1
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. Gut Microbes 2025; 17:2458189. [PMID: 39930324 PMCID: PMC11817531 DOI: 10.1080/19490976.2025.2458189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal growth barrier dysfunction, and aberrant inflammatory responses. Further, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, mostly commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no U.S. Food and Drug Administration (FDA) approved countermeasures that can treat radiation-induced GI injuries. To meet this critical need, Synedgen Inc. has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract, which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy partial body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, Vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory responses mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera while suppressing potentially pathogenic bacteria Enterococcus and Staphylococcus compared with Vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and reducing pro-inflammatory responses. Further development of this drug as an FDA-approved medical countermeasure is of critical importance.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
2
|
Liu S, Wang Q, Luo W, Huang L, Li L, Wu Y, Cai W, Hong J, Philips A, Fernig D, Sutton R, Windsor J, Szatmary P, Liu T, Huang W, Xia Q. Histones are critical toxic factors in gut lymph of severe acute pancreatitis: Neutralization by baicalin and baicalein for protection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156474. [PMID: 39954616 DOI: 10.1016/j.phymed.2025.156474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Whether circulating histones in gut lymph contribute to organ failure and impact of chaiqin chengqi decoction (CQCQD) on histones in severe acute pancreatitis (SAP) remain elusive. PURPOSE To verify the role of histones in gut lymph of SAP and evaluate the effect of the CQCQD on them. METHODS Sodium taurocholate was retrogradely infused into pancreatobiliary duct to induce SAP in rodents. Various regimens of CQCQD were administered intragastrically or via duodenum followed by dynamic gut lymph collection in rats. The impact of gut lymph and histones on endothelial cell viability and lymphocytes was determined. Components of CQCQD in gut lymph were identified by UHPLC-MS and their binding activities with histones were quantified by biolayer interferometry followed by validation in vitro and in vivo in mice. RESULTS The histone level was significantly increased in gut lymph of SAP at various time points assessed, closely correlating with multiple organ injury (MOI) indices and contemporary cell viability. Inhibition of histones reduced cytotoxicity induced by SAP-conditioned gut lymph. CQCQD reduced apoptotic cell death in mesenteric lymph nodes, histone level, and cytotoxicity of gut lymph, alleviating MOI parameters. Baicalin and baicalein were amongst top 13 identified CQCQD components absorbed into gut lymph to actively bind histones, block membrane disruption and calcium influx of lymphocytes, and inhibit their cytotoxicity. Both baicalin and baicalein mitigated histone- and SAP-induced MOI indices in mice. CONCLUSION Histones are key toxic factors in the gut lymph of SAP and their antagonism by baicalin and baicalein offers a novel therapeutic strategy.
Collapse
Affiliation(s)
- Shiyu Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qiqi Wang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wenjuan Luo
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lijia Huang
- West China Biobank, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lan Li
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Yongzi Wu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Wenhao Cai
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Jiwon Hong
- Department of Surgery, Faculty of Medicine and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Anthony Philips
- Department of Surgery, Faculty of Medicine and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - David Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool L69 3GA, United Kingdom
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GA, United Kingdom
| | - John Windsor
- Department of Surgery, Faculty of Medicine and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | - Peter Szatmary
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GA, United Kingdom
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China; West China Biobank, West China Hospital, Sichuan University, Chengdu 610041, PR China; Institute for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Liu X, Ji Y, Lv H, Lv Z, Guo Y, Nie W. Microbiome and metabolome reveal beneficial effects of baicalin on broiler growth performance and intestinal health. Poult Sci 2025; 104:104678. [PMID: 39721274 PMCID: PMC11732485 DOI: 10.1016/j.psj.2024.104678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/28/2024] Open
Abstract
Normal function and health of the intestinal tract were necessary for the growth and development of broilers. Baicalin (BA) possessed a variety of biological activities. The objective of this study was to examine the impact of BA on the growth performance, intestinal barrier function, intestinal microbiota, and mucosal metabolism in broilers. A total of 720 21-day-old broilers were randomly allocated into 3 groups and fed with either basal diet (Con group) or basal diet supplemented with 6 or 12 mg/kg baicalin (BA6 and BA12 groups) for a continuous feeding period of 40 days. Results showed that BA had a trend towards improving (P = 0.086) the 60-day body weight of broilers, and the BA12 group exhibited significantly higher (P < 0.05) average daily gain from day 39 to 60 compared to the Con group. Additionally, in the BA12 group, the ratio of villus height to crypt depth and the expression levels of tight junction protein-related genes significantly increased (P < 0.05), while intestinal permeability significantly decreased (P < 0.05). Supplementation with 12 mg/kg BA significantly enhanced antioxidant capacity, promoted (P < 0.05) crypt proliferation, increased (P < 0.05) immunoglobulin levels, upregulated (P < 0.05) IL-2 and IL-8 mRNA levels, and downregulated (P < 0.05) IL-4 and TGF-β2 mRNA levels. Metabolomics analysis revealed that BA improved the metabolic characteristics of intestinal mucosa, significantly upregulating pathways associated with ascorbate and aldarate metabolism, glyoxylate and dicarboxylate metabolism, phosphatidylinositol signaling system, alpha-linolenic acid metabolism, and galactose metabolism. 16S rRNA sequencing results indicated that BA increased the richness of intestinal microbiota community and the relative abundance of Actinobacteria phylum, while reducing the relative abundance of contains mobile elements, potentially pathogenic, and facultatively anaerobic. Overall, 12 mg/kg BA improved intestinal health by modulating intestinal barrier function, antioxidant capacity, immunity, intestinal microbiota, and intestinal mucosal metabolism levels, ultimately enhancing broiler growth performance.
Collapse
Affiliation(s)
- Xingbo Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yunru Ji
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Huiyuan Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China; Beijing Centre Biology Co., Ltd., Beijing, 102600, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wei Nie
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
4
|
Kiselev VV, Zhigalova MS, Yartsev PA, Novikov SV, Kuzmin AM, Tkeshelashvili DT. [Relationship between intestinal insufficiency syndrome and risk of external intestinal fistulas in patients with severe acute pancreatitis]. Khirurgiia (Mosk) 2025:79-85. [PMID: 40203175 DOI: 10.17116/hirurgia202504179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
OBJECTIVE To identify the risk factors of external intestinal fistulas in patients with severe acute pancreatitis (SAP) and pancreatic necrosis. MATERIAL AND METHODS A retrospective and prospective study included 537 patients (354 (65.9%) men and 183 (34.1%) women) with SAP. Mean age was 51.2±18.5 years. To assess the effectiveness of intensive therapy, patients were divided into 2 groups. The control group (n=207) included patients who underwent examination and treatment according to the national guidelines «Acute pancreatitis», 2020). In the main group (n=330), examination and treatment were supplemented with original protocol. RESULTS. P Atients with intestinal failure score< 5 had no purulent-septic complications and multiple organ failure. Among patients with 6-9 scores, purulent-septic complications were observed in 11.7% of cases, multiple organ failure - 14.8% of cases. Among patients with 10-12 scores, the incidence of purulent-septic complications was 24.6%, multiple organ failure - 30% of cases. Thus, intestinal failure score > 10 is an important prognostic criterion of purulent-septic complications and multiple organ failure. In addition, external intestinal fistulas occurred in 8.5% of patients with intestinal failure score > 10. CONCLUSION. P Rogression of intestinal failure correlates with purulent-septic complications and external intestinal fistulas in patients with SAP and pancreatic necrosis. Original diagnostic algorithm is valuable for early detection of functional gastrointestinal disorders, severity and localization of these changes. Original scoring system makes it possible to predict the risk of purulent-septic complications and multiple organ failure on the first day after admission. Moreover, timely therapy improves treatment outcomes in patients with SAP. In our study, the incidence of purulent-septic complications including external intestinal fistulas decreased from 10% to 7% in the main group.
Collapse
Affiliation(s)
- V V Kiselev
- Sklifosovsky Research Institute for Emergency Care, Moscow, Russia
| | - M S Zhigalova
- Sklifosovsky Research Institute for Emergency Care, Moscow, Russia
| | - P A Yartsev
- Sklifosovsky Research Institute for Emergency Care, Moscow, Russia
| | - S V Novikov
- Sklifosovsky Research Institute for Emergency Care, Moscow, Russia
| | - A M Kuzmin
- Sklifosovsky Research Institute for Emergency Care, Moscow, Russia
| | | |
Collapse
|
5
|
Moreno RJ, Abu Amara R, Ashwood P. Toward a better understanding of T cell dysregulation in autism: An integrative review. Brain Behav Immun 2025; 123:1147-1158. [PMID: 39378971 DOI: 10.1016/j.bbi.2024.10.009] [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: 05/23/2024] [Revised: 08/28/2024] [Accepted: 10/05/2024] [Indexed: 10/10/2024] Open
Abstract
Autism spectrum disorder (ASD) is a highly heterogeneous disorder characterized by impairments in social, communicative, and restrictive behaviors. Over the past 20 years, research has highlighted the role of the immune system in regulating neurodevelopment and behavior. In ASD, immune abnormalities are frequently observed, such as elevations in pro-inflammatory cytokines, alterations in immune cell frequencies, and dysregulated mechanisms of immune suppression. The adaptive immune system - the branch of the immune system conferring cellular immunity - may be involved in the etiology of ASD. Specifically, dysregulated T cell activity, characterized by altered cellular function and increased cytokine release, presence of inflammatory phenotypes and altered cellular signaling, has been consistently observed in several studies across multiple laboratories and geographic regions. Similarly, mechanisms regulating their activation are also disrupted. T cells at homeostasis coordinate the healthy development of the central nervous system (CNS) during early prenatal and postnatal development, and aid in CNS maintenance into adulthood. Thus, T cell dysregulation may play a role in neurodevelopment and the behavioral and cognitive manifestations observed in ASD. Outside of the CNS, aberrant T cell activity may also be responsible for the increased frequency of immune based conditions in the ASD population, such as allergies, gut inflammation and autoimmunity. In this review, we will discuss the current understanding of T cell biology in ASD and speculate on mechanisms behind their dysregulation. This review also evaluates how aberrant T cell biology affects gastrointestinal issues and behavior in the context of ASD.
Collapse
Affiliation(s)
- R J Moreno
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - R Abu Amara
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA
| | - P Ashwood
- Department of Medical Microbiology and Immunology, UC Davis, CA, USA; The M.I.N.D. Institute, University of California at Davis, CA, USA.
| |
Collapse
|
6
|
Li L, Zinger J, Sassen SDT, Juffermans NP, Koch BCP, Endeman H. The relation between inflammatory biomarkers and drug pharmacokinetics in the critically ill patients: a scoping review. Crit Care 2024; 28:376. [PMID: 39563441 PMCID: PMC11577668 DOI: 10.1186/s13054-024-05150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/26/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND The level of inflammation alters drug pharmacokinetics (PK) in critically ill patients. This might compromise treatment efficacy. Understanding the specific effects of inflammation, measured by biomarkers, on drug absorption, distribution, metabolism, and excretion is might help in optimizing dosing strategies. OBJECTIVES This review investigates the relationship between inflammatory biomarkers and PK parameters absorption, distribution, metabolism and excretion (ADME) in critically ill patients, providing insight in the complexity of dosing drugs in critically ill patients. METHOD Following PRISMA guidelines, we conducted a comprehensive search of Medline, Embase, Web of Science, and Cochrane databases (January 1946-November 2023). Studies examining inflammatory biomarkers, PK parameters, or drug exposure in critically ill patients were included. Records were screened by title, abstract, and full text, with any discrepancies resolved through discussion or consultation with a third reviewer. RESULTS Of the 4479 records screened, 31 met our inclusion criteria: 2 on absorption, 7 on distribution, 17 on metabolism, and 6 on excretion. In general, results are only available for a limited number of drugs, and most studies are done only looking at one of the components of ADME. Higher levels of inflammatory biomarkers may increase or decrease drug absorption depending on whether the drug undergoes hepatic first-pass elimination. For drug distribution, inflammation is negatively correlated with drug protein binding capacity, positively correlated with cerebrospinal fluid penetration, and negatively correlated with peritoneal penetration. Metabolizing capacity of most drugs was inversely correlated with inflammatory biomarkers. Regarding excretion, inflammation can lead to reduced drug clearance, except in the neonatal population. CONCLUSION Inflammatory biomarkers can offer valuable information regarding altered PK in critically ill patients. Our findings emphasize the need to consider inflammation-driven PK variability when individualizing drug therapy in this setting, at the same time research is limited to certain drugs and needs further research, also including pharmacodynamics.
Collapse
Affiliation(s)
- Letao Li
- Department of Hospital Pharmacy, Erasmus MC-University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Xinqiao Hospital, Army Medical University, 183 Xinqiao Street, Shapingba District, Chongqing, 400037, China
| | - Julia Zinger
- Department of Hospital Pharmacy, Erasmus MC-University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Sebastiaan D T Sassen
- Department of Hospital Pharmacy, Erasmus MC-University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Nicole P Juffermans
- Department of Intensive Care, Erasmus MC-University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus MC-University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Henrik Endeman
- Department of Intensive Care, Erasmus MC-University Medical Center, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- Department of Intensive Care, OLVG, Oosterpark 9, 1091 AC, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Borges A, Bento L. Organ crosstalk and dysfunction in sepsis. Ann Intensive Care 2024; 14:147. [PMID: 39298039 DOI: 10.1186/s13613-024-01377-0] [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: 03/30/2024] [Accepted: 09/10/2024] [Indexed: 09/21/2024] Open
Abstract
Sepsis is a dysregulated immune response to an infection that leads to organ dysfunction. Sepsis-associated organ dysfunction involves multiple inflammatory mechanisms and complex metabolic reprogramming of cellular function. These mechanisms cooperate through multiple organs and systems according to a complex set of long-distance communications mediated by cellular pathways, solutes, and neurohormonal actions. In sepsis, the concept of organ crosstalk involves the dysregulation of one system, which triggers compensatory mechanisms in other systems that can induce further damage. Despite the abundance of studies published on organ crosstalk in the last decade, there is a need to formulate a more comprehensive framework involving all organs to create a more detailed picture of sepsis. In this paper, we review the literature published on organ crosstalk in the last 10 years and explore how these relationships affect the progression of organ failure in patients with septic shock. We explored these relationships in terms of the heart-kidney-lung, gut-microbiome-liver-brain, and adipose tissue-muscle-bone crosstalk in sepsis patients. A deep connection exists among these organs based on crosstalk. We also review how multiple therapeutic interventions administered in intensive care units, such as mechanical ventilation, antibiotics, anesthesia, nutrition, and proton pump inhibitors, affect these systems and must be carefully considered when managing septic patients. The progression to multiple organ dysfunction syndrome in sepsis patients is still one of the most frequent causes of death in critically ill patients. A better understanding and monitoring of the mechanics of organ crosstalk will enable the anticipation of organ damage and the development of individualized therapeutic strategies.
Collapse
Affiliation(s)
- André Borges
- Intensive Care Unit of Hospital de São José, Unidade de Urgência Médica, Rua José António Serrano, Lisbon, 1150-199, Portugal.
- NOVA Medical School, Campo dos Mártires da Pátria 130, Lisbon, 1169-056, Portugal.
| | - Luís Bento
- Intensive Care Unit of Hospital de São José, Unidade de Urgência Médica, Rua José António Serrano, Lisbon, 1150-199, Portugal
- NOVA Medical School, Campo dos Mártires da Pátria 130, Lisbon, 1169-056, Portugal
| |
Collapse
|
8
|
Wang Y, Deng K, Lin P, Huang L, Hu L, Ye J, Liang J, Ni Y, Tan L. Elevated total bile acid levels as an independent predictor of mortality in pediatric sepsis. Pediatr Res 2024:10.1038/s41390-024-03438-3. [PMID: 39266629 DOI: 10.1038/s41390-024-03438-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/01/2024] [Accepted: 07/12/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND The close relationship between bile acid (BA) metabolism and sepsis has been investigated in recent years, as knowledge of the role of the gut microbiome and metabolomics in sepsis has grown and become more comprehensive. METHODS Patients with sepsis who were admitted to the PICU of the Children's Hospital, Zhejiang University School of Medicine from January 2016 to December 2021 were enrolled in this study. Preoperative non-infectious pediatric patients undergoing elective surgeries in our hospital's department of surgery were recruited as controls during the same period. Clinical data were collected and analyzed. RESULTS 702 children were enrolled, comprising 538 sepsis survivors, 164 sepsis fatalities, and 269 non-infected controls. Statistical analysis revealed that total BA (TBA) increased in both the early and severe stages of pediatric sepsis. In the severe stage, TBA (OR = 2.898, 95% CI 1.946-4.315, p < 0.05) was identified as a risk factor for sepsis. A clinical model identified TBA (the cut-off value is >17.95 µmol/L) as an independent predictor of sepsis mortality with an AUC of 0.842 (95% CI 0.800-0.883), sensitivity of 54.9%, specificity of 96.6%, and HR = 7.658 (95% CI 5.575-10.520). CONCLUSIONS The study showed that elevated TBA was associated with a heightened risk of mortality in pediatric sepsis. IMPACT Many clinical indicators show differences between children with sepsis and the control group, among which the difference in serum total bile acid levels is the most significant. During the hospitalization of the patients, the overall bile acid levels in the sepsis death group were higher and exhibited greater fluctuations compared to the survival group, with significant differences. Serum total bile acid levels can serve as effective biomarker for predicting the prognosis of children with sepsis.
Collapse
Affiliation(s)
- Yanfei Wang
- Department of Surgical ICU, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Kelei Deng
- Department of Surgical ICU, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Peiquan Lin
- Department of Surgical ICU, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Limin Huang
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Lei Hu
- Department of Surgical ICU, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jing Ye
- Department of Surgical ICU, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianfeng Liang
- Department of Medical Statistics, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yan Ni
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| | - Linhua Tan
- Department of Surgical ICU, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
9
|
Suleri A, Gaiser C, Cecil CAM, Dijkzeul A, Neumann A, Labrecque JA, White T, Bergink V, Muetzel RL. Examining longitudinal associations between prenatal exposure to infections and child brain morphology. Brain Behav Immun 2024; 119:965-977. [PMID: 38750701 PMCID: PMC7616133 DOI: 10.1016/j.bbi.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Maternal infection during pregnancy has been identified as a prenatal risk factor for the later development of psychopathology in exposed offspring. Neuroimaging data collected during childhood has suggested a link between prenatal exposure to maternal infection and child brain structure and function, potentially offering a neurobiological explanation for the emergence of psychopathology. Additionally, preclinical studies utilizing repeated measures of neuroimaging data suggest that effects of prenatal maternal infection on the offspring's brain may normalize over time (i.e., catch-up growth). However, it remains unclear whether exposure to prenatal maternal infection in humans is related to long-term differential neurodevelopmental trajectories. Hence, this study aimed to investigate the association between prenatal exposure to infections on child brain development over time using repeated measures MRI data. METHODS We leveraged data from a population-based cohort, Generation R, in which we examined prospectively assessed self-reported infections at each trimester of pregnancy (N = 2,155). We further used three neuroimaging assessments (at mean ages 8, 10 and 14) to obtain cortical and subcortical measures of the offspring's brain morphology with MRI. Hereafter, we applied linear mixed-effects models, adjusting for several confounding factors, to estimate the association of prenatal maternal infection with child brain development over time. RESULTS We found that prenatal exposure to infection in the third trimester was associated with a slower decrease in volumes of the pars orbitalis, rostral anterior cingulate and superior frontal gyrus, and a faster increase in the middle temporal gyrus. In the temporal pole we observed a divergent pattern, specifically showing an increase in volume in offspring exposed to more infections compared to a decrease in volume in offspring exposed to fewer infections. We further observed associations in other frontal and temporal lobe structures after exposure to infections in any trimester, though these did not survive multiple testing correction. CONCLUSIONS Our results suggest that prenatal exposure to infections in the third trimester may be associated with slower age-related growth in the regions: pars orbitalis, rostral anterior cingulate and superior frontal gyrus, and faster age-related growth in the middle temporal gyrus across childhood, suggesting a potential sensitive period. Our results might be interpreted as an extension of longitudinal findings from preclinical studies, indicating that children exposed to prenatal infections could exhibit catch-up growth. However, given the lack of differences in brain volume between various infection groups at baseline, there may instead be either a longitudinal deviation or a subtle temporal deviation. Subsequent well-powered studies that extend into the period of full brain development (∼25 years) are needed to confirm whether the observed phenomenon is indeed catch-up growth, a longitudinal deviation, or a subtle temporal deviation.
Collapse
Affiliation(s)
- Anna Suleri
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Carolin Gaiser
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Neuroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Charlotte A M Cecil
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Biomedical Data Sciences, Molecular Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annet Dijkzeul
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Alexander Neumann
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; The Generation R Study Group, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jeremy A Labrecque
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Tonya White
- Section on Social and Cognitive Developmental Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Veerle Bergink
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, USA; Department of Psychiatry, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Ryan L Muetzel
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, the Netherlands; Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
10
|
Li Z, Xing J, Ma X, Zhang W, Wang C, Wang Y, Qi X, Liu Y, Jian D, Cheng X, Zhu Y, Shi C, Guo Y, Zhao H, Jiang W, Tang H. An orally administered bacterial membrane protein nanodrug ameliorates doxorubicin cardiotoxicity through alleviating impaired intestinal barrier. Bioact Mater 2024; 37:517-532. [PMID: 38698916 PMCID: PMC11063951 DOI: 10.1016/j.bioactmat.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
The cardiotoxicity caused by Dox chemotherapy represents a significant limitation to its clinical application and is a major cause of late death in patients undergoing chemotherapy. Currently, there are no effective treatments available. Our analysis of 295 clinical samples from 132 chemotherapy patients and 163 individuals undergoing physical examination revealed a strong positive correlation between intestinal barrier injury and the development of cardiotoxicity in chemotherapy patients. We developed a novel orally available and intestinal targeting protein nanodrug by assembling membrane protein Amuc_1100 (obtained from intestinal bacteria Akkermansia muciniphila), fluorinated polyetherimide, and hyaluronic acid. The protein nanodrug demonstrated favorable stability against hydrolysis compared with free Amuc_1100. The in vivo results demonstrated that the protein nanodrug can alleviate Dox-induced cardiac toxicity by improving gut microbiota, increasing the proportion of short-chain fatty acid-producing bacteria from the Lachnospiraceae family, and further enhancing the levels of butyrate and pentanoic acids, ultimately regulating the homeostasis repair of lymphocytes in the spleen and heart. Therefore, we believe that the integrity of the intestinal barrier plays an important role in the development of chemotherapy-induced cardiotoxicity. Protective interventions targeting the intestinal barrier may hold promise as a general clinical treatment regimen for reducing Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Junyue Xing
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaohan Ma
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Wanjun Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Chuan Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yingying Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xinkun Qi
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yanhui Liu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Dongdong Jian
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaolei Cheng
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yanjie Zhu
- Department of Pathology, Central Hospital of Kaifeng City, Kaifeng, Henan, 475000, China
| | - Chao Shi
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Yongjun Guo
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| |
Collapse
|
11
|
Hai PD, Tot NH, Thao LT, Khoa Q, Thien DH. Prognostic Value of Acute Gastrointestinal Injury Combined with Disease Severity Scores in Critically Ill Patients. Indian J Crit Care Med 2024; 28:575-580. [PMID: 39130390 PMCID: PMC11310679 DOI: 10.5005/jp-journals-10071-24733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/03/2024] [Indexed: 08/13/2024] Open
Abstract
Background Critically ill patients are at high risk of multiple organ failure syndrome (MODS) and gastrointestinal (GI) injury and dysfunction, which are associated with increased mortality rates. The acute gastrointestinal injury (AGI) scale has shown promise in assessing GI dysfunction. However, the combined utility of AGI with established disease severity scores remains unclear. This study aimed to investigate the performance of AGI in conjunction with modified nutritional risk in critically ill (mNUTRIC), sequential organ failure assessment (SOFA), and acute physiology and chronic health evaluation II (APACHE II) scores for predicting mortality in critically ill patients. Materials and methods A retrospective cross-sectional study was conducted in the intensive care unit (ICU) from May 2021 to December 2021. Demographic and clinical data were collected, including AGI grade, mNUTRIC score, SOFA score, APACHE II score, and mortality. Results Among 93 critically ill patients, AGI was observed in 47.3% of cases, and the in-hospital mortality rate was 30.1%. The area under the curve (AUC) for AGI in predicting in-hospital mortality was 0.67 [95% confidence interval (CI), 0.56, 0.79; p = 0.008], similar to the AUCs of SOFA, APACHE II, and mNUTRIC scores. The combination of AGI with mNUTRIC, APACHE II, or SOFA scores improved the predictive performance compared with AGI alone. Conclusion The AGI grade, in conjunction with disease severity scores, such as mNUTRIC, SOFA, and APACHE II scores, shows promise in predicting mortality in critically ill patients. Integrating AGI into evaluating critically ill patients can enhance prognostic accuracy. How to cite this article Hai PD, Tot NH, Thao LT, Khoa Q, Thien DH. Prognostic Value of Acute Gastrointestinal Injury Combined with Disease Severity Scores in Critically Ill Patients. Indian J Crit Care Med 2024;28(6):575-580.
Collapse
Affiliation(s)
- Pham D Hai
- Medical Intensive Care Unit, 108 Military Central Hospital, Ha Noi, Vietnam
| | - Nguyen H Tot
- Medical Intensive Care Unit, 108 Military Central Hospital, Ha Noi, Vietnam
| | - Le T Thao
- College of Health Sciences, VinUniversity, Ha Noi, Vietnam
| | - Quy Khoa
- College of Health Sciences, VinUniversity, Ha Noi, Vietnam
| | - Dang H Thien
- College of Health Sciences, VinUniversity, Ha Noi, Vietnam
| |
Collapse
|
12
|
Wang Z, Han S, Xiao Y, Zhang Y, Ge Y, Liu X, Gao J. Genetically supported causality between gut microbiota and frailty: a two-sample Mendelian randomization study. Front Microbiol 2024; 15:1324209. [PMID: 38741737 PMCID: PMC11089315 DOI: 10.3389/fmicb.2024.1324209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 03/25/2024] [Indexed: 05/16/2024] Open
Abstract
Background A mounting body of evidence suggests a strong connection between gut microbiota and the risk of frailty. However, the question of causality remains unanswered. In this study, we employed a Mendelian randomization (MR) approach to assess potential causal relationships between gut microbiota and the risk of frailty. Materials and methods Summary statistics for the gut microbiome were obtained from a genome wide association study (GWAS) meta-analysis of the MiBioGen consortium (N = 18,340). Summary statistics for frailty were obtained from a GWAS meta-analysis, including the UK Biobank and TwinGene (N = 175,226). Our primary analysis utilized the inverse variance weighted (IVW) method. To enhance the robustness of our results, we also applied weighted median methods, MR Egger regression, and MR pleiotropy residual sum and outlier test. Finally, we conducted reverse MR analysis to investigate the potential for reverse causality. Results IVW method identified 7 bacterial taxa nominally associated with the risk of FI. Class Bacteroidia (p = 0.033) and genus Eubacterium ruminantium group (p = 0.028) were protective against FI. In addition, class Betaproteobacteria (p = 0.042), genus Allisonella (p = 0.012), genus Bifidobacterium (p = 0.013), genus Clostridium innocuum group (p = 0.036) and genus Eubacterium coprostanoligenes group (p = 0.003) were associated with a higher risk of FI. No pleiotropy or heterogeneity were found. Conclusion The MR analysis indicates a causal relationship between specific gut microbiota and FI, offering new insights into the mechanisms underlying FI mediated by gut microbiota.
Collapse
Affiliation(s)
- Zi Wang
- Yangzhou University Medical College, Yangzhou, China
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Shuai Han
- Yangzhou University Medical College, Yangzhou, China
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Yinggang Xiao
- Yangzhou University Medical College, Yangzhou, China
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Yang Zhang
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Yali Ge
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Xin Liu
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| | - Ju Gao
- Department of Anesthesiology, Institute of Anesthesia, Emergency and Critical Care, Yangzhou University Affiliated Northern Jiangsu People’s Hospital, Yangzhou, China
| |
Collapse
|
13
|
Ziaka M, Exadaktylos A. Pathophysiology of acute lung injury in patients with acute brain injury: the triple-hit hypothesis. Crit Care 2024; 28:71. [PMID: 38454447 PMCID: PMC10918982 DOI: 10.1186/s13054-024-04855-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
It has been convincingly demonstrated in recent years that isolated acute brain injury (ABI) may cause severe dysfunction of peripheral extracranial organs and systems. Of all potential target organs and systems, the lung appears to be the most vulnerable to damage after ABI. The pathophysiology of the bidirectional brain-lung interactions is multifactorial and involves inflammatory cascades, immune suppression, and dysfunction of the autonomic system. Indeed, the systemic effects of inflammatory mediators in patients with ABI create a systemic inflammatory environment ("first hit") that makes extracranial organs vulnerable to secondary procedures that enhance inflammation, such as mechanical ventilation (MV), surgery, and infections ("second hit"). Moreover, accumulating evidence supports the knowledge that gut microbiota constitutes a critical superorganism and an organ on its own, potentially modifying various physiological functions of the host. Furthermore, experimental and clinical data suggest the existence of a communication network among the brain, gastrointestinal tract, and its microbiome, which appears to regulate immune responses, gastrointestinal function, brain function, behavior, and stress responses, also named the "gut-microbiome-brain axis." Additionally, recent research evidence has highlighted a crucial interplay between the intestinal microbiota and the lungs, referred to as the "gut-lung axis," in which alterations during critical illness could result in bacterial translocation, sustained inflammation, lung injury, and pulmonary fibrosis. In the present work, we aimed to further elucidate the pathophysiology of acute lung injury (ALI) in patients with ABI by attempting to develop the "double-hit" theory, proposing the "triple-hit" hypothesis, focused on the influence of the gut-lung axis on the lung. Particularly, we propose, in addition to sympathetic hyperactivity, blast theory, and double-hit theory, that dysbiosis and intestinal dysfunction in the context of ABI alter the gut-lung axis, resulting in the development or further aggravation of existing ALI, which constitutes the "third hit."
Collapse
Affiliation(s)
- Mairi Ziaka
- Clinic for Geriatric Medicine, Center for Geriatric Medicine and Rehabilitation, Kantonsspital Baselland, Bruderholz, Switzerland.
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
14
|
Zhang W, Jin C, Zhang S, Wu L, Li B, Shi M. Gut lymph purification alleviates acute lung injury induced by intestinal ischemia-reperfusion in rats by removing danger-associated molecular patterns from gut lymph. Heliyon 2024; 10:e25711. [PMID: 38371985 PMCID: PMC10873747 DOI: 10.1016/j.heliyon.2024.e25711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/20/2024] Open
Abstract
Background The potential effect of removing danger-associated molecular patterns (DAMPs) from gut lymph on reducing acute lung injury (ALI) induced by gut ischemia-reperfusion injury (GIRI) is uncertain. This study aimed to investigate whether gut lymph purification (GLP) could improve GIRI-induced acute lung injury in rats by clearing danger-associated molecular patterns. Materials and methods Rats were divided into four groups: Sham, GIRI, GIRI + gut lymph drainage (GLD), and GIRI + GLP. After successful modeling, lung tissue samples were collected from rats for hematoxylin-eosin (HE) staining and detection of apoptotic indexes. We detected the DAMPs levels in blood and lymph samples. We observed the microstructure of AEC Ⅱ and measured the expression levels of apoptosis indexes. Results The GIRI group showed destruction of alveolar structure, thickened alveolar walls, and inflammatory cell infiltration. This was accompanied by significantly increased levels of high mobility group protein-1 (HMGB-1) and Interleukin-6 (IL-6), while reduced levels of heat shock protein 70 (HSP 70) and Interleukin-10 (IL-10) in both lymph and serum. In contrast, the lung tissue damage in the GIRI + GLP group was significantly improved compared to the GIRI group. This was evidenced by a reduction in the expression levels of HMGB-1 and IL-6 in both lymph and serum and an increase in HSP 70 and IL-10 levels. Additionally, organelle structure of AEC II was significantly improved in the GIRI + GLP group compared to the GIRI group. Conclusions GLP inhibits inflammation and cell apoptosis in GIRI-induced ALI by blocking the link between DAMPs and mononuclear phagocytes, reducing the severity of ALI.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Critical Care Medicine, The People's Hospital of Leshan, Leshan City, Sichuan Province, 614008, China
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Can Jin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | | | - Linlin Wu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Bohan Li
- Zunyi Medical University, Zunyi, Guizhou, China
| | - Meimei Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. School of Medicine, Northwest University. Xi'an, 710069, Shanxi, China
| |
Collapse
|
15
|
Pesu H, Mbabazi J, Mutumba R, Savolainen O, Olsen MF, Mølgaard C, Michaelsen KF, Ritz C, Filteau S, Briend A, Mupere E, Friis H, Grenov B. Correlates of Plasma Citrulline, a Potential Marker of Enterocyte Mass, among Children with Stunting: A Cross-Sectional Study in Uganda. J Nutr 2024; 154:765-776. [PMID: 38135004 DOI: 10.1016/j.tjnut.2023.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Environmental enteric dysfunction (EED) is associated with stunting. Citrulline, produced in mature enterocytes, may be a valuable biomarker of small intestinal enterocyte mass in the context of EED. OBJECTIVES We aimed to explore the correlates of plasma citrulline (p-cit) in children with stunting. METHODS In a cross-sectional study using baseline data from the community-based MAGNUS (milk affecting growth, cognition and the gut in child stunting) trial (ISRCTN13093195), we explored potential correlates of p-cit in Ugandan children with stunting aged 12-59 mo. Using linear regression in univariate and multivariate models, we explored associations with socioeconomics, diet, micronutrient status, and water, sanitation, and hygiene characteristics. The influence of covariates age, fasting, and systemic inflammation were also explored. RESULTS In 750 children, the mean ± standard deviation age was 32.0 ± 11.7 mo, and height-for-age z-score was -3.02 ± 0.74. P-cit, available for 730 children, differed according to time fasted and was 20.7 ± 8.9, 22.3 ± 10.6 and 24.2 ± 13.1 μmol/L if fasted <2, 2-5 and >5 h, respectively. Positive correlates of p-cit were age [0.07; 95% confidence interval (CI): 0.001, 0.15 μmol/L] and log10 serum insulin-like growth factor-1 (8.88; 95% CI: 5.09, 12.67 μmol/L). With adjustment for systemic inflammation, the association with serum insulin-like growth factor-1 reduced (4.98; 95% CI: 0.94, 9.03 μmol/L). Negative correlates of p-cit included food insecurity, wet season (-3.12; 95% CI: -4.97, -1.26 μmol/L), serum C-reactive protein (-0.15; 95% CI: -0.20, -0.10 μmol/L), serum α1-acid glycoprotein (-5.34; 95% CI: -6.98, -3.70 μmol/L) and anemia (-1.95; 95% CI: -3.72, -0.18 μmol/L). Among the negatively correlated water, sanitation, and hygiene characteristics was lack of soap for handwashing (-2.53; 95% CI: -4.82, -0.25 μmol/L). Many associations attenuated with adjustment for inflammation. CONCLUSIONS Many of the correlates of p-cit are characteristic of populations with a high EED prevalence. Systemic inflammation is strongly associated with p-cit and is implicated in EED and stunting. Adjustment for systemic inflammation attenuates many associations, reflecting either confounding, mediation, or both. This study highlights the complex interplay between p-cit and systemic inflammation.
Collapse
Affiliation(s)
- Hannah Pesu
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Joseph Mbabazi
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Rolland Mutumba
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Otto Savolainen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; Faculty of Health Sciences, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mette F Olsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Christian Mølgaard
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Kim F Michaelsen
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Christian Ritz
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Suzanne Filteau
- Department of Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - André Briend
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark; Tampere Centre for Child Health Research, Tampere University, Tampere, Finland
| | - Ezekiel Mupere
- Department of Paediatrics and Child Health, School of Medicine College of Health Sciences, Makerere University, Kampala, Uganda
| | - Henrik Friis
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Benedikte Grenov
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
16
|
McKnight CA, Diehl LJ, Bergin IL. Digestive Tract and Salivary Glands. HASCHEK AND ROUSSEAUX' S HANDBOOK OF TOXICOLOGIC PATHOLOGY 2024:1-148. [DOI: 10.1016/b978-0-12-821046-8.00001-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
17
|
Cui G, Li S, Ye H, Yang Y, Jia X, Lin M, Chu Y, Feng Y, Wang Z, Shi Z, Zhang X. Gut microbiome and frailty: insight from genetic correlation and mendelian randomization. Gut Microbes 2023; 15:2282795. [PMID: 37990415 PMCID: PMC10730212 DOI: 10.1080/19490976.2023.2282795] [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: 03/30/2023] [Accepted: 11/08/2023] [Indexed: 11/23/2023] Open
Abstract
Observational studies have shown that the gut microbiome is associated with frailty. However, whether these associations underlie causal effects remains unknown. Thus, this study aimed to assess the genetic correlation and causal relationships between the genetically predicted gut microbiome and frailty using linkage disequilibrium score regression (LDSC) and Mendelian Randomization (MR). Summary statistics for the gut microbiome were obtained from a genome-wide association study (GWAS) meta-analysis of the MiBioGen consortium (N = 18,340). Summary statistics for frailty were obtained from a GWAS meta-analysis, including the UK Biobank and TwinGene (N = 175,226). We used LDSC and MR analyses to estimate the genetic correlation and causality between the genetically predicted gut microbiome and frailty. Our findings indicate a suggestive genetic correlation between Christensenellaceae R-7 and frailty. Moreover, we found evidence for suggestive causal effects of twelve genus-level gut microbes on frailty using at least two MR methods. There was no evidence of horizontal pleiotropy or heterogeneity in the MR analysis. This study provides suggestive evidence for a potential genetic correlation and causal association between several genetically predicted gut microbes and frailty. More population-based observational studies and animal experiments are required to clarify this association and the underlying mechanisms.
Collapse
Affiliation(s)
- Guanghui Cui
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Shaojie Li
- School of Public Health, Peking University, Beijing, China
- China Center for Health Development Studies, Peking University, Beijing, China
| | - Hui Ye
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Yao Yang
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Xiaofen Jia
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Miaomiao Lin
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Yingming Chu
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Yue Feng
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Zicheng Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zongming Shi
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| | - Xuezhi Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Peking University First Hospital; Institute of Integrated Traditional Chinese and Western Medicine, Peking University, Beijing, China
| |
Collapse
|
18
|
Arya AK, Hu K, Chen A, Olivas-Garcia Y, Coyne C, Tanaka H, Liu C, Doucet J, Chan T, Hu B. INTRACOLON COOLING INCREASES SURVIVAL RATE IN THE RAT MODEL OF LETHAL HEMORRHAGE. Shock 2023; 60:762-770. [PMID: 37878475 PMCID: PMC10840875 DOI: 10.1097/shk.0000000000002234] [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: 10/27/2023]
Abstract
ABSTRACT Background: The objective of this study was to investigate whether transrectal intracolon (TRIC) cooling can prolong the survival duration in a rat hemorrhagic shock (HS) model. Methods: A lethal HS was induced by bleeding 47% of the total blood volume. A TRIC device was placed into the colon to maintain the intracolon temperature either at 37°C (TRIC37) or at 10°C (TRIC10) post-HS. In the surface cooling (SC) rats, the body temperatures were maintained at the same level as the esophageal temperature of the TRIC10 rats. A separated group of TRIC10 rats were resuscitated (Res) at 90 min post-HS. A total of six groups were as follows: (i) Sham TRIC37 (n = 5), (ii) Sham TRIC10 (n = 5), (iii) HS TRIC37 (n = 5), (iv) HS TRIC10 (n = 6), (v) HS SC (n = 6), and (vi) HS TRIC10 + Res (n = 6). Results: An average post-HS survival time was 18.4 ± 9.4 min in HS TRIC37 and 82 ± 27.82 min in the HS SC group. In striking contrast, the HS TRIC10 group exhibited an average survival time of 150.2 ± 66.43 min. The post-HS blood potassium level rose significantly in the HS TRIC37 and HS SC, whereas it remained unchanged in the TRIC10 groups. Post-HS intestinal damage occurred in HS TRIC37 and HS SC groups but virtually absent in HS TRIC10 groups. After resuscitation at 90 min post-HS, all HS TRIC10 rats were fully recovered from the lethal HS. Conclusions: TRIC10 reversed the high blood potassium level, prevented the intestinal damage, and prolonged the survival duration by sixfold relative to normothermia and by twofold compared with SC post-HS. All TRIC10 rats were successfully resuscitated at 90 min post-HS.
Collapse
Affiliation(s)
- Awadhesh K Arya
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kurt Hu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alice Chen
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Yamileck Olivas-Garcia
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Christopher Coyne
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Hideaki Tanaka
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Chunli Liu
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Jay Doucet
- Department of Surgery, Division of Trauma, University of California San Diego, San Diego, CA, USA
| | - Theodore Chan
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
| | - Bingren Hu
- Departments of Emergency Medicine, University of California San Diego, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Wei YH, Bi RT, Qiu YM, Zhang CL, Li JZ, Li YN, Hu B. The gastrointestinal-brain-microbiota axis: a promising therapeutic target for ischemic stroke. Front Immunol 2023; 14:1141387. [PMID: 37342335 PMCID: PMC10277866 DOI: 10.3389/fimmu.2023.1141387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/22/2023] [Indexed: 06/22/2023] Open
Abstract
Ischemic stroke is a highly complex systemic disease characterized by intricate interactions between the brain and gastrointestinal tract. While our current understanding of these interactions primarily stems from experimental models, their relevance to human stroke outcomes is of considerable interest. After stroke, bidirectional communication between the brain and gastrointestinal tract initiates changes in the gastrointestinal microenvironment. These changes involve the activation of gastrointestinal immunity, disruption of the gastrointestinal barrier, and alterations in gastrointestinal microbiota. Importantly, experimental evidence suggests that these alterations facilitate the migration of gastrointestinal immune cells and cytokines across the damaged blood-brain barrier, ultimately infiltrating the ischemic brain. Although the characterization of these phenomena in humans is still limited, recognizing the significance of the brain-gastrointestinal crosstalk after stroke offers potential avenues for therapeutic intervention. By targeting the mutually reinforcing processes between the brain and gastrointestinal tract, it may be possible to improve the prognosis of ischemic stroke. Further investigation is warranted to elucidate the clinical relevance and translational potential of these findings.
Collapse
Affiliation(s)
| | | | | | | | | | - Ya-nan Li
- *Correspondence: Ya-nan Li, ; Bo Hu,
| | - Bo Hu
- *Correspondence: Ya-nan Li, ; Bo Hu,
| |
Collapse
|
20
|
Conti Bellocchi MC, Crinò SF, De Marchi G, De Pretis N, Ofosu A, Caldart F, Ciccocioppo R, Frulloni L. A Clinical and Pathophysiological Overview of Intestinal and Systemic Diseases Associated with Pancreatic Disorders: Causality or Casualty? Biomedicines 2023; 11:biomedicines11051393. [PMID: 37239064 DOI: 10.3390/biomedicines11051393] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
The relationship between chronic intestinal disease, including inflammatory bowel disease (IBD) and celiac disease (CelD), and pancreatic disorders has been little investigated. Although an increased risk of acute pancreatitis (AP), exocrine pancreatic insufficiency with or without chronic pancreatitis, and chronic asymptomatic pancreatic hyperenzymemia have been described in these patients, the pathogenetic link remains unclear. It may potentially involve drugs, altered microcirculation, gut permeability/motility with disruption of enteric-mediated hormone secretion, bacterial translocation, and activation of the gut-associated lymphoid tissue related to chronic inflammation. In addition, the risk of pancreatic cancer seems to be increased in both IBD and CelD patients with unknown pathogenesis. Finally, other systemic conditions (e.g., IgG4-related disease, sarcoidosis, vasculitides) might affect pancreatic gland and the intestinal tract with various clinical manifestations. This review includes the current understandings of this enigmatic association, reporting a clinical and pathophysiological overview about this topic.
Collapse
Affiliation(s)
| | - Stefano Francesco Crinò
- Diagnostic and Interventional Endoscopy of Pancreas, Pancreas Institute, University of Verona, 37134 Verona, Italy
| | - Giulia De Marchi
- Gastroenterology Unit, Department of Medicine, Pancreas Institute, University of Verona, 37134 Verona, Italy
| | - Nicolò De Pretis
- Gastroenterology Unit, Department of Medicine, Pancreas Institute, University of Verona, 37134 Verona, Italy
| | - Andrew Ofosu
- Division of Gastroenterology and Hepatology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Federico Caldart
- Gastroenterology Unit, Department of Medicine, Pancreas Institute, University of Verona, 37134 Verona, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Pancreas Institute, University of Verona, 37134 Verona, Italy
| | - Luca Frulloni
- Gastroenterology Unit, Department of Medicine, Pancreas Institute, University of Verona, 37134 Verona, Italy
| |
Collapse
|
21
|
Nguyen M, Gautier T, Masson D, Bouhemad B, Guinot PG. Endotoxemia in Acute Heart Failure and Cardiogenic Shock: Evidence, Mechanisms and Therapeutic Options. J Clin Med 2023; 12:jcm12072579. [PMID: 37048662 PMCID: PMC10094881 DOI: 10.3390/jcm12072579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/22/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
Acute heart failure and cardiogenic shock are frequently occurring and deadly conditions. In patients with those conditions, endotoxemia related to gut injury and gut barrier dysfunction is usually described as a driver of organ dysfunction. Because endotoxemia might reciprocally alter cardiac function, this phenomenon has been suggested as a potent vicious cycle that worsens organ perfusion and leading to adverse outcomes. Yet, evidence beyond this phenomenon might be overlooked, and mechanisms are not fully understood. Subsequently, even though therapeutics available to reduce endotoxin load, there are no indications to treat endotoxemia during acute heart failure and cardiogenic shock. In this review, we first explore the evidence regarding endotoxemia in acute heart failure and cardiogenic shock. Then, we describe the main treatments for endotoxemia in the acute setting, and we present the challenges that remain before personalized treatments against endotoxemia can be used in patients with acute heart failure and cardiogenic shock.
Collapse
|
22
|
Prado Y, Aravena C, Aravena D, Eltit F, Gatica S, Riedel CA, Simon F. Small Plastics, Big Inflammatory Problems. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1408:101-127. [PMID: 37093424 DOI: 10.1007/978-3-031-26163-3_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The immune system is the first defense against potentially dangerous chemicals, infections, and damaged cells. Interactions between immune cells and inflammatory mediators increase the coordinated activation of cross-talking signaling pathways, resulting in an acute response necessary to restore homeostasis but potentially detrimental if uncontrolled and prolonged. Plastic production exceeds million tons per year, becoming a global concern due to the stability of its constituent polymers, low density, which allows them to spread easily, and small size, which prevents proper removal by wastewater treatment plants, promoting environmental accumulation and increasing health threats. The interaction between plastic particles and the immune system is still being investigated, owing to growing evidence of increased risk not only for dietary intake due to its presence in food packaging, drinking water, and even fruits and vegetables, but also to emerging evidence of new intake pathways such as respiratory and cutaneous. We discuss in depth the impact of small plastic particles on the immune response across the body, with a focus on the nervous system and peripheral organs and tissues such as the gastrointestinal, respiratory, lymphatic, cardiovascular, and reproductive systems, as well as the involvement in increased susceptibility to worsening concomitant diseases and future perspectives in the exploration of potential therapeutics.
Collapse
Affiliation(s)
- Yolanda Prado
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Cristobal Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Diego Aravena
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Eltit
- Department of Urologic Sciences, University of British Columbia, Vancouver, Canada
- Vancouver Prostate Centre, Vancouver, Canada
| | - Sebastian Gatica
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Claudia A Riedel
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Felipe Simon
- Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.
- Millennium Nucleus of Ion Channel-Associated Diseases, Santiago, Chile.
| |
Collapse
|
23
|
Chen Z, Yang X, Guo J, Jin T, Lin Z, Zhu P, Li J, Li L, Sun X, Du D, Jiang K, He Y, Cai F, Li L, Hu C, Tan Q, Huang W, Deng L, Xia Q. AGI grade-guided chaiqin chengqi decoction treatment for predicted moderately severe and severe acute pancreatitis (CAP trial): study protocol of a randomised, double-blind, placebo-controlled, parallel-group, pragmatic clinical trial. Trials 2022; 23:933. [PMID: 36348365 PMCID: PMC9644559 DOI: 10.1186/s13063-022-06792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/24/2022] [Indexed: 11/09/2022] Open
Abstract
Background Acute pancreatitis (AP) is a common digestive disease with increased incidence globally but without internationally licenced pharmacological therapy. Moderately severe and severe acute pancreatitis (MSAP/SAP) contributes predominately for its morbidities and mortality and has been managed in West China Hospital for decades using the traditional Chinese medicinal formula chaiqin chengqi decoction (CQCQD). The current study tests whether the early administration of CQCQD will result in improved clinical outcomes in predicted MSAP/SAP patients. Methods This is a single-centre, randomised, controlled, double-blind pragmatic clinical trial. AP patients aged 18–75 admitted within 72 h of onset will be assessed at admission for enrolment. We excluded the predicted mild acute pancreatitis (Harmless Acute Pancreatitis Score > 2 at admission) and severe organ failure (Sequential Organ Failure Assessment [SOFA] score of respiratory, cardiovascular, or renal systems > 3) at admission. Eligible patients will be randomly allocated on a 1:1 basis to CQCQD or placebo control administration based on conventional therapy. The administration of CQCQD and placebo is guided by the Acute Gastrointestinal Injury grade-based algorithm. The primary outcome measure will be the duration of respiratory failure (SOFA score of respiratory system ≥ 2) within 28 days after onset. Secondary outcome measures include occurrence of new-onset any organ failure (SOFA score of respiratory, cardiovascular, or renal system ≥ 2) and new-onset persistent organ failure (organ failure lasts > 48 h), dynamic surrogate biochemical markers and clinical severity scores, gut-centred treatment modalities, local complications status, intensive care need and duration, surgical interventions, mortality, and length of hospital stay. Follow-up will be scheduled on 6, 12, and 26 weeks after enrolment to assess AP recurrence, local complications, the requirement for surgical interventions, all-cause mortality, and patient-reported outcomes. Discussion The results of this study will provide high-quality evidence to appraise the efficacy of CQCQD for the early management of AP patients. Trial registration Chictr.org.cn Registry (ChiCTR2000034325). Registered on 2 July, 2020. Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06792-x.
Collapse
|
24
|
The Combined Escherichia coli Nissle 1917 and Tryptophan Treatment Modulates Immune and Metabolome Responses to Human Rotavirus Infection in a Human Infant Fecal Microbiota-Transplanted Malnourished Gnotobiotic Pig Model. mSphere 2022; 7:e0027022. [PMID: 36073800 PMCID: PMC9599269 DOI: 10.1128/msphere.00270-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human rotavirus (HRV) is a major cause of childhood diarrhea in developing countries where widespread malnutrition contributes to the decreased oral vaccine efficacy and increased prevalence of other enteric infections, which are major concerns for global health. Neonatal gnotobiotic (Gn) piglets closely resemble human infants in their anatomy, physiology, and outbred status, providing a unique model to investigate malnutrition, supplementations, and HRV infection. To understand the molecular signatures associated with immune enhancement and reduced diarrheal severity by Escherichia coli Nissle 1917 (EcN) and tryptophan (TRP), immunological responses and global nontargeted metabolomics and lipidomics approaches were investigated on the plasma and fecal contents of malnourished pigs transplanted with human infant fecal microbiota and infected with virulent (Vir) HRV. Overall, EcN + TRP combined (rather than individual supplement action) promoted greater and balanced immunoregulatory/immunostimulatory responses associated with greater protection against HRV infection and disease in malnourished humanized piglets. Moreover, EcN + TRP treatment upregulated the production of several metabolites with immunoregulatory/immunostimulatory properties: amino acids (N-acetylserotonin, methylacetoacetyl-CoA), lipids (gamma-butyrobetaine, eicosanoids, cholesterol-sulfate, sphinganine/phytosphingosine, leukotriene), organic compound (biliverdin), benzenoids (gentisic acid, aminobenzoic acid), and nucleotides (hypoxathine/inosine/xanthine, cytidine-5'-monophosphate). Additionally, the levels of several proinflammatory metabolites of organic compounds (adenosylhomocysteine, phenylacetylglycine, urobilinogen/coproporphyrinogen) and amino acid (phenylalanine) were reduced following EcN + TRP treatment. These results suggest that the EcN + TRP effects on reducing HRV diarrhea in neonatal Gn pigs were at least in part due to altered metabolites, those involved in lipid, amino acid, benzenoids, organic compounds, and nucleotide metabolism. Identification of these important mechanisms of EcN/TRP prevention of HRV diarrhea provides novel targets for therapeutics development. IMPORTANCE Human rotavirus (HRV) is the most common cause of viral gastroenteritis in children, especially in developing countries, where the efficacy of oral HRV vaccines is reduced. Escherichia coli Nissle 1917 (EcN) is used to treat enteric infections and ulcerative colitis while tryptophan (TRP) is a biomarker of malnutrition, and its supplementation can alleviate intestinal inflammation and normalize intestinal microbiota in malnourished hosts. Supplementation of EcN + TRP to malnourished humanized gnotobiotic piglets enhanced immune responses and resulted in greater protection against HRV infection and diarrhea. Moreover, EcN + TRP supplementation increased the levels of immunoregulatory/immunostimulatory metabolites while decreasing the production of proinflammatory metabolites in plasma and fecal samples. Profiling of immunoregulatory and proinflammatory biomarkers associated with HRV perturbations will aid in the identification of treatments against HRV and other enteric diseases in malnourished children.
Collapse
|
25
|
Sandys O, Te Velde A. Raising the Alarm: Environmental Factors in the Onset and Maintenance of Chronic (Low-Grade) Inflammation in the Gastrointestinal Tract. Dig Dis Sci 2022; 67:4355-4368. [PMID: 34981314 DOI: 10.1007/s10620-021-07327-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022]
Abstract
Chronic inflammatory disease of the gastrointestinal (GI) tract is defined by several pathophysiological characteristics, such as dysbiosis of the microbiota, epithelial barrier hyperpermeability, systemic dissemination of endotoxins and chronic inflammation. In addition to well-reported environmental factors in non-communicable disease, such as smoking, diet, and exercise, humans are frequently exposed to myriads more environmental factors, from pesticides to food additives. Such factors are ubiquitous across both our diet and indoor/outdoor environments. A major route of human exposure to these factors is ingestion, which frequently occurs due to their intentional addition (intentional food additives) and/or unintentional contamination (unintentional food contaminants) of food products-often linked to environmental pollution. Understanding how this persistent, diverse exposure impacts GI health is of paramount importance, as deterioration of the GI barrier is proposed to be the first step towards systemic inflammation and chronic disease. Therefore, we aim to evaluate the impact of ingestion of environmental factors on inflammatory processes in the GI tract. In this review, we highlight human exposure to intentional food additives (e.g. emulsifiers, bulking agents) and unintentional food contaminants (e.g. persistent organic pollutants, pesticides, microplastics), then present evidence for their association with chronic disease, modification of the GI microbiota, increased permeability of the GI barrier, systemic dissemination of endotoxins, local (and distal) pro-inflammatory signalling, and induction of oxidative stress and/or endoplasmic reticulum stress. We also propose a link to NLRP3-inflammasome activation. These findings highlight the contribution of common environmental factors towards deterioration of GI health and the induction of pathophysiology associated with onset and maintenance of chronic inflammation in the GI tract.
Collapse
Affiliation(s)
- Oliver Sandys
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam, The Netherlands
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anje Te Velde
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, AGEM, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Ellis PS, Martins RR, Thompson EJ, Farhat A, Renshaw SA, Henriques CM. A subset of gut leukocytes has telomerase-dependent "hyper-long" telomeres and require telomerase for function in zebrafish. Immun Ageing 2022; 19:31. [PMID: 35820929 PMCID: PMC9277892 DOI: 10.1186/s12979-022-00287-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Telomerase, the enzyme capable of elongating telomeres, is usually restricted in human somatic cells, which contributes to progressive telomere shortening with cell-division and ageing. T and B-cells cells are somatic cells that can break this rule and can modulate telomerase expression in a homeostatic manner. Whereas it seems intuitive that an immune cell type that depends on regular proliferation outbursts for function may have evolved to modulate telomerase expression it is less obvious why others may also do so, as has been suggested for macrophages and neutrophils in some chronic inflammation disease settings. The gut has been highlighted as a key modulator of systemic ageing and is a key tissue where inflammation must be carefully controlled to prevent dysfunction. How telomerase may play a role in innate immune subtypes in the context of natural ageing in the gut, however, remains to be determined. RESULTS Using the zebrafish model, we show that subsets of gut immune cells have telomerase-dependent"hyper-long" telomeres, which we identified as being predominantly macrophages and dendritics (mpeg1.1+ and cd45+mhcII+). Notably, mpeg1.1+ macrophages have much longer telomeres in the gut than in their haematopoietic tissue of origin, suggesting that there is modulation of telomerase in these cells, in the gut. Moreover, we show that a subset of gut mpeg1.1+ cells express telomerase (tert) in young WT zebrafish, but that the relative proportion of these cells decreases with ageing. Importantly, this is accompanied by telomere shortening and DNA damage responses with ageing and a telomerase-dependent decrease in expression of autophagy and immune activation markers. Finally, these telomerase-dependent molecular alterations are accompanied by impaired phagocytosis of E. coli and increased gut permeability in vivo. CONCLUSIONS Our data show that limiting levels of telomerase lead to alterations in gut immunity, impacting on the ability to clear pathogens in vivo. These are accompanied by increased gut permeability, which, together, are likely contributors to local and systemic tissue degeneration and increased susceptibility to infection with ageing.
Collapse
Affiliation(s)
- Pam S Ellis
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Raquel R Martins
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Emily J Thompson
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Asma Farhat
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK
| | - Stephen A Renshaw
- The Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, UK
| | - Catarina M Henriques
- The Bateson Centre, MRC-Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing and Department of Oncology and Metabolism, Healthy Lifespan Institute, University of Sheffield Medical School, Sheffield, UK.
| |
Collapse
|
27
|
Tryptophan-rich diet ameliorates chronic unpredictable mild stress induced depression- and anxiety-like behavior in mice: The potential involvement of gut-brain axis. Food Res Int 2022; 157:111289. [DOI: 10.1016/j.foodres.2022.111289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/27/2022]
|
28
|
Jiao J, Liu J, Li Q, Zhang G, Pan C, Luo F, Zhang Q, Qi B, Zhao L, Yin P, Shang D. Gut Microbiota-Derived Diaminopimelic Acid Promotes the NOD1/RIP2 Signaling Pathway and Plays a Key Role in the Progression of Severe Acute Pancreatitis. Front Cell Infect Microbiol 2022; 12:838340. [PMID: 35811665 PMCID: PMC9257083 DOI: 10.3389/fcimb.2022.838340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/02/2022] [Indexed: 11/18/2022] Open
Abstract
Impaired intestinal barrier function and gut microbiota dysbiosis are believed to be related to exacerbation of acute pancreatitis (AP). As a bacterial cell wall peptidoglycan component, diaminopimelic acid (DAP) is a specific ligand of NOD1 that regulates the NOD1/RIP2/NF-kB signaling pathway. Here, we investigated the role of DAP in the crosstalk between the gut microbiota and pancreas during the occurrence of AP. Upregulation of NOD1/RIP2/NF-kB and elevated serum DAP levels were found in severe AP (SAP) model rats. The accumulation of DAP in SAP patients corroborated its ability to serve as an indicator of disease severity. Subsequently, SAP rats were treated with oral administration of the traditional Chinese medicine Qingyi Keli (QYKL) as well as neomycin, which can widely eliminate DAP-containing bacteria. Both QYKL and neomycin intervention ameliorated intestinal and pancreatic damage and systemic inflammation in SAP rats. Through 16S rDNA sequencing, we found that QYKL could rehabilitate the gut microbiota structure and selectively inhibit the overgrowth of enteric bacteria, such as Helicobacter and Lactobacillus, in SAP rats without affecting some protective strains, including Romboutsia and Allobaculum. Interestingly, we demonstrated that the decrease in serum DAP was accompanied by suppression of the NOD1/RIP2/NF-kB signaling pathway in both the intestine and pancreas of the two intervention groups. Taken together, these results suggested that the gut microbiota-DAP-NOD1/RIP2 signaling pathway might play a critical role in the progression of AP and that SAP could be alleviated via intervention in the signaling pathway. Our work provides new potential early warning indicators of SAP and targets for intervention.
Collapse
Affiliation(s)
- Juying Jiao
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianjun Liu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Qi Li
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Guixin Zhang
- Pancreaticobiliary Centre, Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chen Pan
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Fei Luo
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Qingkai Zhang
- Pancreaticobiliary Centre, Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bing Qi
- Pancreaticobiliary Centre, Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Liang Zhao
- Pancreaticobiliary Centre, Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Peiyuan Yin
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
- *Correspondence: Dong Shang, ; Peiyuan Yin,
| | - Dong Shang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- College of Integrative Medicine, Dalian Medical University, Dalian, China
- Pancreaticobiliary Centre, Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- *Correspondence: Dong Shang, ; Peiyuan Yin,
| |
Collapse
|
29
|
Bao Z, Zhang Z, Zhou G, Zhang A, Shao A, Zhou F. Novel Mechanisms and Therapeutic Targets for Ischemic Stroke: A Focus on Gut Microbiota. Front Cell Neurosci 2022; 16:871720. [PMID: 35656406 PMCID: PMC9152006 DOI: 10.3389/fncel.2022.871720] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is the most common type of stroke with limited treatment options. Although the pathological mechanisms and potential therapeutic targets of ischemic stroke have been comprehensively studied, no effective therapies were translated into clinical practice. Gut microbiota is a complex and diverse dynamic metabolic ecological balance network in the body, including a large number of bacteria, archaea, and eukaryotes. The composition, quantity and distribution in gut microbiota are found to be associated with the pathogenesis of many diseases, such as individual immune abnormalities, metabolic disorders, and neurodegeneration. New insight suggests that ischemic stroke may lead to changes in the gut microbiota and the alterations of gut microbiota may determine stroke outcomes in turn. The link between gut microbiota and stroke is expected to provide new perspectives for ischemic stroke treatment. In this review, we discuss the gut microbiota alterations during ischemic stroke and gut microbiota-related stroke pathophysiology and complications. Finally, we highlight the role of the gut microbiota as a potential therapeutic target for ischemic stroke and summarize the microbiome-based treatment options that can improve the recovery of stroke patients.
Collapse
|
30
|
Kornbausch N, Debong MW, Buettner A, Heydel JM, Loos H. Odorant Metabolism in Humans. Angew Chem Int Ed Engl 2022; 61:e202202866. [PMID: 35522818 PMCID: PMC9541901 DOI: 10.1002/anie.202202866] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Indexed: 11/08/2022]
Abstract
Odorants are relatively small molecules which are easily taken up and distributed in the human body. Despite their relevance in everyday life, however, only a limited amount of evidence about their metabolism, pathways, and bioactivities in the human body exists. With this Review, we aim to encourage future interdisciplinary research on the function and mechanisms of the biotransformation of odorants, involving different disciplines such as nutrition, medicine, biochemistry, chemistry, and sensory sciences. Starting with a general overview of the different ways of odorant uptake and enzymes involved in the metabolism of odorants, a more precise description of biotransformation processes and their function in the oral cavity, the nose, the lower respiratory tract (LRT), and the gastrointestinal tract (GIT) is given together with an overview of the different routes of odorant excretion. Finally, perspectives for future research are discussed.
Collapse
Affiliation(s)
- Nicole Kornbausch
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Chemistry and Pharmacy, GERMANY
| | - Marcel W Debong
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Chemistry and Pharmacy, GERMANY
| | - Andrea Buettner
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Chemistry and Pharmacy, GERMANY
| | - Jean-Marie Heydel
- Centre des Sciences du Goût et de l'Alimentation: Centre des Sciences du Gout et de l'Alimentation, Flavour perception: from molecule to behavior, FRANCE
| | - Helene Loos
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Chemistry and Pharmacy, Henkestr. 9, 91054, Erlangen, GERMANY
| |
Collapse
|
31
|
Kornbausch N, Debong MW, Buettner A, Heydel JM, Loos H. Odorant Metabolism in Humans. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Nicole Kornbausch
- Friedrich-Alexander-Universität Erlangen-Nürnberg Chemistry and Pharmacy GERMANY
| | - Marcel W. Debong
- Friedrich-Alexander-Universität Erlangen-Nürnberg Chemistry and Pharmacy GERMANY
| | - Andrea Buettner
- Friedrich-Alexander-Universität Erlangen-Nürnberg Chemistry and Pharmacy GERMANY
| | - Jean-Marie Heydel
- Centre des Sciences du Goût et de l'Alimentation: Centre des Sciences du Gout et de l'Alimentation Flavour perception: from molecule to behavior FRANCE
| | - Helene Loos
- Friedrich-Alexander-Universität Erlangen-Nürnberg Chemistry and Pharmacy Henkestr. 9 91054 Erlangen GERMANY
| |
Collapse
|
32
|
Zheng J, Zhang W, Kang P, Zheng X, He K, Bai H, Yu X. Midazolam Ameliorates Impairment of the Blood-Brain Barrier (BBB) Against LPS. Neurotox Res 2022; 40:751-762. [PMID: 35451708 DOI: 10.1007/s12640-022-00508-4] [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: 02/03/2022] [Revised: 03/27/2022] [Accepted: 04/09/2022] [Indexed: 10/18/2022]
Abstract
Central nervous system (CNS) dysfunction induced by sepsis and pathogenic microbial infections is reported to be closely associated with increased permeability of the blood-brain barrier (BBB), which is mainly mediated by the stimulation of lipopolysaccharide (LPS) on inflammatory signaling. Midazolam is a novel sedative acting on the benzodiazepine receptor, which is recently reported to exert a neuroprotective effect by inhibiting inflammation. The present study will explore the potential repair capacity of Midazolam on LPS-induced damage to the BBB. The in vivo mice model was established by intraperitoneal injection of LPS, while the in vitro model was constructed by stimulating endothelial cells utilizing LPS. We found that the increased malondialdehyde (MDA) level and reduced superoxide dismutase (SOD) activity in the brain cortices, promoted serum concentration of inflammatory factors, and elevated BBB permeability were found in the LPS group, all of which were dramatically reversed by 1 mg/kg and 2 mg/kg Midazolam. Interestingly, Midazolam increased the expression of the tight junction protein zonula occludens-1 (ZO-1). In LPS-challenged in vitro human brain microvascular endothelial cells (HBMECs), the increased concentration of inflammatory factors, reduced trans-endothelial electrical resistance (TEER) level, elevated relative value of trans-endothelial permeability, and downregulated ZO-1 were observed, all of which were pronouncedly alleviated by Midazolam, accompanied by the inhibition on the Ras homolog family member A/ Rho-kinase 2 (RhoA/ROCK-2) pathway. Furthermore, the regulatory effects of Midazolam on ZO-1 expression and the endothelial monolayer permeability in LPS-challenged HBMECs were abolished by the overexpression of RhoA. Collectively, our data imply that Midazolam ameliorated the impairment of the BBB against LPS by regulating the RhoA/ROCK2 pathway.
Collapse
Affiliation(s)
- Juyan Zheng
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Wei Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Bejing, 100730, China
| | - PeiPei Kang
- Department of Anesthesiology, Affiliated Tumor Hospital of Nantong University, Nantong, 226006, China
| | - Xiaojiao Zheng
- Department of Blood Transfusion, The First Affiliated Hospital of Baotou Medical College, Baotou, 014017, China
| | - Kai He
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China
| | - Hong Bai
- Department of Anesthesiology, Wuhai People's Hospital, Wuhai, 016099, China
| | - Xuerong Yu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
33
|
Mesenteric lymph system constitutes the second route in gut-liver axis and transports metabolism-modulating gut microbial metabolites. J Genet Genomics 2022; 49:612-623. [PMID: 35417777 DOI: 10.1016/j.jgg.2022.03.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 12/17/2022]
|
34
|
Bacterial Translocation in Gastrointestinal Cancers and Cancer Treatment. Biomedicines 2022; 10:biomedicines10020380. [PMID: 35203589 PMCID: PMC8962358 DOI: 10.3390/biomedicines10020380] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022] Open
Abstract
In recent years, there has been increasing evidence that gut microbiota is associated with the onset and exacerbation of various diseases, such as gastrointestinal cancer. For instance, it is well known that local inflammation of the intestinal tract in colorectal cancer that is caused by the increased number of Fusobacterium, due to changes in the intestinal bacterial flora, is involved in carcinogenesis. In contrast, gut bacteria or their products, pathogen-associated molecular patterns, not only cause intestinal inflammation but also invade the bloodstream through dysbiosis and gut barrier dysfunction, thereby leading to systemic inflammation, namely bacterial translocation. The involvement of bacterial translocation in the carcinogenesis of gastrointestinal cancers and their prognosis is increasingly being recognized. The Toll-like receptor signaling pathways plays an important role in the carcinogenesis of such cancers. In addition, bacterial translocation influences the treatment of cancers such as surgery and chemotherapy. In this review, we outline the concept of bacterial translocation, summarize the current knowledge on the relationship between gut bacteria and gastrointestinal cancer, and provide future perspectives of this field.
Collapse
|
35
|
Saviano A, Gayani G, Migneco A, Candelli M, Franceschi F, Ojetti V, Zanza C, Longhitano Y. The Gut Microbiota-Brain Axis in Acute Neurological Disease: Focus on Stroke. Rev Recent Clin Trials 2022; 17:240-244. [PMID: 35319389 DOI: 10.2174/1574887117666220321155508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/27/2021] [Accepted: 01/11/2022] [Indexed: 01/15/2023]
Abstract
The gut microbiota is one of the great innovations of modern medicine. In the modern microbiota revolution era, more comprehensive and in-depth studies have been performed regarding the microbial gut communities and their impact on acute and chronic diseases, including those of the nervous system as acute neurological diseases. The microbiota has changed our knowledge of medical conditions; in particular, considering stroke (both ischemic and hemorrhagic), literature studies, experimental and clinical researches indicate that the individual's risk and outcomes are substantially impacted by the gut microbiota. The aim of our review article is to investigate and discuss the recent insights into the emerging role of this complex "gut microbiota-brain axis" in affecting some acute neurological diseases, such as stroke, responsible for a significant number of deaths worldwide. We performed electronic research on PubMed® and collected articles published in the last ten years, finding that changes in the gut microbiota composition could affect various aspects of stroke pathophysiology and individual predisposition, risk, and outcomes. Our review article suggests that there is a strong connection between the gut microbiota and the brain, both in health and in acute neurological diseases such as stroke. Investigating and exploring this relationship can be a challenge useful to learn more about this disabling/deadly condition, and it can be a useful tool to identify novel potential therapeutic approaches, improving an individual's outcomes and life.
Collapse
Affiliation(s)
- Angela Saviano
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, IRCCS Rome, Italy
| | - Gunawardena Gayani
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, IRCCS Rome, Italy
| | - Alessio Migneco
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, IRCCS Rome, Italy
| | - Marcello Candelli
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, IRCCS Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, IRCCS Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Veronica Ojetti
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, IRCCS Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Christian Zanza
- Department of Emergency Medicine, Anesthesia and Critical Care Medicine, Michele and Pietro Ferrero Hospital, Foundation Ospedale Alba-Bra Onlus, Verduno (Cuneo), Italy
- Research Training Innovation Infrastructure, Research and Innovation Department, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| | - Yaroslava Longhitano
- Research Training Innovation Infrastructure, Research and Innovation Department, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, 15121 Alessandria, Italy
| |
Collapse
|
36
|
Factors associated with all-cause mortality at 90 days in hospitalized adult patients who received parenteral nutrition. NUTR HOSP 2022; 39:728-737. [DOI: 10.20960/nh.04106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
37
|
Recent Advances in the Impact of Infection and Inflammation on Stroke Risk and Outcomes. Curr Neurol Neurosci Rep 2022; 22:161-170. [PMID: 35235168 PMCID: PMC8889053 DOI: 10.1007/s11910-022-01179-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2021] [Indexed: 11/17/2022]
Abstract
PURPOSE OF THE REVIEW Inflammation is a key component in the pathogenesis of cerebrovascular diseases. In the past few years, the role of systemic infection and gut dysbiosis in modulating inflammation and stroke risk has been increasingly acknowledged. In this review, we synthesize contemporary literature on the effects of infection and inflammation on stroke risk and outcomes, with a focus on periodontal disease, COVID-19 infection, and gut dysbiosis. RECENT FINDINGS Chronic and acute infections such as periodontitis and COVID-19 induce systemic inflammation that cause atherogenesis and increase cardiac injury and arrhythmias. These infections also directly injure the endothelium leading to worsened secondary inflammation after stroke. Gut dysbiosis engenders a pro-inflammatory state by modulating intestinal lymphocyte populations that can traffic directly to the brain. Additionally, post-stroke immune dysregulation creates a compounding feedback loop of further infections and gut dysbiosis that worsen outcomes. Recent advances in understanding the pathophysiology of how infection and dysbiosis affect the progression of stroke, as well as long-term recovery, have revealed tantalizing glimpses at potential therapeutic targets. We discuss the multidirectional relationship between stroke, infection, and gut dysbiosis, and identify areas for future research to further explore therapeutic opportunities.
Collapse
|
38
|
Ke X, Hu T, Jiang M. cGAS-STING signaling pathway in gastrointestinal inflammatory disease and cancers. FASEB J 2021; 36:e22029. [PMID: 34907606 DOI: 10.1096/fj.202101199r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway has emerged as a key DNA-sensing machinery in innate immunity. Activation of cGAS-STING signaling pathway mediates the production of interferons and proinflammatory cytokines. Although cGAS-STING signaling pathway shows critical function in the maintenance of gut homeostasis, overactive cGAS-STING signaling pathway leads to gastrointestinal (GI) inflammation. Harnessing the effect and mechanism of the cGAS-STING signaling pathway could be beneficial for the development of novel strategies for the treatment of GI diseases. This review presents recent advances regarding the role of cGAS-STING signaling pathway in GI inflammatory disease and cancers and describes perspective therapeutic strategies targeting the signaling pathway.
Collapse
Affiliation(s)
- Xinxin Ke
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Tao Hu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Mizu Jiang
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
39
|
Li Y, Tao Y, Xu J, He Y, Zhang W, Jiang Z, He Y, Liu H, Chen M, Zhang W, Xing Z. Hyperoxia Provokes Time- and Dose-Dependent Gut Injury and Endotoxemia and Alters Gut Microbiome and Transcriptome in Mice. Front Med (Lausanne) 2021; 8:732039. [PMID: 34869425 PMCID: PMC8635731 DOI: 10.3389/fmed.2021.732039] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Oxygen therapy usually exposes patients to hyperoxia, which induces injuries in the lung, the heart, and the brain. The gut and its microbiome play key roles in critical illnesses, but the impact of hyperoxia on the gut and its microbiome remains not very clear. We clarified the time- and dose-dependent effects of hyperoxia on the gut and investigated oxygen-induced gut dysbiosis and explored the underlying mechanism of gut injury by transcriptome analysis. Methods: The C57BL/6 mice were randomly divided into the control group and nine different oxygen groups exposed to hyperoxia with an inspired O2 fraction (FiO2) of 40, 60, and 80% for 24, 72, and 168 h (7 days), respectively. Intestinal histopathological and biochemical analyses were performed to explore the oxygen-induced gut injury and inflammatory response. Another experiment was performed to explore the impact of hyperoxia on the gut microbiome by exposing the mice to hyperoxia (FiO2 80%) for 7 days, with the 16S rRNA sequencing method. We prolonged the exposure (up to 14 days) of the mice to hyperoxia (FiO2 80%), and gut transcriptome analysis and western blotting were carried out to obtain differentially expressed genes (DEGs) and signaling pathways related to innate immunity and cell death. Results: Inhaled oxygen induced time- and dose-dependent gut histopathological impairment characterized by mucosal atrophy (e.g., villus shortening: 80% of FiO2 for 24 h: P = 0.008) and enterocyte death (e.g., apoptosis: 40% of FiO2 for 7 days: P = 0.01). Administered time- and dose-dependent oxygen led to intestinal barrier dysfunction (e.g., endotoxemia: 80% of FiO2 for 72 h: P = 0.002) and potentiated gut inflammation by increasing proinflammatory cytokines [e.g., tumor necrosis factor alpha (TNF-α): 40% of FiO2 for 24 h: P = 0.003)] and reducing anti-inflammatory cytokines [Interleukin 10 (IL-10): 80% of FiO2 for 72 h: P < 0.0001]. Hyperoxia induced gut dysbiosis with an expansion of oxygen-tolerant bacteria (e.g., Enterobacteriaceae). Gut transcriptome analysis identified 1,747 DEGs and 171 signaling pathways and immunoblotting verified TLR-4, NOD-like receptor, and apoptosis signaling pathways were activated in oxygen-induced gut injury. Conclusions: Acute hyperoxia rapidly provokes gut injury in a time- and dose-dependent manner and induces gut dysbiosis, and an innate immune response is involved in an oxygen-induced gut injury.
Collapse
Affiliation(s)
- Yunhang Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuanfa Tao
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yihuai He
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wen Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhigang Jiang
- Department of Statistics, Zunyi Medical University, Zunyi, China
| | - Ying He
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Houmei Liu
- Department of Endodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wei Zhang
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhouxiong Xing
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
40
|
Liu C, Yuan D, Crawford R, Sarkar R, Hu B. Directly Cooling Gut Prevents Mortality in the Rat Model of Reboa Management of Lethal Hemorrhage. Shock 2021; 56:813-823. [PMID: 33555843 PMCID: PMC8329109 DOI: 10.1097/shk.0000000000001744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Resuscitative endovascular balloon occlusion of the aorta (REBOA) is a lifesaving technique for the management of lethal torso hemorrhage. Its benefit, however, must be weighed against the lethal distal organ ischemia-reperfusion injury (IRI). This study uses a novel direct gut cooling technique to manage the distal organ IRI. METHODS A rat lethal hemorrhage model was established by bleeding of 50% of the estimated total blood volume via inferior vena cava. A novel TransRectal Intra-Colon (TRIC) temperature management device was positioned in the descending colon either to maintain intra-colon temperature at 37°C or 12°C. The upper body temperature was maintained at as close to 37°C as possible in both groups. A 2F Fogarty balloon catheter was inserted via the femoral artery into the descending thoracic aorta for the implementation of REBOA. After REBOA, the balloon was deflated, and the shed blood was returned. The temperature managements were continued for additional 180 to 270 min during the post-REBOA period. RESULTS All rats subjected to REBOA management of lethal hemorrhage at 37°C had severe histopathological gut and abdominal organ IRI, severe functional deficits, and died within 24 h with 100% mortality. By contrast, directly cooling the colon to 10°C to 12°C with the novel TRIC device abolished mortality, and dramatically improved ABG parameters, prevented the abdominal organ injury, and reduced the functional deficits during the 7-day post-REBOA period. CONCLUSIONS Direct trans-rectal colon cooling during REBOA management of lethal hemorrhage offers extraordinary functional improvement and amazing tissue protection, and abolishes mortality.
Collapse
Affiliation(s)
- Chunli Liu
- Veterans Affairs Maryland Health Center System,10 North Greene Street, Baltimore, MD 21201
| | - Dong Yuan
- Departments of Anesthesiology and Surgery, Shock Trauma and Anesthesiology Research Center University of Maryland School of Medicine, Baltimore, MD
| | - Robert Crawford
- Departments of Anesthesiology and Surgery, Shock Trauma and Anesthesiology Research Center University of Maryland School of Medicine, Baltimore, MD
| | - Rajabrata Sarkar
- Departments of Anesthesiology and Surgery, Shock Trauma and Anesthesiology Research Center University of Maryland School of Medicine, Baltimore, MD
| | - Bingren Hu
- Departments of Anesthesiology and Surgery, Shock Trauma and Anesthesiology Research Center University of Maryland School of Medicine, Baltimore, MD
- Veterans Affairs Maryland Health Center System,10 North Greene Street, Baltimore, MD 21201
| |
Collapse
|
41
|
Debras C, Chazelas E, Srour B, Julia C, Schneider É, Kesse-Guyot E, Agaësse C, Druesne-Pecollo N, Andreeva VA, Wendeu-Foyet G, Galan P, Hercberg S, Deschasaux-Tanguy M, Touvier M. Fermentable Oligosaccharides, Disaccharides, Monosaccharides and Polyols (FODMAPs) and cancer risk in the prospective NutriNet-Santé cohort. J Nutr 2021; 152:1059-1069. [PMID: 36967163 DOI: 10.1093/jn/nxab379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/02/2021] [Accepted: 10/25/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Fermentable Oligosaccharides, Disaccharides and Monosaccharides And Polyols (FODMAPs) have been shown to be involved in gastrointestinal disorders. In view of their pro-inflammatory potential and their interactions with the gut microbiota, their contribution to the etiology of other chronic diseases such as cancer has been postulated. However, no epidemiological study has investigated this hypothesis so far. OBJECTIVE Our objective was to investigate the associations between FODMAP intake (total and by type) and cancer risk (overall, breast, prostate and colorectal) in a large prospective cohort. DESIGN The study was based on the NutriNet-Santé cohort (2009-2020); 104,909 adult participants without cancer at baseline were included in our analyses (median follow-up time = 7.7y, 78.7% women, mean age at baseline 42.1y (SD = 14.5)). Baseline dietary intakes were obtained from repeated 24h-dietary records linked to a detailed food composition table. Associations between FODMAP intake (expressed in quintiles, Q) and cancer risks were assessed by Cox proportional hazard models adjusted for a large range of lifestyle, sociodemographic and anthropometric variables. RESULTS Total FODMAP intake was associated with increased overall cancer risk (n = 3374 incident cases, HR for sex-specific Quintile 5 versus Quintile 1: 1.21; 95%CI: 1.02, 1.44; P-trend = 0.04). In particular, oligosaccharides were associated with cancer risk: a trend was observed for overall cancer (HR Q5 vs. Q1: 1.10; 95%CI: 0.97, 1.25; P-trend = 0.04) and colorectal cancer (n = 272, HR Q5 vs. Q1: 1.78; 95%CI: 1.13-2.79; P-trend = 0.02). CONCLUSION Results from this large population-based study on French adults from the NutriNet-Santé cohort show a significant association between FODMAP intake and the risk of cancer development. Further epidemiological and experimental studies are needed to confirm these results and provide data on the potential underlying mechanisms.
Collapse
Affiliation(s)
- Charlotte Debras
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France.
| | - Eloi Chazelas
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| | - Bernard Srour
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| | - Chantal Julia
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; Public Health Department, Avicenne Hospital, AP-HP, Bobigny, France
| | - Élodie Schneider
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France
| | - Emmanuelle Kesse-Guyot
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| | - Cédric Agaësse
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France
| | - Nathalie Druesne-Pecollo
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| | - Valentina A Andreeva
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France
| | - Gaëlle Wendeu-Foyet
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| | - Pilar Galan
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France
| | - Serge Hercberg
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France; Public Health Department, Avicenne Hospital, AP-HP, Bobigny, France
| | - Mélanie Deschasaux-Tanguy
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| | - Mathilde Touvier
- Sorbonne Paris Nord University, INSERM U1153, INRAe U1125, CNAM, Nutritional Epidemiology Research Team (EREN), Epidemiology and Statistics Research Center, University of Paris (CRESS), Bobigny, France; French Network for Nutrition AND Cancer Research (NACRe network), Jouy-en-Josas, France
| |
Collapse
|
42
|
Andrei S, Nguyen M, Berthoud V, Morgant MC, Bouhemad B, Guinot PG. Evaluation of the Oxiris Membrane in Cardiogenic Shock Requiring Extracorporeal Membrane Oxygenation Support: Study Protocol for a Single Center, Single-Blind, Randomized Controlled Trial. Front Cardiovasc Med 2021; 8:738496. [PMID: 34708091 PMCID: PMC8544809 DOI: 10.3389/fcvm.2021.738496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Veno-arterial extracorporeal membrane oxygenation (VA-ECMO) is the rescue treatment proposed to patients with refractory cardiogenic shock. The VA-ECMO implantation promotes inflammation and ischemia-reperfusion injuries through the VA-ECMO flow, causing digestive mucosa barrier disrupture and inducing translocation of bacterial wall components-Lipopolysaccharides (LPS) with further inflammation and circulatory impairment. LPS is a well-studied surrogate indicator of bacterial translocation. Oxiris membrane is a promising and well-tolerated device that can specifically remove LPS. The main study aim is to compare the LPS elimination capacity of Oxiris membrane vs. a non-absorbant classical renal replacement (RRT) membrane in patients with cardiogenic shock requiring VA-ECMO. Methods: ECMORIX is a randomized, prospective, single-center, single-blind, parallel-group, controlled study. It compares the treatment with Oxiris membrane vs. the standard continuous renal replacement therapy care in patients with cardiogenic shock support by peripheral VA-ECMO. Forty patients will be enrolled in both treatment groups. The primary endpoint is the value of LPS serum levels after 24 h of treatment. LPS serum levels will be monitored during the first 72 h of treatment, as clinical and cardiac ultrasound parameters, biological markers of inflammation and 30-day mortality. Discussion: Oxiris membrane appears to be beneficial in controlling the VA-ECMO-induced ischemia-reperfusion inflammation by LPS removal. ECMORIX results will be of major importance in the management of severe cases requiring VA-ECMO and will bring pathophysiological insights about the LPS role in this context. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT04886180.
Collapse
Affiliation(s)
- Stefan Andrei
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- Anaesthesiology and Critical Care Department, Carol Davila University of Medicine, Bucharest, Romania
| | - Maxime Nguyen
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- University of Burgundy Franche Comté, Dijon, France
| | - Vivien Berthoud
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
| | | | - Belaid Bouhemad
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- University of Burgundy Franche Comté, Dijon, France
| | - Pierre-Grégoire Guinot
- Anaesthesiology and Critical Care Department, Dijon Bourgogne University Hospital, Dijon, France
- University of Burgundy Franche Comté, Dijon, France
| |
Collapse
|
43
|
Zhang X, Li N, Chen Q, Qin H. Fecal Microbiota Transplantation Modulates the Gut Flora Favoring Patients With Functional Constipation. Front Microbiol 2021; 12:700718. [PMID: 34690948 PMCID: PMC8529243 DOI: 10.3389/fmicb.2021.700718] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal dysmotility is common in many diseases and is correlated with gut microbiota dysbiosis and systemic inflammation. Functional constipation (FC) is the most typical manifestation of intestinal hypomotility and reduces patients' quality of life. Some studies have reported that fecal micriobiota transplantation (FMT) may be an effective and safe therapy for FC as it corrects intestinal dysbiosis. This study was conducted to evaluate how FMT remodels the gut microbiome and to determine a possible correlation between certain microbes and clinical symptoms in constipated individuals. Data were retrospectively collected on 18 patients who underwent FMT between January 1, 2019 and June 30, 2020. The fecal bacterial genome was detected by sequencing the V3-V4 hypervariable regions of the 16S rDNA gene. Fecal short chain fatty acids (SCFAs) were detected by gas chromatography-mass spectrometry, and serum inflammatory factor concentrations were detected via enzyme-linked immunosorbent assay. Comparing the changes in fecal microbiome compositions before and after FMT revealed a significant augmentation in the alpha diversity and increased abundances of some flora such as Clostridiales, Fusicatenibacter, and Paraprevotella. This was consistent with the patients experiencing relief from their clinical symptoms. Abundances of other flora, including Lachnoanaerobaculum, were decreased, which might correlate with the severity of patients' constipation. Although no differences were found in SCFA production, the butyric acid concentration was correlated with both bacterial alterations and clinical symptoms. Serum IL-8 levels were significantly lower after FMT than at baseline, but IL-4, IL-6, IL-10, and IL-12p70 levels were not noticeably changed. This study showed how FMT regulates the intestinal microenvironment and affects systemic inflammation in constipated patients, providing direction for further research on the mechanisms of FMT. It also revealed potential microbial targets for precise intervention, which may bring new breakthroughs in treating constipation.
Collapse
Affiliation(s)
| | | | - Qiyi Chen
- Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huanlong Qin
- Intestinal Microenvironment Treatment Center, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
44
|
Seilitz J, Grafver I, Kiszakiewicz L, Oikonomakis I, Jansson K, Axelsson B, Nilsson KF. A Randomized Porcine Study in Low Cardiac Output of Vasoactive and Inotropic Drug Effects on the Gastrointestinal Tract. Shock 2021; 56:308-317. [PMID: 33443363 PMCID: PMC8529897 DOI: 10.1097/shk.0000000000001726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/22/2020] [Accepted: 01/07/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Splanchnic vasodilation by inodilators is an argument for their use in critical cardiac dysfunction. To isolate peripheral vasoactivity from inotropy, such drugs were investigated, and contrasted to vasopressors, in a fixed low cardiac output (CO) model resembling acute cardiac dysfunction effects on the gastrointestinal tract. We hypothesized that inodilators would vasodilate and preserve the aerobic metabolism in the splanchnic circulation in low CO. METHODS In anesthetized pigs, CO was lowered to 60% of baseline by partial inferior caval vein balloon inflation. The animals were randomized to placebo (n = 8), levosimendan (24 μg kg-1 bolus, 0.2 μg kg-1 min-1, n = 7), milrinone (50 μg kg-1 bolus, 0.5 μg kg-1 min-1, n = 7), vasopressin (0.001, 0.002 and 0.006 U kg-1 min-1, 1 h each, n = 7) or norepinephrine (0.04, 0.12, and 0.36 μg kg-1 min-1, 1 h each, n = 7). Hemodynamic variables including mesenteric blood flow were collected. Systemic, mixed-venous, mesenteric-venous, and intraperitoneal metabolites were analyzed. RESULTS Cardiac output was stable at 60% in all groups, which resulted in systemic hypotension, low superior mesenteric artery blood flow, lactic acidosis, and increased intraperitoneal concentrations of lactate. Levosimendan and milrinone did not change any circulatory variables, but levosimendan increased blood lactate concentrations. Vasopressin and norepinephrine increased systemic and mesenteric vascular resistances at the highest dose. Vasopressin increased mesenteric resistance more than systemic, and the intraperitoneal lactate concentration and lactate/pyruvate ratio. CONCLUSION Splanchnic vasodilation by levosimendan and milrinone may be negligible in low CO, thus rejecting the hypothesis. High-dose vasopressors may have side effects in the splanchnic circulation.
Collapse
Affiliation(s)
- Jenny Seilitz
- Department of Cardiothoracic and Vascular Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Isabelle Grafver
- Department of Cardiothoracic and Vascular Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Lars Kiszakiewicz
- Department of Anaesthesiology and Intensive Care, Skaraborg Hospital, Skövde, Sweden
| | - Ioannis Oikonomakis
- Department of Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Kjell Jansson
- Department of Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Birger Axelsson
- Department of Cardiothoracic and Vascular Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Kristofer F. Nilsson
- Department of Cardiothoracic and Vascular Surgery, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
45
|
Mishra A, Singh KP. Protective effect of neurotensin receptor-1 agonist PD 149163 against lipopolysaccharide-induced gut toxicity in mice. Drug Chem Toxicol 2021; 45:2399-2410. [PMID: 34334065 DOI: 10.1080/01480545.2021.1954698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The interaction between neuroendocrine and immune components of the gut maintains the organism's physical and psychological health. Its disruption may reflect in disease conditions such as inflammatory bowel disease (IBD) and mental illness. The lipopolysaccharide (LPS) disrupts the endocrine-immune homeostasis resulting in gut toxicity. The Neurotensin receptor-1 (NTR-1) agonist PD 149163 (PD) acts as an atypical antipsychotic drug in psychiatric illness, but its role in modulating gut pathophysiology remains unknown. Therefore, the aim of the present study was to evaluate the protective effect of PD against LPS-induced gut toxicity. Swiss albino female mice (12 weeks) were divided into six groups (n = 6/group): (I) Control, (II) LPS (1 mg/kg, for 5 days), (III) LPS (1 mg/kg, for 5 days)+PD low (100 µg/kg, for 21 days), (IV) LPS (1 mg/kg, for 5 days)+PD high (300 µg/kg, for 21 days), (V) PD low (100 µg/kg, for 21 days), and (VI) PD high (300 µg/kg, for 21 days). Drugs were given intraperitoneal in the morning. PD administration prevented the LPS-induced gut inflammation observed in damage of epithelial barrier, disruption of goblet cells, and condensation of lamina propria (LP). The LPS-induced oxidative stress characterized by decreased superoxide dismutase (SOD) activity and increased lipid hydroperoxide (LOOH) (p < 0.001 for both), and enhanced interleukine-6 (IL-6) & tumor necrosis factor-α (TNF-α) (p < 0.001 for both) as well as immunointensity of NT (p < 0.01) and NTR-1 (p < 0.05) were reversed and normalized to control after PD treatment. Thus, the anti-inflammatory, anti-oxidative, and cell proliferation properties of PD modulate the gut toxicity in LPS-challenged mice.
Collapse
Affiliation(s)
- Ankit Mishra
- Neurobiology Lab, Department of Zoology, University of Allahabad, Prayagraj, India
| | - K P Singh
- Neurobiology Lab, Department of Zoology, University of Allahabad, Prayagraj, India
| |
Collapse
|
46
|
Honore PM, Redant S, Preseau T, Moorthamers S, Kaefer K, Barreto Gutierrez L, Attou R, Gallerani A, De Bels D. Studying polymyxin B-immobilized hemoperfusion therapy to remove endotoxin in infection with GN bacteria has conflicting results: We are not sure! J Crit Care 2021; 65:228-229. [PMID: 34252651 DOI: 10.1016/j.jcrc.2021.06.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Patrick M Honore
- ICU Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium; Faculty of Medicine of the ULB University, Belgium.
| | - Sebastien Redant
- ICU Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium.
| | - Thierry Preseau
- Emergency Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium.
| | - Sofie Moorthamers
- Emergency Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium.
| | - Keitiane Kaefer
- ICU Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium
| | | | - Rachid Attou
- ICU Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium.
| | - Andrea Gallerani
- ICU Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium.
| | - David De Bels
- ICU Dept, Centre Hospitalier Universitaire Brugmann, Brussels, Belgium.
| |
Collapse
|
47
|
Arya AK, Hu K, Subedi L, Li T, Hu B. Focal intra-colon cooling reduces organ injury and systemic inflammation after REBOA management of lethal hemorrhage in rats. Sci Rep 2021; 11:13696. [PMID: 34211011 PMCID: PMC8249469 DOI: 10.1038/s41598-021-93064-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/14/2021] [Indexed: 12/04/2022] Open
Abstract
Resuscitative endovascular balloon occlusion of the aorta (REBOA) is a lifesaving maneuver for the management of lethal torso hemorrhage. However, its prolonged use leads to distal organ ischemia-reperfusion injury (IRI) and systemic inflammatory response syndrome (SIRS). The objective of this study is to investigate the blood-based biomarkers of IRI and SIRS and the efficacy of direct intestinal cooling in the prevention of IRI and SIRS. A rat lethal hemorrhage model was produced by bleeding 50% of the total blood volume. A balloon catheter was inserted into the aorta for the implementation of REBOA. A novel TransRectal Intra-Colon (TRIC) device was placed in the descending colon and activated from 10 min after the bleeding to maintain the intra-colon temperature at 37 °C (TRIC37°C group) or 12 °C (TRIC12°C group) for 270 min. The upper body temperature was maintained at as close to 37 °C as possible in both groups. Blood samples were collected before hemorrhage and after REBOA. The organ injury biomarkers and inflammatory cytokines were evaluated by ELISA method. Blood based organ injury biomarkers (endotoxin, creatinine, AST, FABP1/L-FABP, cardiac troponin I, and FABP2/I-FABP) were all drastically increased in TRIC37°C group after REBOA. TRIC12°C significantly downregulated these increased organ injury biomarkers. Plasma levels of pro-inflammatory cytokines TNF-α, IL-1b, and IL-17F were also drastically increased in TRIC37°C group after REBOA. TRIC12°C significantly downregulated the pro-inflammatory cytokines. In contrast, TRIC12°C significantly upregulated the levels of anti-inflammatory cytokines IL-4 and IL-10 after REBOA. Amazingly, the mortality rate was 100% in TRIC37°C group whereas 0% in TRIC12°C group after REBOA. Directly cooling the intestine offered exceptional protection of the abdominal organs from IRI and SIRS, switched from a harmful pro-inflammatory to a reparative anti-inflammatory response, and mitigated mortality in the rat model of REBOA management of lethal hemorrhage.
Collapse
Affiliation(s)
- Awadhesh K Arya
- Departments of Anesthesiology, Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kurt Hu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lalita Subedi
- Departments of Anesthesiology, Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tieluo Li
- Departments of Anesthesiology, Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bingren Hu
- Departments of Anesthesiology, Shock, Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, USA.
- Veterans Affairs Maryland Health Center System, 10 North Greene Street, Baltimore, MD, USA.
| |
Collapse
|
48
|
Abstract
This review attempts to unveil the possible mechanisms underlying how gut lymph affects lung and further gives rise to acute respiratory distress syndrome, as well as potential interventional targets under the condition of ischemia-reperfusion injury. We searched electronic databases including PubMed, MEDLINE, Cochrane Central Register of Controlled Trials, Google Scholar, Web of Science, and Embase to identify relevant literatures published up to December 2019. We enrolled the literatures including the Mesh Terms of “gut lymph or intestinal lymph and acute lung injury or acute respiratory distress syndrome.” Gut is considered to be the origin of systemic inflammation and the engine of multiple organ distress syndrome in the field of critical care medicine, whereas gut lymph plays a pivotal role in initiation of ischemia-reperfusion injury-induced acute respiratory distress syndrome. In fact, in the having been established pathologic model of sepsis leading to multiple organ dysfunction named by Gut Lymph theory, a variety of literatures showed the position and role of changes in gut lymph components in the initiation of systemic inflammatory response, which allows us to screen out potential intervention targets to pave the way for future clinic and basic research.
Collapse
|
49
|
Arias AV, Lucas DJ, Shafi NI. Respiratory Syncytial Virus Bronchiolitis Complicated by Necrotizing Enterocolitis: A Case Series. Pediatrics 2021; 147:peds.2020-022707. [PMID: 33824182 DOI: 10.1542/peds.2020-022707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2020] [Indexed: 11/24/2022] Open
Abstract
In rare instances, severe respiratory syncytial virus (RSV) infections of the lower respiratory tract can cause life-threatening extrapulmonary complications. In this report, we describe 4 previously healthy, term and late-preterm infants admitted to the PICU with respiratory failure due to RSV bronchiolitis who developed necrotizing enterocolitis shortly after admission. All infants exhibited progressive abdominal distention, had typical radiographic findings, and developed simple or complex ascites. In addition to being managed with broad-spectrum antibiotics and bowel rest, 1 infant was treated with colon resection and ileostomy, 2 had peritoneal drainage procedures for ascites, and one of those later developed small bowel strictures treated with delayed resection and anastomosis. Three were discharged from the hospital without further complications; 1 died of septic shock. In this case series, we describe development of necrotizing enterocolitis in otherwise healthy neonates with severe RSV disease in the absence of traditional risk factors. We hypothesize that a dysregulated proinflammatory response associated with severe RSV disease may alter intestinal blood flow and compromise barriers to bacterial translocation. Enteral feeding intolerance, septic ileus, and/or complex ascites may represent important clinical corollaries in these patients.
Collapse
Affiliation(s)
- Anita V Arias
- Division of Pediatric Critical Care, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, Tennessee;
| | - Donald J Lucas
- Division of Pediatric Surgery, Naval Medical Center San Diego, California; and.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Nadeem I Shafi
- Division of Pediatric Critical Care, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, Tennessee
| |
Collapse
|
50
|
Sun X, Zhu S, Tonnessen TI, Yang R. Bile is a promising gut nutrient that inhibits intestinal bacterial translocation and promotes gut motility via an interleukin-6-related pathway in an animal model of endotoxemia. Nutrition 2021; 84:111064. [PMID: 33418232 DOI: 10.1016/j.nut.2020.111064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES People who are critically ill have high rates of endotoxemia that can significantly decrease bile flow and increase bile cytokines, the latter of which might worsen their condition. Bile acids are nutrient-signaling hormones that have a significant impact on gut barrier function and motility, and the gut is considered the origin of systemic inflammation. Therefore, healthy exogenous bile could be a promising gut nutrient for critical illness, so the biomedical role of bile in endotoxemia was investigated in this study. METHODS Twelve rats were injected with lipopolysaccharide (LPS) and randomized into a group with sham operation) and a group with bile external drainage (n = 6 for each group); six rats with sham operation served as the control group. In addition, interleukin-6 (IL-6) knockout mice and macrophages were treated with LPS. RESULTS Compared to the control animals, the group with LPS injection and sham operation had significantly increased levels of gut permeability, gut bacterial translocation, gut mucosal tumor necrosis factor α, IL-6 transcripts, and serum tumor necrosis factor α and IL-6. Compared to group with sham operation and LPS injection, bile external drainage (in LPS-challenged rats) increased gut bacterial translocation by 10 times, and this detrimental effect was associated with prolonged intestinal transit time, increased serum IL-6 concentration, and up-regulated gut mucosal IL-6 transcripts. Moreover, bile selectively inhibited LPS-stimulated macrophages in IL-6 release, which can activate gastrointestinal submucosal neurons to promote motility. Knocking out IL-6 significantly reduced gut bacterial translocation in endotoxemic mice. CONCLUSIONS Bile is a promising gut nutrient that inhibits gut bacterial translocation and promotes gut motility via an IL-6-related pathway in experimental endotoxemia.
Collapse
Affiliation(s)
- Xiujing Sun
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tor Inge Tonnessen
- Department of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Runkuan Yang
- Department of Emergencies and Critical Care, Oslo University Hospital, Oslo, Norway; Department of Critical Care Medicine, University of Pittsburgh Medical School, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|