offering novel evidence for the existence of the blood microbiome. Identification of the blood microbiome provides novel insights into characteristics and diagnostics of bacteremia in the patients. Further study is required to assess the possible implications of the blood microbiome in health and diseases.
Collapse
Affiliation(s)
- Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Chenyang Wang
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Chun Tang
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Xiaofan Zhao
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Qin He
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Jieshou Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
48
|
Physiological Translocation of Lactic Acid Bacteria during Pregnancy Contributes to the Composition of the Milk Microbiota in Mice. Nutrients 2017; 10:nu10010014. [PMID: 29295502 PMCID: PMC5793242 DOI: 10.3390/nu10010014] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/24/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023] Open
Abstract
The human milk microbiota is a complex and diverse ecosystem that seems to play a relevant role in the mother-to-infant transmission of microorganisms during early life. Bacteria present in human milk may arise from different sources, and recent studies suggest that at least some of them may be originally present in the maternal digestive tract and may reach the mammary gland through an endogenous route during pregnancy and lactation. The objective of this work was to elucidate whether some lactic acid bacteria are able to translocate and colonize the mammary gland and milk. For this purpose, two lactic acid bacteria strains (Lactococcus lactis MG1614 and Lactobacillus salivarius PS2) were transformed with a plasmid containing the lux genes; subsequently, the transformed strains were orally administered to pregnant mice. The murine model allowed the visualization, isolation, and Polymerase Chain Reaction (PCR)-detection of the transformed bacteria in different body locations, including mammary tissue and milk, reinforcing the hypothesis that physiological translocation of maternal bacteria during pregnancy and lactation may contribute to the composition of the mammary and milk microbiota.
Collapse
|
49
|
Role for FimH in Extraintestinal Pathogenic Escherichia coli Invasion and Translocation through the Intestinal Epithelium. Infect Immun 2017; 85:IAI.00581-17. [PMID: 28808163 DOI: 10.1128/iai.00581-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/27/2022] Open
Abstract
The translocation of bacteria across the intestinal epithelium of immunocompromised patients can lead to bacteremia and life-threatening sepsis. Extraintestinal pathogenic Escherichia coli (ExPEC), so named because this pathotype infects tissues distal to the intestinal tract, is a frequent cause of such infections, is often multidrug resistant, and chronically colonizes a sizable portion of the healthy population. Although several virulence factors and their roles in pathogenesis are well described for ExPEC strains that cause urinary tract infections and meningitis, they have not been linked to translocation through intestinal barriers, a fundamentally distant yet important clinical phenomenon. Using untransformed ex situ human intestinal enteroids and transformed Caco-2 cells, we report that ExPEC strain CP9 binds to and invades the intestinal epithelium. ExPEC harboring a deletion of the gene encoding the mannose-binding type 1 pilus tip protein FimH demonstrated reduced binding and invasion compared to strains lacking known E. coli virulence factors. Furthermore, in a murine model of chemotherapy-induced translocation, ExPEC lacking fimH colonized at levels comparable to that of the wild type but demonstrated a statistically significant reduction in translocation to the kidneys, spleen, and lungs. Collectively, this study indicates that FimH is important for ExPEC translocation, suggesting that the type 1 pilus is a therapeutic target for the prevention of this process. Our study also highlights the use of human intestinal enteroids in the study of enteric diseases.
Collapse
|
50
|
Nascimento BB, Cartelle CT, Noviello MDL, Pinheiro BV, de Almeida Vitor RW, Souza DDG, de Vasconcelos Generoso S, Cardoso VN, Martins FDS, Nicoli JR, Arantes RME. Influence of indigenous microbiota on experimental toxoplasmosis in conventional and germ-free mice. Int J Exp Pathol 2017; 98:191-202. [PMID: 28895246 DOI: 10.1111/iep.12236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 06/04/2017] [Indexed: 01/12/2023] Open
Abstract
Toxoplasmosis represents one of the most common zoonoses worldwide. Its agent, Toxoplasma gondii, causes a severe innate pro-inflammatory response. The indigenous intestinal microbiota promotes host animal homoeostasis and may protect the host against pathogens. Germ-free (GF) animals provide an important tool for the study of interactions between host and microbiota. In this study, we assessed the role of indigenous microorganisms in disease development utilizing a murine toxoplasmosis model, which includes conventional (CV) and GF NIH Swiss mice. CV and GF mice orally inoculated with T. gondii had similar survival curves. However, disease developed differently in the two animal groups. In CV mice, intestinal permeability increased and levels of intestinal pro-inflammatory cytokines were altered. In GF animals, there were discrete epithelial degenerative changes and mucosal oedema, but the liver and lungs displayed significant lesions. We conclude that, despite similar survival curves, CV animals succumb to an exaggerated inflammatory response, whereas GF mice fail to produce an adequate systemic response.
Collapse
Affiliation(s)
- Bruna B Nascimento
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Christiane T Cartelle
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria de L Noviello
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Breno V Pinheiro
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo W de Almeida Vitor
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Danielle da G Souza
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Simone de Vasconcelos Generoso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Valbert N Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Flaviano Dos S Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jacques R Nicoli
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rosa M E Arantes
- Departamento de Patologia Geral, Laboratório de Neuro-Imunopatologia Experimental (NIPE), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|