1
|
Keirns BH, Medlin AR, Maki KA, McClanahan K, Fruit SE, Sciarrillo CM, Hart SM, Joyce J, Lucas EA, Emerson SR. Biomarkers of intestinal permeability are associated with inflammation in metabolically healthy obesity but not normal-weight obesity. Am J Physiol Heart Circ Physiol 2024; 327:H1135-H1145. [PMID: 39212768 DOI: 10.1152/ajpheart.00381.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Systemic inflammation is reported in normal-weight obesity (NWO) and metabolically healthy obesity (MHO), which may be linked to their increased cardiovascular disease (CVD) risk. Yet, drivers of this inflammation remain unclear. We characterized factors known to influence inflammatory status (i.e., intestinal permeability, adipose tissue, diet quality, microbiota), and their relationships with measured inflammation, in NWO and MHO, healthy control subjects (CON), and metabolically unhealthy obesity (MUO; N = 80; n = 20/group). Serum indicators of intestinal permeability and inflammation were assessed by ELISA and/or multiplex. Total, visceral, and percent body fat were measured with dual-energy X-ray absorptiometry (DXA). Fecal microbiota composition was assessed via 16S rRNA sequencing (n = 9-10/group). For C-reactive protein (CRP), MUO > NWO > CON (P < 0.0001). In MHO, CRP was intermediate and similar to both MUO and NWO. Lipopolysaccharide binding protein (LBP) and the ratio of LBP to soluble CD14 (sCD14) were higher in MHO and MUO vs. CON/NWO (P < 0.0001). Across correlation and regression analyses, LBP consistently displayed the strongest relationships with CRP in the entire sample (r = 0.78; β = 0.57; P < 0.0001) and in MHO (r = 0.74; P < 0.01) but not NWO (r = 0.37; P = 0.11). Shannon index was higher in CON compared with MUO (P < 0.05) and inversely correlated with CRP in the full sample (r = -0.37; P < 0.05). These data are consistent with the notion that intestinal permeability is associated with low-grade inflammation in MHO, which could be implicated in this population's reported CVD risk.NEW & NOTEWORTHY This is the first study to our knowledge to examine biomarkers of intestinal permeability in normal-weight obesity and one of few assessing microbiota compositions in this population. Additionally, we report that individuals with metabolically healthy obesity and metabolically unhealthy obesity displayed similar evidence of intestinal permeability, which was more strongly associated with systemic inflammation than total and visceral adipose tissue mass.
Collapse
Affiliation(s)
- Bryant H Keirns
- Department of Nutrition and Health Science, Ball State University, Muncie, Indiana, United States
| | - Austin R Medlin
- Department of Health & Wellness Design, Indiana University School of Public Health, Bloomington, Indiana, United States
| | - Katherine A Maki
- Translational Biobehavioral and Health Disparities Branch, National Institutes of Health Clinical Center, Bethesda, Maryland, United States
| | - Kristen McClanahan
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Sarah E Fruit
- Department of Nutrition and Health Science, Ball State University, Muncie, Indiana, United States
| | - Christina M Sciarrillo
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Samantha M Hart
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Jill Joyce
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Edralin A Lucas
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| | - Sam R Emerson
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, Oklahoma, United States
| |
Collapse
|
2
|
Kang J, Lee M, Park M, Lee J, Lee S, Park J, Koyanagi A, Smith L, Nehs CJ, Yon DK, Kim T. Slow gut transit increases the risk of Alzheimer's disease: An integrated study of the bi-national cohort in South Korea and Japan and Alzheimer's disease model mice. J Adv Res 2024; 65:283-295. [PMID: 38097171 PMCID: PMC11518944 DOI: 10.1016/j.jare.2023.12.010] [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: 09/17/2023] [Revised: 11/20/2023] [Accepted: 12/12/2023] [Indexed: 01/02/2024] Open
Abstract
INTRODUCTION Although the association between Alzheimer's disease (AD) and constipation is controversial, its causality and underlying mechanisms remain unknown. OBJECTIVES To investigate the potential association between slow gut transit and AD using epidemiological data and a murine model. METHODS We conducted a bi-national cohort study in South Korea (discovery cohort, N=3,130,193) and Japan (validation cohort, N=4,379,285) during the pre-observation period to determine the previous diagnostic history (2009-2010) and the follow-up period (2011-2021). To evaluate the causality, we induced slow gut transit using loperamide in 5xFAD transgenic mice. Changes in amyloid-beta (Aβ) and other markers were examined using ELISA, qRT-PCR, RNA-seq, and behavioral tests. RESULTS Constipation was associated with an increased risk of AD in the discovery cohort (hazard ratio, 2.04; 95% confidence interval [CI], 2.01-2.07) and the validation cohort (hazard ratio; 2.82; 95% CI, 2.61-3.05). We found that loperamide induced slower gut transit in 5xFAD mice, increased Aβ and microglia levels in the brain, increased transcription of genes related to norepinephrine secretion and immune responses, and decreased the transcription of defense against bacteria in the colonic tissue. CONCLUSION Impaired gut transit may contribute to AD pathogenesis via the gut-brain axis, thus suggesting a cyclical relationship between intestinal barrier disruption and Aβ accumulation in the brain. We propose that gut transit or motility may be a modifiable lifestyle factor in the prevention of AD, and further clinical investigations are warranted.
Collapse
Affiliation(s)
- Jiseung Kang
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea; Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, United States; Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Myeongcheol Lee
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea; Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Mincheol Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jibeom Lee
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Jaeyu Park
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea; Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea
| | - Ai Koyanagi
- Research and Development Unit, Parc Sanitari Sant Joan de Deu, Barcelona, Spain
| | - Lee Smith
- Centre for Health, Performance and Wellbeing, Anglia Ruskin University, Cambridge, UK
| | - Christa J Nehs
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, United States; Division of Sleep Medicine, Harvard Medical School, Boston, MA, United States
| | - Dong Keon Yon
- Center for Digital Health, Medical Science Research Institute, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea; Department of Regulatory Science, Kyung Hee University, Seoul, Republic of Korea; Department of Pediatrics, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul, Republic of Korea.
| | - Tae Kim
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea.
| |
Collapse
|
3
|
Ahmadi S, Hasani A, Khabbaz A, Poortahmasbe V, Hosseini S, Yasdchi M, Mehdizadehfar E, Mousavi Z, Hasani R, Nabizadeh E, Nezhadi J. Dysbiosis and fecal microbiota transplant: Contemplating progress in health, neurodegeneration and longevity. Biogerontology 2024; 25:957-983. [PMID: 39317918 DOI: 10.1007/s10522-024-10136-4] [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: 05/21/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
The gut-brain axis plays an important role in mental health. The intestinal epithelial surface is colonized by billions of commensal and transitory bacteria, known as the Gut Microbiota (GM). However, potential pathogens continuously stimulate intestinal immunity when they find the place. The last two decades have witnessed several studies revealing intestinal bacteria as a key factor in the health-disease balance of the gut, as well as disease-emergent in other parts of the body. Various neurological processes, such as cognition, learning, and memory, could be affected by dysbiosis in GM. Additionally, the aging process and longevity are related to systemic inflammation caused by dysbiosis. Commensal GM affects brain development, behavior, and healthy aging suggesting that building changes in GM might be a potential therapeutic method. The innovation in GM dysbiosis is intervention by Fecal Microbiota Transplantation (FMT), which has been confirmed as a therapy for recurrent Clostridium difficile infections and is promising for other clinical disorders, such as Parkinson's disease, Multiple Sclerosis (MS), Alzheimer's disease, and depression. Additionally, FMT may be possible to promote healthy aging, and extend longevity. This review aims to connect dysbiosis, neurological disorders, and aging and the potential of FMT as a therapeutic strategy to treat these disorders, and to enhance the quality of life in the elderly.
Collapse
Affiliation(s)
- Somayeh Ahmadi
- Infectious and Tropical Diseases Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Students Research Committee, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alka Hasani
- Infectious and Tropical Diseases Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Clinical Research Development Unit, Sina Educational, Research and Treatment Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Aytak Khabbaz
- Neurosciences Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasbe
- Infectious and Tropical Diseases Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samaneh Hosseini
- Neurosciences Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Yasdchi
- Neurosciences Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Mehdizadehfar
- Neurosciences Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Mousavi
- Department of Psychology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roqaiyeh Hasani
- School of Medicine, Istanbul Okan University, Tuzla, 34959, Istanbul, Turkey
| | - Edris Nabizadeh
- Infectious and Tropical Diseases Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Nezhadi
- Infectious and Tropical Diseases Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Wang S, Liu Y, Liu S, Qin Z, Lu J, Zhang R, Yuan H. Consensus gene co-expression analysis across multiple intestinal tissues to identify key genes and pathways associated with abdominal fat deposition in broilers. Br Poult Sci 2024:1-11. [PMID: 39466128 DOI: 10.1080/00071668.2024.2410367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/28/2024] [Indexed: 10/29/2024]
Abstract
1. Abdominal fat deposition (AFD) is regulated by multiple intestinal tissues, and changes in the function of intestinal tissues are associated with AFD. Currently, integration of transcriptomic data across multiple intestinal tissues to explore excessive AFD has rarely been reported in broilers.2. In this study, a consensus gene co-expression network across the duodenum, jejunum, ileum and caecum of high- and low-abdominal fat broiler lines (HL and LL) was constructed using a publicly available transcriptomic data set. Combining the results of functional enrichment analyses and differential gene expression analyses, this investigated the genes and biological pathways across the four intestinal tissues that might influence AFD.3. In one expression module, NDUFA5, NDUFS6, NDUFA4, NDUFS4, ATP5H, ATP5J and ATP5C1 were significantly enriched in the oxidative phosphorylation pathway, with GPX2 and GSR significantly enriched in the glutathione metabolism pathway. These genes were significantly downregulated in the four intestinal tissues of the HL compared to LL chickens, which may be associated with AFD by increasing intestinal permeability.4. Lipid metabolism relevant genes were identified in other modules (ALDH7A1, ACSBG1, THEM4 and DECR1), which may be linked to AFD through regulation of lipid metabolism. Interestingly, in the first module, 12 genes were significantly enriched in the proteasome pathway and significantly downregulated in the four intestinal tissues in HL birds compared to LL birds, indicating a link between the proteasome and AFD.
Collapse
Affiliation(s)
- S Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Y Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - S Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Z Qin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - J Lu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - R Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - H Yuan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, Heilongjiang, China
| |
Collapse
|
5
|
Partida-Rodríguez O, Brown EM, Woodward SE, Cirstea M, Reynolds LA, Petersen C, Vogt SL, Peña-Díaz J, Thorson L, Arrieta MC, Hernández EG, Rojas-Velázquez L, Moran P, González Rivas E, Serrano-Vázquez A, Pérez-Juárez H, Torres J, Ximénez C, Finlay BB. Fecal microbiota transplantation from protozoa-exposed donors downregulates immune response in a germ-free mouse model, its role in immune response and physiology of the intestine. PLoS One 2024; 19:e0312775. [PMID: 39466773 PMCID: PMC11515975 DOI: 10.1371/journal.pone.0312775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024] Open
Abstract
Intestinal parasites are part of the intestinal ecosystem and have been shown to establish close interactions with the intestinal microbiota. However, little is known about the influence of intestinal protozoa on the regulation of the immune response. In this study, we analyzed the regulation of the immune response of germ-free mice transplanted with fecal microbiota (FMT) from individuals with multiple parasitic protozoans (P) and non-parasitized individuals (NP). We determined the production of intestinal cytokines, the lymphocyte populations in both the colon and the spleen, and the genetic expression of markers of intestinal epithelial integrity. We observed a general downregulation of the intestinal immune response in mice receiving FMT-P. We found significantly lower intestinal production of the cytokines IL-6, TNF, IFN-γ, MCP-1, IL-10, and IL-12 in the FMT-P. Furthermore, a significant decrease in the proportion of CD3+, CD4+, and Foxp3+ T regulatory cells (Treg) was observed in both, the colon and spleen with FMT-P in contrast to FMT-NP. We also found that in FMT-P mice there was a significant decrease in tjp1 expression in all three regions of the small intestine; ocln in the ileum; reg3γ in the duodenum and relmβ in both the duodenum and ileum. We also found an increase in colonic mucus layer thickness in mice colonized with FMT-P in contrast with FMT-NP. Finally, our results suggest that gut protozoa, such as Blastocystis hominis, Entamoeba coli, Endolimax nana, Entamoeba histolytica/E. dispar, Iodamoeba bütschlii, and Chilomastix mesnili consortia affect the immunoinflammatory state and induce functional changes in the intestine via the gut microbiota. Likewise, it allows us to establish an FMT model in germ-free mice as a viable alternative to explore the effects that exposure to intestinal parasites could have on the immune response in humans.
Collapse
Affiliation(s)
- Oswaldo Partida-Rodríguez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Eric M. Brown
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sarah E. Woodward
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa A. Reynolds
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Microbiology, Faculty of Science, University of Victoria, Victoria, Canada
| | - Charisse Petersen
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Stefanie L. Vogt
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Jorge Peña-Díaz
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Lisa Thorson
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Marie-Claire Arrieta
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Eric G. Hernández
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Liliana Rojas-Velázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Patricia Moran
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Enrique González Rivas
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Angélica Serrano-Vázquez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Horacio Pérez-Juárez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas y Parasitarias, Instituto Mexicano del Seguro Social (IMSS), Mexico, Mexico
| | - Cecilia Ximénez
- Unidad de Investigación en Medicina Experimental, Hospital General de Mexico, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - B. B. Finlay
- Michael Smith Laboratories, Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, Faculty of Science, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
6
|
Yang X, Fang Z, Shang L. Functional PEGylated bacteria reinforce the intestinal mucosal barrier by effective mucus-penetration. Sci Bull (Beijing) 2024; 69:2964-2966. [PMID: 39138112 DOI: 10.1016/j.scib.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Affiliation(s)
- Xinyuan Yang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zhonglin Fang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
7
|
Piccioni A, Spagnuolo F, Candelli M, Voza A, Covino M, Gasbarrini A, Franceschi F. The Gut Microbiome in Sepsis: From Dysbiosis to Personalized Therapy. J Clin Med 2024; 13:6082. [PMID: 39458032 PMCID: PMC11508704 DOI: 10.3390/jcm13206082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/21/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Sepsis is a complex clinical syndrome characterized by an uncontrolled inflammatory response to an infection that may result in septic shock and death. Recent research has revealed a crucial link between sepsis and alterations in the gut microbiota, showing that the microbiome could serve an essential function in its pathogenesis and prognosis. In sepsis, the gut microbiota undergoes significant dysbiosis, transitioning from a beneficial commensal flora to a predominance of pathobionts. This transformation can lead to a dysfunction of the intestinal barrier, compromising the host's immune response, which contributes to the severity of the disease. The gut microbiota is an intricate system of protozoa, fungi, bacteria, and viruses that are essential for maintaining immunity and metabolic balance. In sepsis, there is a reduction in microbial heterogeneity and a predominance of pathogenic bacteria, such as proteobacteria, which can exacerbate inflammation and negatively influence clinical outcomes. Microbial compounds, such as short-chain fatty acids (SCFAs), perform a crucial task in modulating the inflammatory response and maintaining intestinal barrier function. However, the role of other microbiota components, such as viruses and fungi, in sepsis remains unclear. Innovative therapeutic strategies aim to modulate the gut microbiota to improve the management of sepsis. These include selective digestive decontamination (SDD), probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT), all of which have shown potential, although variable, results. The future of sepsis management could benefit greatly from personalized treatment based on the microbiota. Rapid and easy-to-implement tests to assess microbiome profiles and metabolites associated with sepsis could revolutionize the disease's diagnosis and management. These approaches could not only improve patient prognosis but also reduce dependence on antibiotic therapies and promote more targeted and sustainable treatment strategies. Nevertheless, there is still limited clarity regarding the ideal composition of the microbiota, which should be further characterized in the near future. Similarly, the benefits of therapeutic approaches should be validated through additional studies.
Collapse
Affiliation(s)
- Andrea Piccioni
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Fabio Spagnuolo
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Marcello Candelli
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
| | - Antonio Voza
- Department of Emergency Medicine, IRCCS-Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Marcello Covino
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Antonio Gasbarrini
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Medical and Surgical Science Department, Fondazione Policlinico Universitario A. Gemelli-IRCCS, 00168 Rome, Italy
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy; (A.P.); (M.C.); (M.C.); (F.F.)
- Faculty of Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| |
Collapse
|
8
|
Shanmugasundaram S, Karmakar S. Excess dietary sugar and its impact on periodontal inflammation: a narrative review. BDJ Open 2024; 10:78. [PMID: 39379356 PMCID: PMC11461508 DOI: 10.1038/s41405-024-00265-w] [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: 07/26/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION Sugar is omnipresent in the current food environment and sugar consumption has drastically risen over the past century. Extensive evidence highlights the negative health consequences of consuming excess dietary sugars, leading the World Health Organization (WHO) and the American Heart Association (AHA) to devise guidelines to restrict sugar intake. According to the WHO's Global Oral Health Status Report of 2022, oral diseases and severe periodontitis are a massive public health problem, and dietary sugars are a modifiable risk factor. METHODS We conducted a literature review using key databases to summarise the health effects of excessive sugar consumption and their potential role in periodontal inflammation. RESULTS AND CONCLUSION Available evidence suggests that excess dietary fructose and sucrose can cause low-grade systemic inflammation; and induce dysbiosis in both gut and the oral microbiota. Also, dietary sugar is potentially addictive and hypercaloric and its overconsumption can lead to obesity, metabolic syndrome, and other risk factors for periodontal inflammation. Hence, an unbalanced diet with excess dietary sugars holds the potential to initiate and aggravate periodontal inflammation. In the modern food environment that enables and facilitates a high-sugar diet, adopting a diverse diet and restricting sugar intake according to WHO and AHA guidelines seem beneficial to systemic and periodontal health. Since clinical evidence is limited, future research should study the effectiveness of dietary interventions that control sugar consumption in preventing and managing the global public health problem of periodontal inflammation.
Collapse
Affiliation(s)
- Shashikiran Shanmugasundaram
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India.
| | - Shaswata Karmakar
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
9
|
Feathers A, Lovasi GS, Grigoryan Z, Beem K, Datta SK, Faleck DM, Socci T, Maggi R, Swaminath A. Crohn's Disease Mortality and Ambient Air Pollution in New York City. Inflamm Bowel Dis 2024; 30:1732-1739. [PMID: 37934758 DOI: 10.1093/ibd/izad243] [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: 03/08/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND The worldwide increase in Crohn's disease (CD) has accelerated alongside rising urbanization and accompanying decline in air quality. Air pollution affects epithelial cell function, modulates immune responses, and changes the gut microbiome composition. In epidemiologic studies, ambient air pollution has a demonstrated relationship with incident CD and hospitalizations. However, no data exist on the association of CD-related death and air pollution. METHODS We conducted an ecologic study comparing the number of CD-related deaths of individuals residing in given zip codes, with the level of air pollution from nitric oxide, nitrogen dioxide, sulfur dioxide (SO2), and fine particulate matter. Air pollution was measured by the New York Community Air Survey. We conducted Pearson correlations and a Poisson regression with robust standard errors. Each pollution component was modeled separately. RESULTS There was a higher risk of CD-related death in zip codes with higher levels of SO2 (incidence rate ratio [IRR], 1.16; 95% confidence interval [CI], 1.06-1.27). Zip codes with higher percentage of Black or Latinx residents were associated with lower CD-related death rates in the SO2 model (IRR, 0.58; 95% CI, 0.35-0.98; and IRR, 0.13; 95% CI, 0.05-0.30, respectively). There was no significant association of either population density or area-based income with the CD-related death rate. CONCLUSIONS In New York City from 1993 to 2010, CD-related death rates were higher among individuals from neighborhoods with higher levels of SO2 but were not associated with levels of nitric oxide, nitrogen dioxide, and fine particulate matter. These findings raise an important and timely public health issue regarding exposure of CD patients to environmental SO2, warranting further exploration.
Collapse
Affiliation(s)
| | - Gina S Lovasi
- Urban Health Collaborative, Dornsife School of Public Health, Drexel University, Philadelphia, PA, USA
| | - Zoya Grigoryan
- Department of Internal Medicine, Lenox Hill Hospital, New York, NY, USA
| | | | - Samit K Datta
- Gastroenterology, Department at Skagit Regional Health in Mt. Vernon, WA
| | - David M Faleck
- Gastroenterology, Hepatology and Nutrition Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas Socci
- Division of Gastroenterology, Lenox Hill Hospital, New York, NY, USA
| | - Rachel Maggi
- Division of Gastroenterology, Lenox Hill Hospital, New York, NY, USA
| | - Arun Swaminath
- Division of Gastroenterology, Lenox Hill Hospital, New York, NY, USA
| |
Collapse
|
10
|
Gao Y, Guo M, Chen J, Sun Y, Wang M. A ginseng polysaccharide protects intestinal barrier integrity in high-fat diet-fed obese mice. Int J Biol Macromol 2024; 277:133976. [PMID: 39029823 DOI: 10.1016/j.ijbiomac.2024.133976] [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: 09/19/2023] [Revised: 05/14/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
A novel polysaccharide, GPH1, was extracted and isolated from ginseng. Structural analysis of GPH1 revealed a molecular weight of 7.321 × 105 Da and the presence of glucose and galactose components in a 30.2: 1 molar ratio. Results of methylation and NMR analyses indicated the GPH1 backbone consisted of →1)-α-Glc-(3→ and →1)-α-Glc-(6→. The anti-obesity activity of GPH1 was assessed by HFD-induced obesity mouse model. GPH1 was found to significantly reduced body weight, alleviated liver lipid accumulation and inflammatory damage. Meanwhile, GPH1 treatment increased the expression of tight junction proteins, including zonula occludens-1 (ZO-1) and claudin-1, while also regulating the intestinal microbiota of obese mice by promoting proliferation of beneficial bacteria with known anti-obesity effects, including s_Akkermansia muciniphila, s_Lactobacillus intestinalis, s_Lactobacillus reuteri, s_Streptococcus hyointestinalis, and s_Lactococcus garvieae. Our findings demonstrated that GPH1 is a practical natural dietary supplement with potential therapeutic effects on obesity.
Collapse
Affiliation(s)
- Yanan Gao
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130021, China; College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Mingkun Guo
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130021, China; College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jiaqi Chen
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130021, China; College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yue Sun
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130021, China; College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130021, China
| | - Mingxing Wang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun 130021, China.
| |
Collapse
|
11
|
Acharya B, Tofthagen M, Maciej-Hulme ML, Suissa MR, Karlsson NG. Limited support for a direct connection between prebiotics and intestinal permeability - a systematic review. Glycoconj J 2024; 41:323-342. [PMID: 39287885 DOI: 10.1007/s10719-024-10165-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The intestinal barrier is a selective interface between the body´s external and the internal environment. Its layer of epithelial cells is joined together by tight junction proteins. In intestinal permeability (IP), the barrier is compromised, leading to increased translocation of luminal contents such as large molecules, toxins and even microorganisms. Numerous diseases including Inflammatory Bowel Disease (IBD), Coeliac disease (CD), autoimmune disorders, and diabetes are believed to be associated with IP. Dietary interventions, such as prebiotics, may improve the intestinal barrier. Prebiotics are non-digestible food compounds, that promote the growth and activity of beneficial bacteria in the gut. This systematic review assesses the connection between prebiotic usage and IP. PubMed and Trip were used to identify relevant studies conducted between 2010-2023. Only six studies were found, which all varied in the characteristics of the population, study design, and types of prebiotics interventions. Only one study showed a statistically significant effect of prebiotics on IP. Alteration of intestinal barrier function was measured by lactulose/mannitol, chromium-labelled Ethylenediaminetetraacetic acid (51Cr-EDTA), lactulose/rhamnose, and sucralose/erythritol excretion as well as zonulin and glucagon-like peptide 2 levels. Three studies also conducted gut microbiota assessment, and one of them showed statistically significant improvement of the gut microbiome. This study also reported a decrease in zonulin level. The main conclusion from this review is that there is a lack of human studies in this important field. Futhermore, large population studies and using standardized protocols, would be required to properly assess the impact of prebiotic intervention and improvement on IP.
Collapse
Affiliation(s)
- Binayak Acharya
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marthe Tofthagen
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marissa L Maciej-Hulme
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Michal Rachel Suissa
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Niclas G Karlsson
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway.
| |
Collapse
|
12
|
Adams L, Li X, Burchmore R, Goodwin RJA, Wall DM. Microbiome-derived metabolite effects on intestinal barrier integrity and immune cell response to infection. MICROBIOLOGY (READING, ENGLAND) 2024; 170:001504. [PMID: 39392674 PMCID: PMC11469068 DOI: 10.1099/mic.0.001504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
The gut microbiota exerts a significant influence on human health and disease. While compositional changes in the gut microbiota in specific diseases can easily be determined, we lack a detailed mechanistic understanding of how these changes exert effects at the cellular level. However, the putative local and systemic effects on human physiology that are attributed to the gut microbiota are clearly being mediated through molecular communication. Here, we determined the effects of gut microbiome-derived metabolites l-tryptophan, butyrate, trimethylamine (TMA), 3-methyl-4-(trimethylammonio)butanoate (3,4-TMAB), 4-(trimethylammonio)pentanoate (4-TMAP), ursodeoxycholic acid (UDCA), glycocholic acid (GCA) and benzoate on the first line of defence in the gut. Using in vitro models of intestinal barrier integrity and studying the interaction of macrophages with pathogenic and non-pathogenic bacteria, we could ascertain the influence of these metabolites at the cellular level at physiologically relevant concentrations. Nearly all metabolites exerted positive effects on barrier function, but butyrate prevented a reduction in transepithelial resistance in the presence of the pathogen Escherichia coli, despite inducing increased apoptosis and exerting increased cytotoxicity. Induction of IL-8 was unaffected by all metabolites, but GCA stimulated increased intra-macrophage growth of E. coli and tumour necrosis-alpha (TNF-α) release. Butyrate, 3,4-TMAB and benzoate all increased TNF-α release independent of bacterial replication. These findings reiterate the complexity of understanding microbiome effects on host physiology and underline that microbiome metabolites are crucial mediators of barrier function and the innate response to infection. Understanding these metabolites at the cellular level will allow us to move towards a better mechanistic understanding of microbiome influence over host physiology, a crucial step in advancing microbiome research.
Collapse
Affiliation(s)
- Lauren Adams
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| | - Xiang Li
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| | - Richard Burchmore
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| | - Richard J. A. Goodwin
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Daniel M. Wall
- School of Infection and Immunology, College of Medical, Veterinary and Life Sciences, Sir Graeme Davies Building, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|
13
|
Garrido G, Garrido-Suárez BB, Martínez-Tapia N, Valdés-González M, Ortega-Mardones A. Antidiarrheal effect of Psidium guajava L. extract in acute diarrhea: a systematic review. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7731-7753. [PMID: 38578668 DOI: 10.1002/jsfa.13515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 01/21/2024] [Accepted: 04/03/2024] [Indexed: 04/06/2024]
Abstract
Acute diarrheal diseases are a leading cause of childhood mortality and morbidity worldwide. Psidium guajava has been globally used for its antidiarrheal potential. We conducted a systematic review of scientific articles published up to the year 2021, which included in vivo pre-clinical tests and clinical trials involving patients with acute infectious diarrhea to verify the antidiarrheal, antibacterial and antispasmodic effects of galenic preparations or phytopharmaceuticals from P. guajava. PRISMA and Rayyan were used as tools for the selection of studies collected in four databases (Pubmed, Scopus, Web of Science and Science Direct). The keywords used to carry out the search were: 'Psidium guajava', 'guava', 'antidiarrhe*' and 'diarrhe*', joined by Boolean operators 'OR' or 'AND'. The characteristics of studies in animal models of acute diarrhea induction, as well as in vivo and in vitro motility and microbiological tests linked with its main pathophysiological mechanisms, were collected. Twenty-three articles were included. Twenty (87%) of these reported heterogenic preclinical studies, predominating pharmacological studies of efficacy against conventional antidiarrheal agents, which utilized relevant outcomes and models of infectious diarrhea from the top pathogens in the clinic along with classical castor oil-induced diarrhea associated with motility tests. Only three articles (13%) corresponded to clinical trials investigating the efficacy, dose and safety of these preparations. Most studies reported positive results and significant mechanistic evidence from antibacterial, anti-motility, anti-secretory and protective/anti-inflammatory perspectives. However, further studies are needed to define the clinical significance and safety treatment with P. guajava extracts. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Gabino Garrido
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Universidad Católica del Norte, Antofagasta, Chile
| | | | - Nicolás Martínez-Tapia
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Universidad Católica del Norte, Antofagasta, Chile
| | - Marisela Valdés-González
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Universidad Católica del Norte, Antofagasta, Chile
| | - Andrea Ortega-Mardones
- Departamento Procesos de Diagnóstico y Evaluación, Facultad Ciencias de la Salud, Universidad Católica de Temuco, Temuco, Chile
| |
Collapse
|
14
|
Su J, Dai Y, Wu X, Zhou X, Fang X, Ge X, Zhao L. Maslinic acid alleviates alcoholic liver injury in mice and regulates intestinal microbiota via the gut-liver axis. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7928-7938. [PMID: 38837352 DOI: 10.1002/jsfa.13624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/01/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Maslinic acid (MA), a pentacyclic triterpene acid, is widely distributed in natural plants and mainly found in the fruit and leaves of olives and hawthorn. MA has been reported as having many health-promoting functions, such as anticancer, anti-inflammation and neuroprotective activities. According to previous study, hawthorn extract has certain hepatoprotective effects. However, the detailed mechanism is still unclear, especially the effect of MA on gut microbiota. RESULTS Our study reveals that MA effectively counteracts alcohol-induced liver injury and oxidative stress. It mitigates alcohol-induced intestinal barrier damage, reverses increased permeability and reduces translocation of lipopolysaccharide (LPS). This prevents LPS/Toll-like receptor 4 activation, leading to decreased TNF-α and IL-1β production. Furthermore, MA rebalances gut microbiota by reversing harmful bacterial abundance and enhancing beneficial bacteria post-alcohol consumption. CONCLUSION MA, through modulation of gut microbiota, alleviates alcohol-induced liver injury via the gut-liver axis. These findings support the potential use of MA as a functional food ingredient for preventing or treating alcoholic liver disease. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jingwen Su
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Yuan Dai
- Jiangsu Yanghe Distillery Co. Ltd, Suqian, China
| | - Xianyao Wu
- Jinling High School Hexi Campus International Department, Nanjing, China
| | - Xinhu Zhou
- Jiangsu Yanghe Distillery Co. Ltd, Suqian, China
| | - Xianying Fang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
- Jinpu Research institute, Nanjing Forestry University, Nanjing, China
| | - Xiangyang Ge
- Jiangsu Yanghe Distillery Co. Ltd, Suqian, China
| | - Linguo Zhao
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, Nanjing Forestry University, Nanjing, China
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
- Jinpu Research institute, Nanjing Forestry University, Nanjing, China
| |
Collapse
|
15
|
Lacy BE, Cangemi DJ. Opioids and the Gastrointestinal Tract: The Role of Peripherally Active µ-Opioid Receptor Antagonists in Modulating Intestinal Permeability. Am J Gastroenterol 2024; 119:1970-1978. [PMID: 38870087 PMCID: PMC11446513 DOI: 10.14309/ajg.0000000000002887] [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: 12/18/2023] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Opioid receptors are found throughout the gastrointestinal tract, including the large intestine. Many patients treated with opioids experience opioid-induced constipation (OIC). Laxatives are not effective in most patients, and in those who do initially respond, the efficacy of laxatives generally diminishes over time. In addition, OIC does not spontaneously resolve for most patients. However, complications of opioids extend far beyond simply slowing gastrointestinal transit. Opioid use can affect intestinal permeability through a variety of mechanisms. Toll-like receptors are a crucial component of innate immunity and are tightly regulated within the gut epithelium. Pathologic µ-opioid receptor (MOR) and toll-like receptor signaling, resulting from chronic opioid exposure, disrupts intestinal permeability leading to potentially harmful bacterial translocation, elevated levels of bacterial toxins, immune activation, and increased cytokine production. Peripherally active MOR antagonists, including methylnaltrexone, are effective at treating OIC. Benefits extend beyond simply blocking the MOR; these agents also act to ameliorate opioid-induced disrupted intestinal permeability. In this review, we briefly describe the physiology of the gastrointestinal epithelial border and discuss the impact of opioids on gastrointestinal function. Finally, we consider the use of peripherally active MOR antagonists to treat disrupted intestinal permeability resulting from opioid use and discuss the potential for improved morbidity and mortality in patients treated with methylnaltrexone for opioid-induced bowel disorders.
Collapse
|
16
|
Hirano A, Kadoya H, Takasu M, Iwakura T, Kajimoto E, Tatsugawa R, Matsuura T, Kurumatani H, Yamamoto T, Kidokoro K, Kishi S, Nagasu H, Sasaki T, Kashihara N. Effects of Beraprost on Intestinal Microcirculation and Barrier Function in a Mouse Model of Renal Failure. Microcirculation 2024:e12889. [PMID: 39348278 DOI: 10.1111/micc.12889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/07/2024] [Accepted: 09/10/2024] [Indexed: 10/02/2024]
Abstract
OBJECTIVE Endothelial dysfunction plays an important role in the pathogenesis of chronic kidney disease. Prostacyclin (PGI2), an endothelial cell-produced endogenous prostaglandin, plays a crucial role in maintaining endothelial function. However, its effects on intestinal microcirculation and barrier function are not fully understood. We hypothesized that PGI2 improves intestinal microcirculation and barrier function via endothelial protective effects. METHODS ICR and ICGN (a spontaneous nephrotic model) mice were used in this study. Intestinal microcirculation was visualized in vivo to investigate PGI2 effects. Beraprost served as PGI2. PGI2 administration spanned 4 weeks, following which we assessed its influence on intestinal endothelial, intestinal barrier, and renal functions. RESULTS We visualized intestinal microcirculation and endothelial glycocalyx in the intestinal blood vessels. Beraprost administration induced a 1.2-fold dilatation of the vascular diameter of the small intestine. Intestinal blood flow in ICGN mice was significantly reduced compared that in ICR mice but improved with beraprost administration. ICGN mice exhibited reduced serum albumin levels, decreased ambulation, an imbalance in intestinal reactive oxygen species (ROS)/nitric oxide (NO), and impaired tight junctions; all were ameliorated by beraprost administration. CONCLUSIONS Beraprost improves intestinal microcirculation and barrier function by ameliorating ROS/NO imbalances, thereby reducing physical inactivity during renal failure.
Collapse
Affiliation(s)
- Akira Hirano
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hiroyuki Kadoya
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
- Department of General Geriatric Medicine, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Masanobu Takasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Tsukasa Iwakura
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Eriko Kajimoto
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Rie Tatsugawa
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | | | | | - Toshiya Yamamoto
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
- Department of General Geriatric Medicine, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kengo Kidokoro
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Seiji Kishi
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Tamaki Sasaki
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
- Department of Medical Science, Kawasaki Medical School, Kurashiki, Okayama, Japan
| |
Collapse
|
17
|
Lombardo GE, Navarra M, Cremonini E. A flavonoid-rich extract of bergamot juice improves high-fat diet-induced intestinal permeability and associated hepatic damage in mice. Food Funct 2024; 15:9941-9953. [PMID: 39263833 DOI: 10.1039/d4fo02538e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Consumption of high-fat diets (HFDs) is a contributing factor to obesity, insulin resistance and non-alcoholic fatty liver disease (NAFLD). Several studies suggested the protective role of bioactives present in Citrus fruits against the above mentioned chronic metabolic conditions. In this study, we evaluated if a flavonoid-rich extract of Citrus bergamia (bergamot) juice (BJe) could inhibit HFD-induced intestinal permeability and endotoxemia and, through this mechanism, mitigate the associated hepatic damage in C57BL/6J mice. After 12 weeks of the treatment, HFD consumption caused high body weight (BW) gain, hyperinsulinemia, hyperglycemia, and dyslipidemia, which were mitigated by BJe (50 mg per kg BW) supplementation. Furthermore, supplementation with BJe prevented HFD-induced liver alterations, including increased plasma alanine aminotransferase (ALT) activity, increased hepatic lipid deposition, high NAS, and fibrosis. Mice fed a HFD for 12 weeks showed (i) a decrease in small intestine tight junction protein levels (ZO-1, occludin, and claudin-1), (ii) increased intestinal permeability, and (iii) endotoxemia. All these adverse events were mitigated by BJe supplementation. Linking the capacity of BJe to prevent HFD-associated endotoxemia, supplementation with this extract decreased the HFD-induced overexpression of hepatic TLR-4, downstream signaling pathways (MyD88, NF-κB and MAPK), and the associated inflammation, evidenced by increased MCP-1, TNF-α, IL-6, iNOS, and F4/80 levels. Overall, we suggest that BJe could mitigate the harmful consequences of western style diet consumption on liver physiology by protecting the gastrointestinal tract from permeabilization and associated metabolic endotoxemia.
Collapse
Affiliation(s)
- Giovanni E Lombardo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.
- Prof. Antonio Imbesi Foundation, Messina, Italy
- Department of Medicine and Surgery, "Kore" University of Enna, Enna, Italy
- Department of Nutrition, University of California, Davis, USA.
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy.
| | | |
Collapse
|
18
|
Zhu Y, He H, Sun W, Wu J, Xiao Y, Peng Y, Hu P, Jin M, Liu P, Zhang D, Xie T, Huang L, He W, Wei M, Wang L, Xu X, Tang Y. IgA nephropathy: gut microbiome regulates the production of hypoglycosilated IgA1 via the TLR4 signaling pathway. Nephrol Dial Transplant 2024; 39:1624-1641. [PMID: 38402460 PMCID: PMC11427068 DOI: 10.1093/ndt/gfae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Immunoglobulin A nephropathy (IgAN) is a major cause of primary glomerulonephritis characterized by mesangial deposits of galactose-deficient IgA1 (Gd-IgA1). Toll-like receptors (TLRs), particularly TLR4, are involved in the pathogenesis of IgAN. The role of gut microbiota on IgAN patients was recently investigated. However, whether gut microbial modifications of Gd-IgA1 through TLR4 play a role in IgAN remains unclear. METHODS We recruited subjects into four groups, including 48 patients with untreated IgAN, 22 treated IgAN patients (IgANIT), 22 primary membranous nephropathy and 31 healthy controls (HCs). Fecal samples were collected to analyze changes in gut microbiome. Gd-IgA1 levels, expression of TLR4, B-cell stimulators and intestinal barrier function were evaluated in all subjects. C57BL/6 mice were treated with a broad-spectrum antibiotic cocktail to deplete the gut microbiota and then gavaged with fecal microbiota transplanted from clinical subjects of every group. Gd-IgA1 and TLR4 pathway were detected in peripheral blood mononuclear cells (PBMCs) from IgAN and HCs co-incubated with lipopolysaccharide (LPS) and TLR4 inhibitor. RESULTS Compared with the other three groups, different compositions and decreased diversity demonstrated gut dysbiosis in the untreated IgAN group, especially the enrichment of Escherichia-Shigella. Elevated Gd-IgA1 levels were found in untreated IgAN patients and correlated with gut dysbiosis, TLR4, B-cell stimulators, indexes of intestinal barrier damage and proinflammatory cytokines. In vivo, mice colonized with gut microbiota from IgAN and IgANIT patients mimicked the IgAN phenotype with the activation of TLR4/MyD88/nuclear factor-κB pathway and B-cell stimulators in the intestine, and had with enhanced proinflammatory cytokines. In vitro, LPS activated TLR4/MyD88/NF-κB pathway, B-cell stimulators and proinflammatory cytokines in PBMCs of IgAN patients. This process may induce the overproduction of Gd-IgA1, which was inhibited by TLR4 inhibitors. CONCLUSIONS Our results illustrated that the gut-kidney axis is involved in the pathogenesis of IgAN. Gut dysbiosis could stimulate the overproduction of Gd-IgA1 via TLR4 signaling pathway production and B-cell stimulators.
Collapse
Affiliation(s)
- Yifan Zhu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Haidong He
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Weiqian Sun
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Jiajun Wu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Yong Xiao
- Department of Emergency, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Yinshun Peng
- School of Public Health, Fudan University, Shanghai, China
| | - Ping Hu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Meiping Jin
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Ping Liu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - DongLiang Zhang
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Ting Xie
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Lusheng Huang
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Weiming He
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Suzhou, P.R. China
| | - Minggang Wei
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Soochow University, Jiangsu Suzhou, P.R. China
| | - Lishun Wang
- Center for Traditional Chinese Medicine and Gut Microbiota, Institute of Fudan-Minhang Academic Health System, Minhang Hospital, Fudan University, Shanghai, China
| | - Xudong Xu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| | - Yuyan Tang
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
19
|
Miralles A, Ramis G, Pallarés FJ, Párraga-Ros E, Seva J. Medium- and Long-Term Immune Responses in the Small Intestine in Piglets from Oral Vaccination against Escherichia coli. Animals (Basel) 2024; 14:2779. [PMID: 39409727 PMCID: PMC11476012 DOI: 10.3390/ani14192779] [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: 08/28/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Post-weaning stress, together with Escherichia coli, are two of the key factors in the occurrence of post-weaning diarrhea. There are different commercial vaccines that induce immunity at the local or systemic level, improving farm health and avoiding economic losses in the pork industry. That is why the objective of this study was to evaluate the effect of an oral enterotoxigenic E. coli F4/F18 vaccine on immunity and intestinal integrity in the middle and long term after inoculation. The gene expression of the biomarkers indicative of cellular infiltration (calprotectin, CAL), tight junction proteins (occludin, OCL; zonulin, ZON; and claudin, CLA) and a panel of proinflammatory (interleukins, IL: IL1α, IL1β, IL6, IL8, IL12p35 and IL12p40; interferons, IFN: IFNα and IFNγ; and tumoral necrosis factor, TNF: TNFα) and anti-inflammatory mediator cytokines (TGFβ and IL10) were analyzed, as well as histomorphology in jejunum and ileum, the cell density of goblet cells, intraepithelial lymphocytes and IgA-producing cells. Differences were observed in ZON, CLA and CAL, with greater gene expression in observed in vaccinated piglets at 42 days post vaccination (dpv) in the ileum. Regarding the expression of cytokines, the vaccinated animals showed significant differences in IL1α, IL6, IL12p35, IL12p40, IFNα, IFNγ, TNFα and TGFβ at 42 dpv in the jejunum or ileum. The villi showed greater height in the vaccinated piglets and the ratio between villus height and crypt depth was significantly greater in the vaccinated group in the jejunum at 84 dpv. The count of IgA-producing cells shows higher values for the unvaccinated group in the ileum, while intraepithelial lymphocytes show a significant increase in both jejunum and ileum in vaccinated piglets. We can conclude that oral vaccination against E. coli produces an evident effect, which manifests itself even in the middle and long term after the challenge, including immune response, decrease in antimicrobials usage, better histological structure in intestine and the improvement of performance.
Collapse
Affiliation(s)
- Aida Miralles
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (A.M.); (E.P.-R.); (J.S.)
- CEFU, S.A., 30840 Alhama de Murcia, Spain
| | - Guillermo Ramis
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), 30120 Murcia, Spain
| | - Francisco J. Pallarés
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Universidad de Córdoba, 14071 Córdoba, Spain;
| | - Ester Párraga-Ros
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (A.M.); (E.P.-R.); (J.S.)
| | - Juan Seva
- Departamento de Anatomía y Anatomía Patológica Comparadas, Facultad de Veterinaria, Universidad de Murcia, 30100 Murcia, Spain; (A.M.); (E.P.-R.); (J.S.)
| |
Collapse
|
20
|
Brown K, Funk K, Figueroa Barrientos A, Bailey A, Shrader S, Feng W, McClain CJ, Song ZH. The Modulatory Effects and Therapeutic Potential of Cannabidiol in the Gut. Cells 2024; 13:1618. [PMID: 39404382 PMCID: PMC11475737 DOI: 10.3390/cells13191618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Cannabidiol (CBD) is a major non-psychotropic phytocannabinoid that exists in the Cannabis sativa plant. CBD has been found to act on various receptors, including both cannabinoid and non-cannabinoid receptors. In addition, CBD has antioxidant effects that are independent of receptors. CBD has demonstrated modulatory effects at different organ systems, such as the central nervous system, immune system, and the gastrointestinal system. Due to its broad effects within the body and its safety profile, CBD has become a topic of therapeutic interest. This literature review summarizes previous research findings with regard to the effect of CBD on the gastrointestinal (GI) system, including its effects at the molecular, cellular, organ, and whole-body levels. Both pre-clinical animal studies and human clinical trials are reviewed. The results of the studies included in this literature review suggest that CBD has significant impact on intestinal permeability, the microbiome, immune cells and cytokines. As a result, CBD has been shown to have therapeutic potential for GI disorders such as inflammatory bowel disease (IBD). Furthermore, through interactions with the gut, CBD may also be helpful in the treatment of disorders outside the GI system, such as non-alcoholic liver disease, postmenopausal disorders, epilepsy, and multiple sclerosis. In the future, more mechanistic studies are warranted to elucidate the detailed mechanisms of action of CBD in the gut. In addition, more well-designed clinical trials are needed to explore the full therapeutic potential of CBD on and through the gut.
Collapse
Affiliation(s)
- Kevin Brown
- College of Medicine and Life Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Kyle Funk
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Alexa Figueroa Barrientos
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Ashly Bailey
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Sarah Shrader
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Wenke Feng
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Craig J. McClain
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Zhao-Hui Song
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40292, USA
| |
Collapse
|
21
|
Schaller ML, Sykes MM, Mecano J, Solanki S, Huang W, Rebernick RJ, Beydoun S, Wang E, Bugarin-Lapuz A, Shah YM, Leiser SF. Fmo5 plays a sex-specific role in goblet cell maturation and mucus barrier formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588360. [PMID: 38645243 PMCID: PMC11030302 DOI: 10.1101/2024.04.05.588360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background and Aims The intestine plays a key role in metabolism, nutrient and water absorption, and provides both physical and immunological defense against dietary and luminal antigens. The protective mucosal lining in the intestine is a critical component of intestinal barrier that when compromised, can lead to increased permeability, a defining characteristic of inflammatory bowel disease (IBD), among other intestinal diseases. Here, we define a new role for the flavin-containing monooxygenase (FMO) family of enzymes in maintaining a healthy intestinal epithelium. Methods Using Caenorhabditis elegans we measure intestinal barrier function, actin expression, and intestinal damage response. In mice, we utilize an intestine-specific, tamoxifen- inducible knockout model of the mammalian homolog of Cefmo-2 , Fmo5, and assess histology, mucus barrier thickness, and goblet cell physiology. We also treat mice with the ER chaperone Tauroursodeoxycholic acid (TUDCA). Results In nematodes, we find Cefmo-2 is necessary and sufficient for intestinal barrier function, intestinal actin expression, and is induced by intestinal damage. In mice, we find striking changes to the intestine within two weeks following Fmo5 disruption. Alterations include sex-dependent changes in colon epithelial histology, goblet cell localization, and mucus barrier formation. These changes are significantly more severe in female mice, mirroring differences observed in IBD patients. Furthermore, we find increased protein folding stress in Fmo5 knockout animals and successfully rescue the severe female phenotype with addition of a chemical ER chaperone. Conclusions Together, our results identify a highly conserved and novel role for Fmo5 in the mammalian intestine and support a key role for Fmo5 in maintenance of ER/protein homeostasis and proper mucus barrier formation.
Collapse
|
22
|
Bakinowska E, Stańska W, Kiełbowski K, Szwedkowicz A, Boboryko D, Pawlik A. Gut Dysbiosis and Dietary Interventions in Rheumatoid Arthritis-A Narrative Review. Nutrients 2024; 16:3215. [PMID: 39339815 PMCID: PMC11435214 DOI: 10.3390/nu16183215] [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: 08/11/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive autoimmune disease. The pathogenesis of RA is complex and involves interactions between articular cells, such as fibroblast-like synoviocytes, and immune cells. These cells secrete pro-inflammatory cytokines, chemokines, metalloproteinases and other molecules that together participate in joint degradation. The current evidence suggests the important immunoregulatory role of the gut microbiome, which can affect susceptibility to diseases and infections. An altered microbiome, a phenomenon known as gut dysbiosis, is associated with the development of inflammatory diseases. Importantly, the profile of the gut microbiome depends on dietary habits. Therefore, dietary elements and interventions can indirectly impact the progression of diseases. This review summarises the evidence on the involvement of gut dysbiosis and diet in the pathogenesis of RA.
Collapse
Affiliation(s)
- Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Agata Szwedkowicz
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Dominika Boboryko
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
23
|
Yan X, Bai X, Fu R, Duan Z, Zeng W, Zhu C. Ginsenoside compound K alleviates D-galactose-induced mild cognitive impairment by modulating gut microbiota-mediated short-chain fatty acid metabolism. Food Funct 2024; 15:9037-9052. [PMID: 39150321 DOI: 10.1039/d4fo03216k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The occurrence and progression of mild cognitive impairment (MCI) are closely related to dysbiosis of the gut microbiota. Ginsenoside compound K (CK), a bioactive component of ginseng, has been shown to alleviate gut microbiota dysbiosis and neural damage. However, the mechanisms by which CK regulates the gut microbiota to improve MCI remain unexplored. In this study, an MCI mouse model induced by D-galactose was used, and 16S rRNA gene sequencing, metabolomics, transcriptomics, and integrative multi-omics analyses were employed to investigate the potential mechanisms by which CK alleviates MCI through modulation of the gut microbiota. The results demonstrated that CK repaired intestinal barrier dysfunction caused by MCI, improved blood-brain barrier (BBB) integrity, inhibited activation of microglial cells and astrocytes, and significantly ameliorated MCI. Furthermore, CK enhanced gut microbiota diversity, notably enriched beneficial bacteria such as Akkermansia, and modulated the levels of short-chain fatty acids (SCFAs), particularly increasing propionate, thereby alleviating gut microbiota dysbiosis caused by MCI. Germ-free experiments confirmed that gut microbiota is a key factor for ginsenoside CK in relieving MCI. Further investigation revealed that CK regulated the TLR4-MyD88-NF-κB signaling pathway through modulation of gut microbiota-mediated propionate metabolism, significantly reducing systemic inflammation and alleviating MCI. Our findings provide a new theoretical basis for using CK as a potential means of modulating the gut microbiota for the treatment of MCI.
Collapse
Affiliation(s)
- Xiaojun Yan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China.
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Xue Bai
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China.
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China.
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China.
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Wen Zeng
- Xi'an Honghui Hospital, 710054, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China.
- Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| |
Collapse
|
24
|
Dora D, Szőcs E, Soós Á, Halasy V, Somodi C, Mihucz A, Rostás M, Mógor F, Lohinai Z, Nagy N. From bench to bedside: an interdisciplinary journey through the gut-lung axis with insights into lung cancer and immunotherapy. Front Immunol 2024; 15:1434804. [PMID: 39301033 PMCID: PMC11410641 DOI: 10.3389/fimmu.2024.1434804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
This comprehensive review undertakes a multidisciplinary exploration of the gut-lung axis, from the foundational aspects of anatomy, embryology, and histology, through the functional dynamics of pathophysiology, to implications for clinical science. The gut-lung axis, a bidirectional communication pathway, is central to understanding the interconnectedness of the gastrointestinal- and respiratory systems, both of which share embryological origins and engage in a continuous immunological crosstalk to maintain homeostasis and defend against external noxa. An essential component of this axis is the mucosa-associated lymphoid tissue system (MALT), which orchestrates immune responses across these distant sites. The review delves into the role of the gut microbiome in modulating these interactions, highlighting how microbial dysbiosis and increased gut permeability ("leaky gut") can precipitate systemic inflammation and exacerbate respiratory conditions. Moreover, we thoroughly present the implication of the axis in oncological practice, particularly in lung cancer development and response to cancer immunotherapies. Our work seeks not only to synthesize current knowledge across the spectrum of science related to the gut-lung axis but also to inspire future interdisciplinary research that bridges gaps between basic science and clinical application. Our ultimate goal was to underscore the importance of a holistic understanding of the gut-lung axis, advocating for an integrated approach to unravel its complexities in human health and disease.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Emőke Szőcs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Ádám Soós
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Viktória Halasy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Csenge Somodi
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Melinda Rostás
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, Hungary
| | - Fruzsina Mógor
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
25
|
Al-Failakawi A, Al-Jarallah A, Rao M, Khan I. The Role of Claudins in the Pathogenesis of Dextran Sulfate Sodium-Induced Experimental Colitis: The Effects of Nobiletin. Biomolecules 2024; 14:1122. [PMID: 39334888 PMCID: PMC11430412 DOI: 10.3390/biom14091122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The pathogenesis of inflammatory bowel diseases such as ulcerative colitis and Crohn's disease is not well understood. This study investigated the roles and regulation of the claudin-1, -2, -3, and -4 isoforms in the pathogenesis of ulcerative colitis, and the potential therapeutic effects of nobiletin. METHODS Colitis was induced in rats by administering dextran sulfate sodium [DSS] in drinking water for seven days. Animals were treated daily with nobiletin [oral, 60 mg/Kg body weight] and studied in four groups, C [non-colitis control], D [DSS-induced colitis], CN [nobiletin-treated non-colitis control], and DN [nobiletin-treated DSS-induced colitis]. On day seven, the animals were sacrificed, and colonic tissues were collected and analyzed. RESULTS Both macroscopic and microscopic findings suggest the progression of colitis. In the inflamed colon, claudin-1 and -4 proteins were decreased, claudin-2 increased, while the claudin-3 protein remained unchanged. Except for claudin-1, these changes were not paralleled by mRNA expression, indicating a complex regulatory mechanism. Uniform β-actin expression along with consistent quality and yield of total RNA indicated selectivity of these changes. Nobiletin treatment reversed these changes. CONCLUSIONS Altered expression of the claudin isoforms -1, -2, and -4 disrupts tight junctions, exposing the lamina propria to microflora, leading to electrolyte disturbance and the development of ulcerative colitis. Nobiletin with its anti-inflammatory properties may be useful in IBD.
Collapse
Affiliation(s)
- Asmaa Al-Failakawi
- Department of Biochemistry, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait; (A.A.-F.); (A.A.-J.)
| | - Aishah Al-Jarallah
- Department of Biochemistry, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait; (A.A.-F.); (A.A.-J.)
| | - Muddanna Rao
- Departments of Anatomy, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait;
| | - Islam Khan
- Department of Biochemistry, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait; (A.A.-F.); (A.A.-J.)
| |
Collapse
|
26
|
Snelson M, Vanuytsel T, Marques FZ. Breaking the Barrier: The Role of Gut Epithelial Permeability in the Pathogenesis of Hypertension. Curr Hypertens Rep 2024; 26:369-380. [PMID: 38662328 PMCID: PMC11324679 DOI: 10.1007/s11906-024-01307-2] [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] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE OF THE REVIEW To review what intestinal permeability is and how it is measured, and to summarise the current evidence linking altered intestinal permeability with the development of hypertension. RECENT FINDINGS Increased gastrointestinal permeability, directly measured in vivo, has been demonstrated in experimental and genetic animal models of hypertension. This is consistent with the passage of microbial substances to the systemic circulation and the activation of inflammatory pathways. Evidence for increased gut permeability in human hypertension has been reliant of a handful of blood biomarkers, with no studies directly measuring gut permeability in hypertensive cohorts. There is emerging literature that some of these putative biomarkers may not accurately reflect permeability of the gastrointestinal tract. Data from animal models of hypertension support they have increased gut permeability; however, there is a dearth of conclusive evidence in humans. Future studies are needed that directly measure intestinal permeability in people with hypertension.
Collapse
Affiliation(s)
- Matthew Snelson
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Australia
- Victorian Heart Institute, Monash University, Melbourne, Australia
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Francine Z Marques
- Hypertension Research Laboratory, School of Biological Sciences, Monash University, Melbourne, Australia.
- Victorian Heart Institute, Monash University, Melbourne, Australia.
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
27
|
Xu X, Yuan J, Zhu M, Gao J, Meng X, Wu Y, Li X, Tong P, Chen H. The potential of orally exposed risk factors and constituents aggravating food allergy: Possible mechanism and target cells. Compr Rev Food Sci Food Saf 2024; 23:e70014. [PMID: 39230383 DOI: 10.1111/1541-4337.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/10/2024] [Accepted: 08/18/2024] [Indexed: 09/05/2024]
Abstract
Food allergy is a significant concern for the health of humans worldwide. In addition to dietary exposure of food allergens, genetic and environmental factors also play an important role in the development of food allergy. However, only the tip of the iceberg of risk factors in food allergy has been identified. The importance of food allergy caused by orally exposed risk factors and constituents, including veterinary drugs, pesticides, processed foods/derivatives, nanoparticles, microplastics, pathogens, toxins, food additives, dietary intake of salt/sugar/total fat, vitamin D, and therapeutic drugs, are highlighted and discussed in this review. Moreover, the epithelial barrier hypothesis, which is closely associated with the occurrence of food allergy, is also introduced. Additionally, several orally exposed risk factors and constituents that have been reported to disrupt the epithelial barrier are elucidated. Finally, the possible mechanisms and key immune cells of orally exposed risk factors and constituents in aggravating food allergy are overviewed. Further work should be conducted to define the specific mechanism by which these risk factors and constituents are driving food allergy, which will be of central importance to the targeted therapy of food allergy.
Collapse
Affiliation(s)
- Xiaoqian Xu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Jin Yuan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Mengting Zhu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Xuanyi Meng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Yong Wu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- College of Food Science & Technology, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Ping Tong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, P. R. China
- Sino-German Joint Research Institute, Nanchang University, Nanchang, P. R. China
- Jiangxi Province Key Laboratory of Food Allergy, Nanchang, P. R. China
| |
Collapse
|
28
|
Yoshida M, Kanda N, Kashiwagi S, Wakimoto Y, Ohbe H, Nakamura K. Relationship between very early enteral nutrition and persistent inflammation, immunosuppression, and catabolism syndrome in cardiovascular surgery patients: a propensity score-matched study. Am J Clin Nutr 2024; 120:610-618. [PMID: 39038737 DOI: 10.1016/j.ajcnut.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/11/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Early enteral nutrition (EN) is recommended for patients with critical illness to maintain intestinal immunity. However, the optimal timing of the commencement of EN remains unclear, particularly after cardiovascular surgery. OBJECTIVES We herein focused on Persistent Inflammation, Immunosuppression, and Catabolism Syndrome (PICS) as a predisposing immunodeficiency and investigated its association with very early EN (VEEN) (<24 h) in patients who underwent cardiovascular surgery. METHODS In this retrospective study, we used an administrative claims database with laboratory examinations between 2008 and 2021 to identify adult patients admitted to the intensive care unit after cardiovascular surgery. Patients who received EN the day after surgery were assigned to the EN <24 h group, whereas those who received EN on day 2 or day 3 were assigned to the control group. The primary outcome was a composite of the incidence of PICS and mortality on day 14 after surgery. We defined PICS as patients who were hospitalized for >14 day and meeting ≥2 of the following conditions: a lymphocyte count <800/μL, albumin <3.0 g/dL, and C-reactive protein >2.0 mg/dL. We compared the 2 groups using propensity score analysis. RESULTS A propensity score matching generated 2082 pairs. The primary outcome was significantly lower in the EN <24 h group than in the control group on days 14 {risk difference [95% confidence interval (CI)]: -3.1% [-5.9%, -0.3%]} and 28 (risk difference [95% CI]: -2.1% [-3.7%, -0.4%]). Mortality did not significantly differ between the 2 groups. The length of hospital stay was significantly shorter in the EN <24 h group: the difference (95% CI) was -2.2 (-3.7, -0.7) d. CONCLUSIONS Among patients who underwent cardiovascular surgery, VEEN provided on the day after surgery was associated with a lower incidence of PICS and a shorter length of hospital stay than EN provided 2 day or 3 day after surgery.
Collapse
Affiliation(s)
- Minoru Yoshida
- Department of Emergency and Critical Care Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naoki Kanda
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Hitachi, Japan; Division of General Internal Medicine, Jichi Medical University, Tochigi, Japan
| | - Shizuka Kashiwagi
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuji Wakimoto
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Hitachi, Japan
| | - Hiroyuki Ohbe
- Division of Emergency and Critical Care Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kensuke Nakamura
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, Hitachi, Japan; Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| |
Collapse
|
29
|
Qian G, Zang H, Tang J, Zhang H, Yu J, Jia H, Zhang X, Zhou J. Lactobacillus gasseri ATCC33323 affects the intestinal mucosal barrier to ameliorate DSS-induced colitis through the NR1I3-mediated regulation of E-cadherin. PLoS Pathog 2024; 20:e1012541. [PMID: 39250508 PMCID: PMC11412683 DOI: 10.1371/journal.ppat.1012541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/19/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Inflammatory bowel disease (IBD) is an immune system disorder primarily characterized by colitis, the exact etiology of which remains unclear. Traditional treatment approaches currently yield limited efficacy and are associated with significant side effects. Extensive research has indicated the potent therapeutic effects of probiotics, particularly Lactobacillus strains, in managing colitis. However, the mechanisms through which Lactobacillus strains ameliorate colitis require further exploration. In our study, we selected Lactobacillus gasseri ATCC33323 from the intestinal microbiota to elucidate the specific mechanisms involved in modulation of colitis. Experimental findings in a DSS-induced colitis mouse model revealed that L. gasseri ATCC33323 significantly improved physiological damage in colitic mice, reduced the severity of colonic inflammation, decreased the production of inflammatory factors, and preserved the integrity of the intestinal epithelial structure and function. It also maintained the expression and localization of adhesive proteins while improving intestinal barrier permeability and restoring dysbiosis in the gut microbiota. E-cadherin, a critical adhesive protein, plays a pivotal role in this protective mechanism. Knocking down E-cadherin expression within the mouse intestinal tract significantly attenuated the ability of L. gasseri ATCC33323 to regulate colitis, thus confirming its protective role through E-cadherin. Finally, transcriptional analysis and in vitro experiments revealed that L. gasseri ATCC33323 regulates CDH1 transcription by affecting NR1I3, thereby promoting E-cadherin expression. These findings contribute to a better understanding of the specific mechanisms by which Lactobacillus strains alleviate colitis, offering new insights for the potential use of L. gasseri as an alternative therapy for IBD, particularly in dietary supplementation.
Collapse
Affiliation(s)
- Guanru Qian
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Hui Zang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jingtong Tang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Hao Zhang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jiankang Yu
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Huibiao Jia
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Xinzhuang Zhang
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| | - Jianping Zhou
- Department of Gastrointestinal Surgery & Hernia and Abdominal Wall Surgery, the First Hospital, China Medical University, Shenyang, China
- Department of Shenyang Medical Nutrition Clinical Medical Research Center, Shenyang, China
| |
Collapse
|
30
|
Wang Y, Wang Q, Wang G, Zhang Q, Guo Y, Su X, Tang Y, Koci M, Zhang J, Ma Q, Zhao L. Rutin, a natural flavonoid glycoside, ameliorates zearalenone induced liver inflammation via inhibiting lipopolysaccharide gut leakage and NF-κB signaling pathway in mice. Food Chem Toxicol 2024; 191:114887. [PMID: 39053873 DOI: 10.1016/j.fct.2024.114887] [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: 04/08/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Zearalenone (ZEN) poses a potential threat on human and animal health partly through the nuclear factor (NF)-κB signaling pathway. In silico study suggested that rutin effective against TLR4 and NF-κB. A wetting test was designed to evaluate the effect and underlying mechanism of rutin in alleviating ZEN-induced inflammation in animals. Twenty-four female mice were randomly divided into 4 groups: control (basal diet), ZEN group (basal diet + ZEN), rutin group (basic diet + rutin), Z + R group (basal diet + rutin + ZEN). Results showed that rutin effectively alleviated ZEN-induced inflammation and damage of liver and jejunum in mice. Rutin addition reduced the content of lipopolysaccharide (LPS) in serum and liver mainly by improving the intestinal barrier function resulted from the production increase of short-chain fatty acids (SCFA). In sum, this study showed that rutin alleviated ZEN-induced liver inflammation and injury by modulating the gut microbiota, increasing the production of SCFA and improving intestinal barrier function, leading to the decrease of LPS in liver and the inhibition of MyD88 independent NF-κB signaling pathway in mice. Specifically, these findings may provide useful insights into the screening of functional natural compounds and its action mechanism to alleviate ZEN induced liver inflammation.
Collapse
Affiliation(s)
- Yanan Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Qianqian Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Gaigai Wang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Qiongqiong Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yongpeng Guo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Xin Su
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Yu Tang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Matthew Koci
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, 27695, USA.
| | - Jianyun Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Qiugang Ma
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Lihong Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Poultry Nutrition and Feed Technology Innovation Team, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
31
|
Bertens CA, Seymour DJ, Penner GB. Validation of an in vivo dual permeability marker technique to characterize regional gastrointestinal tract permeability in mid lactation Holstein cows during short-term feed restriction. J Dairy Sci 2024:S0022-0302(24)01103-2. [PMID: 39218063 DOI: 10.3168/jds.2024-25142] [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: 05/10/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
This study evaluated the impact of short-term feed restriction in lactating dairy cows on regional permeability of the gastrointestinal tract (GIT), and the recovery of DMI, ruminal pH, and milk yield. In addition, sampling methods for a novel dual marker technique to characterize total GIT and post ruminal permeability were validated. Six ruminally cannulated lactating Holstein cows were blocked by parity (3 primiparous, 3 multiparous; 189 DIM ± 25.2) and enrolled in a crossover design. Experimental periods included a 5-d baseline phase (BASE), 5-d challenge phase (CHAL), and 2 weeks of recovery (REC1 and REC2). During CHAL cows received either 100% ad libitum feed intake (AL) or 40% of ad libitum feed intake (FR). To assess, total-tract and post-ruminal permeability, equimolar doses of Cr-EDTA and Co-EDTA were infused on d 3 of CHAL into the rumen and abomasum (0.369 mmol/kg BW). Following infusions, total urine and feces were collected every 8 h over 96 h, and blood samples were collected at h 0, 1, 2, 3, 4, 6, 8, 12, 16, 20, 24, 32, 40, 48, and 64. The plasma area under the curve (AUC) for Cr and Co were calculated. By design, DMI for FR was reduced by 60% during CHAL and remained 19% lower than AL during REC1 but was not different from AL in REC2. Mean ruminal pH for FR was greatest during CHAL and the least during REC1, with no differences detected between AL and FR in REC2. The duration that pH was < 5.8 was the least for FR during CHAL and greatest during REC1 which were different from AL and were no longer different between treatments in REC2. Milk yield was the least for FR during CHAL and REC1 and no longer different from AL in REC2. Feed restriction reduced milk fat, protein, and lactose yields by 26, 31% and 31%, respectively. Plasma Cr AUC was 34% greater and Co AUC tended to be 35% greater for FR than AL on d 3 of CHAL. Urinary Cr recovery after 48-h was not affected by treatment; however, urinary Co recovery was 36% greater for FR than AL. Positive correlations between plasma AUC and urinary recovery for Cr and Co were detected. It was determined that blood samples collected at h 2, 8, 20, 40, and 48 could predict the total plasma Cr and Co AUC within 1.9% and 6.2%, respectively. In summary, short-term FR in lactating dairy cows increases permeability of the total GIT and may increase permeability of the post-ruminal regions with more than 60% of the permeability occurring post-ruminally. After FR, cows experienced low ruminal pH and a sustained reduction in milk yield. When utilizing Cr- and Co-EDTA to evaluate regional GIT permeability, plasma AUC can be used as an alternate to urinary Cr and Co excretion. In addition, blood samples collected at h 2, 8, 20, 40, and 48 result in adequate prediction accuracy, at least when comparing GIT permeability for lactating dairy cows exposed to AL and FR.
Collapse
Affiliation(s)
- C A Bertens
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada S7N 5A8
| | - D J Seymour
- Trouw Nutrition R&D, P.O. Box 200, 5830 AE Boxmeer, the Netherlands; Centre for Nutrition Modelling, Department of Animal Biosciences, University of Guelph, ON, Canada N1G 2W1
| | - G B Penner
- Department of Animal and Poultry Science, University of Saskatchewan, Saskatoon, SK, Canada S7N 5A8.
| |
Collapse
|
32
|
Keivanlou MH, Amini-Salehi E, Sattari N, Hashemi M, Saberian P, Prabhu SV, Javid M, Mirdamadi A, Heidarzad F, Bakhshi A, Letafatkar N, Zare R, Hassanipour S, Nayak SS. Gut microbiota interventions in type 2 diabetes mellitus: An umbrella review of glycemic indices. Diabetes Metab Syndr 2024; 18:103110. [PMID: 39213690 DOI: 10.1016/j.dsx.2024.103110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/18/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND We aimed to explore how probiotics, prebiotics, or synbiotics impact glycemic indices in patients with diabetes mellitus. METHOD A comprehensive search was conducted on PubMed, Scopus, and Web of Science from inception up to April 2023. The random-effects model was employed for the study analysis. Furthermore, sensitivity and subgroup analyses were conducted to investigate potential sources of heterogeneity. AMSTAR2 checklist was used to determine the quality of studies. Comprehensive meta-analysis version 3 was used for the study analysis. RESULT A total of 31 studies were included in the final analysis. Based on the results of the meta-analysis, gut microbial therapy could significantly decrease serum fasting blood glucose levels in patients with type 2 diabetes mellitus (effect size: -0.211; 95 % CI: -0.257, -0.164; P < 0.001). Additionally, significant associations were also found between gut microbial therapy and improved serum levels of fasting insulin, glycated hemoglobin, and homeostatic model assessment for insulin resistance (effect size: -0.087; 95 % confidence interval: -0.120, -0.053; P < 0.001; effect size: -0.166; 95 % confidence interval: -0.200, -0.132; P < 0.001; effect size: -0.230; 95 % confidence interval: -0.288, -0.172; P < 0.001, respectively). CONCLUSION Our results revealed promising effects of gut microbiota modulation on glycemic profile of patients with type 2 diabetes mellitus. The use of these agents as additional treatments can be considered.
Collapse
Affiliation(s)
- Mohammad-Hossein Keivanlou
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hashemi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Parsa Saberian
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Mona Javid
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Arian Mirdamadi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Forough Heidarzad
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Bakhshi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Negin Letafatkar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Zare
- Student Research Committee, Larestan University of Medical Sciences, Larestan, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sandeep Samethadka Nayak
- Department of Internal Medicine, Yale New Haven Health Bridgeport Hospital , Bridgeport, CT, USA
| |
Collapse
|
33
|
Herrera-Quintana L, Vázquez-Lorente H, Hinojosa-Nogueira D, Plaza-Diaz J. Relationship between Infant Feeding and the Microbiome: Implications for Allergies and Food Intolerances. CHILDREN (BASEL, SWITZERLAND) 2024; 11:1030. [PMID: 39201963 PMCID: PMC11353207 DOI: 10.3390/children11081030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/03/2024]
Abstract
Childhood is a critical period for immune system development, which is greatly influenced by the gut microbiome. Likewise, a number of factors affect the gut microbiome composition and diversity, including breastfeeding, formula feeding, and solid foods introduction. In this regard, several studies have previously demonstrated that breastfeeding promotes a favorable microbiome. In contrast, formula feeding and the early incorporation of certain solid foods may adversely affect microbiome development. Additionally, there is increasing evidence that disruptions in the early microbiome can lead to allergic conditions and food intolerances. Thus, developing strategies to promote optimal infant nutrition requires an understanding of the relationship between infant nutrition and long-term health. The present review aims to examine the relationship between infant feeding practices and the microbiome, as well as its implications on allergies and food intolerances in infants. Moreover, this study synthesizes existing evidence on how different eating habits influence the microbiome. It highlights their implications for the prevention of allergies and food intolerances. In conclusion, introducing allergenic solid foods before six months, alongside breastfeeding, may significantly reduce allergies and food intolerances risks, being also associated with variations in gut microbiome and related complications.
Collapse
Affiliation(s)
- Lourdes Herrera-Quintana
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Héctor Vázquez-Lorente
- Department of Physiology, Schools of Pharmacy and Medicine, University of Granada, 18071 Granada, Spain; (L.H.-Q.); (H.V.-L.)
- Biomedical Research Center, Health Sciences Technology Park, University of Granada, 18016 Granada, Spain
| | - Daniel Hinojosa-Nogueira
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Laboratorio del Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario de Málaga (Virgen de la Victoria), 29590 Málaga, Spain;
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Campus de Cartuja s/n, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS, GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
34
|
Yan S, Liu T, Zhao H, Zhao C, Zhu Y, Dai W, Sun W, Wang H, Sun J, Zhao L, Xu D. Colorectal cancer-specific microbiome in peripheral circulation and cancer tissues. Front Microbiol 2024; 15:1422536. [PMID: 39234556 PMCID: PMC11371800 DOI: 10.3389/fmicb.2024.1422536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/02/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction Accumulating evidence has supported that gut microbiota and metabolite profiles play indispensable roles in the pathogenesis of colorectal cancer (CRC), which ranks as the third most common cancer and the second leading cause of cancer-related deaths worldwide. However, alterations in tumoral or circulating microbiomes in CRC remain incompletely understood. It has been well-documented that tissue or serum microbiomes with low microbial biomass could be screened by use of 2bRAD sequencing for microbiome (2bRAD-M) at the species resolution. Methods In order to validate the microbial biomarkers distinguishing CRC and the variations in microorganisms present in serum and tumors, we performed 2bRAD-M to characterize the microbiomes in serum and cancer tissues of CRC patients with and without lymph node or liver metastasis. Results The composition of dominated microbiota in serum was different from that of tissue samples, while the microbial community composition of tumors was similar to that of the tumor-adjacent tissues. The analysis of α-diversity and β-diversity has revealed notable variations in serum microbiota diversities in CRC patients, particularly those with liver metastasis. Multiple CRC-specific microbial species, such as Moraxella A cinereus, Flavobacterium sp001800905, and Acinetobacter albensis, were identified in serum. Complicated functions and KEGG pathways were also confirmed in CRC according to the metastasis status. Discussion This study has found significant alterations in the microbial compositions and diversities in CRC and CRC-specific microbial species in both circulation and cancer tissues, which may serve as promising biomarkers for the screening, diagnosis and prognosis prediction of CRC. In particular, CRC-specific bacterial taxa are promising markers, holding transformative potentials in establishing personalized screening and risk stratification, refining much earlier non-invasive diagnostic approaches, and enhancing diagnostic sensitivity.
Collapse
Affiliation(s)
- Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery, Affiliated Hospital of Shandong Second Medical University, Weifang, China
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Tie Liu
- Department of Anorectal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Haobin Zhao
- Central Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Chunbo Zhao
- Department of Anorectal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Yuxin Zhu
- Central Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Wenqing Dai
- Central Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Wenchang Sun
- Central Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Honggang Wang
- Clinical Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Junxi Sun
- Department of Anorectal Surgery, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| | - Lu Zhao
- Central Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Laibo Biotechnology Co., Ltd., Jinan, China
| | - Donghua Xu
- Central Laboratory, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
- Department of Rheumatology and Immunology, Weifang People's Hospital, Shandong Second Medical University, Weifang, China
| |
Collapse
|
35
|
Li Z, Wan M, Wang M, Duan J, Jiang S. Modulation of gut microbiota on intestinal permeability: A novel strategy for treating gastrointestinal related diseases. Int Immunopharmacol 2024; 137:112416. [PMID: 38852521 DOI: 10.1016/j.intimp.2024.112416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/07/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Accumulating evidence emphasizes the critical reciprocity between gut microbiota and intestinal barrier function in maintaining the gastrointestinal homeostasis. Given the fundamental role caused by intestinal permeability, which has been scrutinized as a measurable potential indicator of perturbed barrier function in clinical researches, it seems not surprising that recent decades have been marked by augmented efforts to determine the interaction between intestinal microbes and permeability of the individual. However, despite the significant progress in characterizing intestinal permeability and the commensal bacteria in the intestine, the mechanisms involved are still far from being thoroughly revealed. In the present review, based on multiomic methods, high-throughput sequencing and molecular biology techniques, the impacts of gut microbiota on intestinal permeability as well as their complex interaction networks are systematically summarized. Furthermore, the diseases related to intestinal permeability and main causes of changes in intestinal permeability are briefly introduced. The purpose of this review is to provide a novel prospection to elucidate the correlation between intestinal microbiota and permeability, and to explore a promising solution for diagnosis and treatment of gastrointestinal related diseases.
Collapse
Affiliation(s)
- Zhuotong Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Meiyu Wan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Mingyang Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing 210023, PR China.
| |
Collapse
|
36
|
Rodríguez-Ramírez R, Fernández Peralbo MA, Mendía I, Long JCD, Sousa C, Cebolla Á. Urinary excretion of gluten immunoreactive peptides as an indicator of gastrointestinal function after fasting and dietary provocation in healthy volunteers. Front Immunol 2024; 15:1433304. [PMID: 39161759 PMCID: PMC11330814 DOI: 10.3389/fimmu.2024.1433304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Introduction Understanding intestinal permeability is paramount for elucidating gastrointestinal health and pathology. The size and nature of the molecule traversing the intestinal barrier offer crucial insights into various acute and chronic diseases, as well as the evolution of some conditions. This study aims to assess the urinary excretion kinetics of gluten immunogenic peptides (u-GIP), a unique class of dietary peptides detectable in urine, in volunteers under controlled dietary conditions. This evaluation should be compared to established probes like lactulose, a non-digestible disaccharide indicative of paracellular permeability, and mannitol, reflecting transcellular permeability. Methods Fifteen participants underwent simultaneous ingestion of standardized doses of gluten (10 g), lactulose (10 g), and mannitol (1 g) under fasting conditions for at least 8 hours pre-ingestion and during 6 hours post-ingestion period. Urine samples were collected over specified time intervals. Excretion patterns were analyzed, and correlations between the lactulose-to-mannitol ratio (LMR) and u-GIP parameters were assessed. Results The majority of u-GIP were detected within the first 12 hours post-ingestion. Analysis of the variability in cumulative excretion across two sample collection ranges demonstrated that lactulose and u-GIP exhibited similar onset and excretion dynamics, although GIP reached its maximum peak earlier than either lactulose or mannitol. Additionally, a moderate correlation was observed between the LMR and u-GIP parameters within the longest urine collection interval, indicating potential shared characteristics among permeability pathways. These findings suggest that extending urine collection beyond 6 hours may enhance data reliability. Discussion This study sheds light on the temporal dynamics of u-GIP in comparison to lactulose and mannitol, established probes for assessing intestinal permeability. The resemblance between u-GIP and lactulose excretion patterns aligns with the anticipated paracellular permeability pathway. The capacity to detect antigenic food protein fragments in urine opens novel avenues for studying protein metabolism and monitoring pathologies related to the digestive and intestinal systems.
Collapse
Affiliation(s)
- Raquel Rodríguez-Ramírez
- Research and Development Department, Biomedal S.L., Seville, Spain
- Inorganic Chemistry Department, Faculty of Science, University of Granada, Granada, Spain
| | | | - Irati Mendía
- Research and Development Department, Biomedal S.L., Seville, Spain
| | | | - Carolina Sousa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Ángel Cebolla
- Research and Development Department, Biomedal S.L., Seville, Spain
| |
Collapse
|
37
|
Alizadeh M, Wong U, Siaton BC, Patil SA, George L, Raufman JP, Scott WH, von Rosenvinge EC, Ravel J, Cross RK. Inflammatory Bowel Disease-Associated Arthritis Is Associated with Concomitant Autoimmune and Inflammatory Disorders. Dig Dis Sci 2024; 69:2961-2969. [PMID: 38902460 DOI: 10.1007/s10620-024-08478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/05/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Extraintestinal Manifestations (EIMs) are a common and potentially debilitating complication of Inflammatory Bowel Diseases (IBD), sometimes requiring additional treatment beyond those used to control intestinal disease. IBD-associated arthritis (IAA), a form of spondyloarthritis, is associated with several factors including disease location, sex, and IBD type. However, much remains unknown about other clinical factors predicting development of EIMs. Our goal was to identify additional factors associated with IAA. METHODS Participants in the LOCATION-IBD cohort were included in this analysis. We performed univariate and multivariate analysis of demographics, clinical data, and patient-reported outcomes data. RESULTS The LOCATION-IBD cohort included 182 participants with (n = 53) and without (n = 110) joint EIMs and with joint pain of unclear etiology (n = 19). In a multivariate analysis comparing those with and without joint EIMs, female sex (OR = 2.5, p = 0.014), the presence of concomitant autoimmune and inflammatory disorders (OR = 2.5, p = 0.038), and Crohn's disease (OR = 2.9, p = 0.026) were associated with the presence of joint EIMs. CONCLUSION This analysis reveals patients with IAA are more likely to have concomitant autoimmune disorders. Further studies are needed to confirm this association, understand the mechanisms underlying the common pathogenesis of these concurrent disorders, and evaluate their impact on the treatment of IAA.
Collapse
Affiliation(s)
- Madeline Alizadeh
- Institute for Genome Sciences, University of Maryland School of Medicine, HSF III, 670 W Baltimore St, Baltimore, MD, 21201, USA.
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA.
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA.
| | - Uni Wong
- Department of Veterans Affairs, Washington DC Veterans Health Administration, Washington, DC, USA
| | - Bernadette C Siaton
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seema A Patil
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Lauren George
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - William H Scott
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Erik C von Rosenvinge
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, HSF III, 670 W Baltimore St, Baltimore, MD, 21201, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Raymond K Cross
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, 22 S Greene St, Baltimore, MD, 21201, USA
| |
Collapse
|
38
|
Cao YY, Wang ZH, Pan YJ, Qi YP, Chen Q, Qin XM, Wang T, Shen GG, Jiang XG, Lu WH. The dual effects of dexmedetomidine on intestinal barrier and intestinal motility during sepsis. Surgery 2024; 176:379-385. [PMID: 38762380 DOI: 10.1016/j.surg.2024.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 03/13/2024] [Accepted: 03/25/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Sepsis, characterized by dysregulated host responses to infection, remains a critical global health concern, with high morbidity and mortality rates. The gastrointestinal tract assumes a pivotal role in sepsis due to its dual functionality as a protective barrier against injurious agents and as a regulator of motility. Dexmedetomidine, an α2-adrenergic agonist commonly employed in critical care settings, exhibits promise in influencing the maintenance of intestinal barrier integrity during sepsis. However, its impact on intestinal motility, a crucial component of intestinal function, remains incompletely understood. METHODS In this study, we investigated dexmedetomidine's multifaceted effects on intestinal barrier function and motility during sepsis using both in vitro and in vivo models. Sepsis was induced in Sprague-Dawley rats via cecal ligation and puncture. Rats were treated with dexmedetomidine post-cecal ligation and puncture, and various parameters were assessed to elucidate dexmedetomidine's impact. RESULTS Our findings revealed a dichotomous influence of dexmedetomidine on intestinal physiology. In septic rats, dexmedetomidine administration resulted in improved intestinal barrier integrity, as evidenced by reduced mucosal hyper-permeability and morphological alterations. However, a contrasting effect was observed on intestinal motility, as dexmedetomidine treatment inhibited both the frequency and amplitude of contractions in isolated intestinal strips and decreased the distance of ink migration in vivo. Additionally, dexmedetomidine suppressed the secretion of pro-motility hormones while having no influence on hormones that inhibit intestinal peristalsis. CONCLUSION The study revealed that during sepsis, dexmedetomidine exhibited protective effects on barrier integrity, although concurrently it hindered intestinal motility, partly attributed to its modulation of pro-motility hormone secretion. These findings underscore the necessity of a comprehensive understanding of dexmedetomidine's impact on multiple facets of gastrointestinal physiology in sepsis management, offering potential implications for therapeutic strategies and patient care.
Collapse
Affiliation(s)
- Ying-Ya Cao
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Zhong-Han Wang
- Department of Anesthesiology, The People's Hospital of Bozhou, Bozhou, Anhui, China
| | - You-Jun Pan
- Department of Critical Care Medicine, Wuhu Hospital, East China Normal University (The Second People's Hospital, Wuhu), Wuhu, Anhui, China
| | - Yu-Peng Qi
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Qun Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Xue-Mei Qin
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Tong Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Guang-Gui Shen
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China
| | - Xiao-Gan Jiang
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China.
| | - Wei-Hua Lu
- Department of Critical Care Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu Anhui, China; Anhui Province Clinical Research Center for Critical Respiratory Medicine, Wuhu Anhui, China.
| |
Collapse
|
39
|
Lacy BE, Wise JL, Cangemi DJ. Leaky Gut Syndrome: Myths and Management. Gastroenterol Hepatol (N Y) 2024; 20:264-272. [PMID: 39193076 PMCID: PMC11345991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Leaky gut syndrome is a condition widely popularized in the lay literature, although it is not currently accepted as a formal medical diagnosis. Multiple gastrointestinal symptoms are ascribed to leaky gut syndrome, including diarrhea, bloating, distension, abdominal pain, and dyspeptic symptoms of early satiety, nausea, and postprandial fullness. The etiology and pathophysiology of leaky gut syndrome are multifactorial; a preceding gastrointestinal infection, inflammatory bowel disease, and certain medications may be relevant factors in some patients. The diagnosis of leaky gut syndrome is problematic. Although patients are frequently informed that the diagnosis can be readily made using results from blood work or stool studies, no validated test currently exists to make this diagnosis. Patients report a variety of myths about the etiology, diagnosis, and treatment of leaky gut syndrome, which can cause alarm and can frequently lead to expensive, unnecessary tests and unproven, sometimes dangerous treatments. This article reviews some of the most common myths about leaky gut syndrome and provides data from the scientific literature to correct these statements. Management strategies, based on data, are provided when available.
Collapse
Affiliation(s)
- Brian E. Lacy
- Division of Gastroenterology & Hepatology, Mayo Clinic Jacksonville, Jacksonville, Florida
| | - Journey L. Wise
- Graduate Research Education Program, Mayo Clinic Rochester, Rochester, Minnesota
| | - David J. Cangemi
- Division of Gastroenterology & Hepatology, Mayo Clinic Jacksonville, Jacksonville, Florida
| |
Collapse
|
40
|
Mickiewicz-Góra D, Sznurkowska K, Skonieczna-Żydecka K, Drozd A, Borkowska A, Zagierski M, Troch J, Szlagatys-Sidorkiewicz A. Markers of Intestinal Permeability and Inflammation in Enterally Fed Children with Cerebral Palsy. Nutrients 2024; 16:2447. [PMID: 39125328 PMCID: PMC11314602 DOI: 10.3390/nu16152447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Cerebral palsy (CP) results in non-progressive damage to the central nervous system, leading to functional disorders of the gastrointestinal tract and requiring enteral nutrition via gastrostomy in some patients. The aim of the study was to assess the impact of enteral nutrition on intestinal inflammation expressed by stool calprotectin and intestinal permeability determined by fecal zonulin and IFABP, and to determine whether CP affects these parameters. The study group consisted of 30 children with CP, fed enterally (Cerebral Palsy Enteral Nutrition-CPEN), and two reference groups: 24 children with CP, fed orally with a standard diet (CPC-Cerebral Palsy Controls) and 24 healthy children (HC-healthy controls). The differences between these groups and between the combined CP groups (CPG and CPEN + CPC) and HC were analyzed. Fecal zonulin, calprotectin, and intestinal fatty acid-binding protein 2 (IFABP2) levels were determined by ELISA. The concentrations of fecal calprotectin and zonulin were significantly higher in the CPEN group than in the CPC group (p = 0.012, p = 0.025). When comparing the CPG (n = 53) with the HC group (n = 24), statistically significant differences were observed for calprotectin (p = 0.000018, higher in the CPG) and IFABP (p = 0.021, higher in HC). Enteral nutrition was associated in our cohort with increased fecal calprotectin and zonulin. Children with cerebral palsy presented with increased fecal calprotectin but not increased intestinal permeability expressed by stool zonulin.
Collapse
Affiliation(s)
- Dorota Mickiewicz-Góra
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Katarzyna Sznurkowska
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | | | - Arleta Drozd
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland;
| | - Anna Borkowska
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Maciej Zagierski
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Joanna Troch
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| | - Agnieszka Szlagatys-Sidorkiewicz
- Department of Paediatrics, Gastroenterology, Allergology & Paediatric Nutrition, Medical University of Gdansk, 80-803 Gdansk, Poland; (A.B.); (M.Z.); (J.T.); (A.S.-S.)
| |
Collapse
|
41
|
Yin C, Zhang M, Jin S, Zhou Y, Ding L, Lv Q, Huang Z, Zhou J, Chen J, Wang P, Zhang S, You Q. Mechanism of Salvia miltiorrhiza Bunge extract to alleviate Chronic Sleep Deprivation-Induced cognitive dysfunction in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155725. [PMID: 38772181 DOI: 10.1016/j.phymed.2024.155725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/16/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
BACKGROUND Bidirectional communication between the gut microbiota and the brain may play an essential role in the cognitive dysfunction associated with chronic sleep deprivation(CSD). Salvia miltiorrhiza Bunge (Danshen, DS), a famous Chinese medicine and functional tea, is extensively used to protect learning and memory capacities, although the mechanism of action remains unknown. PURPOSE The purpose of this research was to explore the efficacy and the underlying mechanism of DS in cognitive dysfunction caused by CSD. METHODS DS chemical composition was analyzed by UPLC-QTOF-MS/MS. Forty rats were randomly assigned to five groups (n = 8): control (CON), model (MOD), low- (1.35 g/kg, DSL), high-dose (2.70 g/kg, DSH) DS group, and Melatonin(100 mg/kg, MT) group. A CSD rat model was established over 21 days. DS's effects and the underlying mechanism were explored using the open-field test(OFT), Morris water-maze(MWM), tissue staining(Hematoxylin and Eosin Staining, Nissl staining, Alcian blue-periodic acid SCHIFF staining, and Immunofluorescence), enzyme-linked immunosorbent assay, Western blot, quantitative real-time polymerase chain reaction(qPCR), and 16S rRNA sequencing. RESULTS We demonstrated that CSD caused gut dysbiosis and cognitive dysfunction. Furthermore, 16S rRNA sequencing demonstrated that Firmicutes and Proteobacteria were more in fecal samples from model group rats, whereas Bacteroidota and Spirochaetota were less. DS therapy, on the contrary hand, greatly restored the gut microbial community, consequently alleviating cognitive impairment in rats. Further research revealed that DS administration reduced systemic inflammation via lowering intestinal inflammation and barrier disruption. Following that, DS therapy reduced Blood Brain Barrier(BBB) and neuronal damage, further decreasing neuroinflammation in the hippocampus(HP). Mechanistic studies revealed that DS therapy lowered lipopolysaccharide (LPS) levels in the HP, serum, and colon, consequently blocking the TLR4/MyD88/NF-κB signaling pathway and its downstream pro-inflammatory products(IL-1β, IL-6, TNF-α, iNOS, and COX2) in the HP and colon. CONCLUSION DS treatment dramatically improved spatial learning and memory impairments in rats with CSD by regulating the composition of the intestinal flora, preserving gut and brain barrier function, and reducing inflammation mediated by the LPS-TLR4 signaling pathway. Our findings provide novel insight into the mechanisms by which DS treats cognitive dysfunction caused by CSD.
Collapse
Affiliation(s)
- Chao Yin
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Meiya Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Shuna Jin
- Hubei Shizhen Laboratory, Wuhan 430065, PR China; School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuan Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Ding
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Qing Lv
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Zixuan Huang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Jiaqi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Jianmei Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Ping Wang
- Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China.
| | - Shunbo Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Qiuyun You
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Engineering Research Center of TCM Protection Technology and New Product Development for the Elderly Brain Health, Ministry of Education, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China.
| |
Collapse
|
42
|
Kunath BJ, De Rudder C, Laczny CC, Letellier E, Wilmes P. The oral-gut microbiome axis in health and disease. Nat Rev Microbiol 2024:10.1038/s41579-024-01075-5. [PMID: 39039286 DOI: 10.1038/s41579-024-01075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
The human body hosts trillions of microorganisms throughout many diverse habitats with different physico-chemical characteristics. Among them, the oral cavity and the gut harbour some of the most dense and diverse microbial communities. Although these two sites are physiologically distinct, they are directly connected and can influence each other in several ways. For example, oral microorganisms can reach and colonize the gastrointestinal tract, particularly in the context of gut dysbiosis. However, the mechanisms of colonization and the role that the oral microbiome plays in causing or exacerbating diseases in other organs have not yet been fully elucidated. Here, we describe recent advances in our understanding of how the oral and intestinal microbiota interplay in relation to their impact on human health and disease.
Collapse
Affiliation(s)
- Benoit J Kunath
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
43
|
Nuszkiewicz J, Kukulska-Pawluczuk B, Piec K, Jarek DJ, Motolko K, Szewczyk-Golec K, Woźniak A. Intersecting Pathways: The Role of Metabolic Dysregulation, Gastrointestinal Microbiome, and Inflammation in Acute Ischemic Stroke Pathogenesis and Outcomes. J Clin Med 2024; 13:4258. [PMID: 39064298 PMCID: PMC11278353 DOI: 10.3390/jcm13144258] [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: 05/16/2024] [Revised: 07/13/2024] [Accepted: 07/20/2024] [Indexed: 07/28/2024] Open
Abstract
Acute ischemic stroke (AIS) remains a major cause of mortality and long-term disability worldwide, driven by complex and multifaceted etiological factors. Metabolic dysregulation, gastrointestinal microbiome alterations, and systemic inflammation are emerging as significant contributors to AIS pathogenesis. This review addresses the critical need to understand how these factors interact to influence AIS risk and outcomes. We aim to elucidate the roles of dysregulated adipokines in obesity, the impact of gut microbiota disruptions, and the neuroinflammatory cascade initiated by lipopolysaccharides (LPS) in AIS. Dysregulated adipokines in obesity exacerbate inflammatory responses, increasing AIS risk and severity. Disruptions in the gut microbiota and subsequent LPS-induced neuroinflammation further link systemic inflammation to AIS. Advances in neuroimaging and biomarker development have improved diagnostic precision. Here, we highlight the need for a multifaceted approach to AIS management, integrating metabolic, microbiota, and inflammatory insights. Potential therapeutic strategies targeting these pathways could significantly improve AIS prevention and treatment. Future research should focus on further elucidating these pathways and developing targeted interventions to mitigate the impacts of metabolic dysregulation, microbiome imbalances, and inflammation on AIS.
Collapse
Affiliation(s)
- Jarosław Nuszkiewicz
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Beata Kukulska-Pawluczuk
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Katarzyna Piec
- Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland; (B.K.-P.); (K.P.)
| | - Dorian Julian Jarek
- Student Research Club of Medical Biology and Biochemistry, Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Karina Motolko
- Student Research Club of Neurology, Department of Neurology, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 9 M. Skłodowskiej—Curie St., 85-094 Bydgoszcz, Poland;
| | - Karolina Szewczyk-Golec
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 24 Karłowicza St., 85-092 Bydgoszcz, Poland;
| |
Collapse
|
44
|
Schneck NA, Moghieb A, Teague C, Perez HL. Development and application of a multi-sugar assay to assess intestinal permeability. Bioanalysis 2024; 16:849-861. [PMID: 39023344 PMCID: PMC11457621 DOI: 10.1080/17576180.2024.2374168] [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: 04/16/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
Aim: Bioanalytical assays to measure rhamnose, erythritol, lactulose and sucralose in human urine and plasma were developed to support an indomethacin challenge study for intestinal permeability assessment in healthy participants.Methods: The multi-sugar assays utilized 5-μl sample matrix and a simple chemical derivatization with acetic anhydride, followed by RPLC-MS/MS detection.Results: Rhamnose and erythritol quantification was established between 1.00-1,000 μg/ml in urine and 250-250,000 ng/ml in plasma. For lactulose and sucralose, dynamic ranges of 0.1-100 μg/ml (urine) and 25-25,000 ng/ml (plasma) were applied for biological measurements.Conclusion: This work helped overcome some of the common analytical challenges associated with the bioanalysis of mono- and disaccharides and achieved improved limits of quantification.
Collapse
Affiliation(s)
- Nicole A Schneck
- GSK, Biomarker Platforms, Precision Medicine, GSK, 1250 S Collegeville Rd, Collegeville, PA19426, USA
| | - Ahmed Moghieb
- GSK, Biomarker Platforms, Precision Medicine, GSK, 1250 S Collegeville Rd, Collegeville, PA19426, USA
| | - Claire Teague
- GSK, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Hermes Licea Perez
- GSK, Biomarker Platforms, Precision Medicine, GSK, 1250 S Collegeville Rd, Collegeville, PA19426, USA
| |
Collapse
|
45
|
Zhang D, Li J, Zhang B, Shao Y, Wang Z. Two Doses of Zn Induced Different Microbiota Profiles and Dietary Zinc Supplementation Affects the Intestinal Microbial Profile, Intestinal Microarchitecture and Immune Response in Pigeons. Animals (Basel) 2024; 14:2087. [PMID: 39061548 PMCID: PMC11273959 DOI: 10.3390/ani14142087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
We aimed to explore the effects of two different doses of Zn on the fecal microbiota in pigeons and the correlation between these effects and intestinal immune status. Zn doses affected pigeon growth performance, and pigeons in the T60 (60 mg/kg Zn) and T90 (90 mg/kg Zn) groups exhibited higher villus height and crypt depth in duodenum and ileum compared to the control group, respectively. Supplementation with Zn increased the expression of the IL8, CD798, TJP and NKTR genes (p < 0.05), while enhancing serum immunoglobulin (Ig) G, IgM, and IgA concentrations compared to the control pigeons (p < 0.05). T60 treatment reduced relative Actinobacteriota abundance, while Lactobacillus spp. abundance was highest in the T90 group compared to the two other groups. The core functional genera significantly associated with immune indices in these pigeons were Rhodococcus erythropolis and Lactobacillus ponti. Our findings will help facilitate the application of dietary Zn intake in pig production.
Collapse
Affiliation(s)
| | | | | | - Yuxin Shao
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (D.Z.); (J.L.); (B.Z.)
| | - Zheng Wang
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China; (D.Z.); (J.L.); (B.Z.)
| |
Collapse
|
46
|
Xiao JH, Wang Y, Zhang XM, Wang WX, Zhang Q, Tang YP, Yue SJ. Intestinal permeability in human cardiovascular diseases: a systematic review and meta-analysis. Front Nutr 2024; 11:1361126. [PMID: 39086542 PMCID: PMC11289889 DOI: 10.3389/fnut.2024.1361126] [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: 01/05/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Background There is a link between cardiovascular diseases and intestinal permeability, but it is not clear. This review aimed to elucidate intestinal permeability in cardiovascular diseases by meta-analysis. Methods Multidisciplinary electronic databases were searched from the database creation to April 2023. All included studies were assessed for risk of bias according to the Joanna Briggs Institute Critical Appraisal Checklist. The heterogeneity of each study was estimated using the I2 statistic, and the data were analyzed using Review Manager 5.3 and Stata 16.0. Results In total, studies in 13 pieces of literature were included in the quantitative meta-analysis. These studies were conducted among 1,321 subjects mostly older than 48. Patients had higher levels of intestinal permeability markers (lipopolysaccharide, d-lactate, zonulin, serum diamine oxidase, lipopolysaccharide-binding protein, intestinal fatty acid binding protein, and melibiose/rhamnose) than controls (standard mean difference SMD = 1.50; 95% CI = 1.31-1.88; p < 0.00001). Similarly, lipopolysaccharide levels were higher in patients than in controls (SMD = 1.61; 95% CI = 1.02-2.21; p < 0.00001); d-lactate levels were higher in patients than in controls (SMD = 1.16; 95% CI = 0.23-2.08; p = 0.01); zonulin levels were higher in patients than in controls (SMD = 1.74; 95% CI = 1.45-2.03; p < 0.00001); serum diamine oxidase levels were higher in patients than in controls (SMD = 2.51; 95% CI = 0.29-4.73; p = 0.03). Conclusion The results of the meta-analysis verified that the intestinal barrier was damaged and intestinal permeability was increased in patients with cardiovascular diseases. These markers may become a means of the diagnosis and treatment of cardiovascular diseases. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=414296, identifier CRD42023414296.
Collapse
Affiliation(s)
- Jiang-Hong Xiao
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Yu Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Xi-Mei Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Wen-Xiao Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qiao Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi University of Chinese Medicine, Xi’an, China
- International Joint Research Center on Resource Utilization and Quality Evaluation of Traditional Chinese Medicine of Hebei Province, Hebei University of Chinese Medicine, Shijiazhuang, China
| |
Collapse
|
47
|
Zhang J, Huang Y, Li H, Xu P, Liu Q, Sun Y, Zhang Z, Wu T, Tang Q, Jia Q, Xia Y, Xu Y, Jing X, Li J, Mo L, Xie W, Qu A, He J, Li Y. B3galt5 functions as a PXR target gene and regulates obesity and insulin resistance by maintaining intestinal integrity. Nat Commun 2024; 15:5919. [PMID: 39004626 PMCID: PMC11247088 DOI: 10.1038/s41467-024-50198-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Pregnane X receptor (PXR) has been reported to regulate glycolipid metabolism. The dysfunction of intestinal barrier contributes to metabolic disorders. However, the role of intestinal PXR in metabolic diseases remains largely unknown. Here, we show that activation of PXR by tributyl citrate (TBC), an intestinal-selective PXR agonist, improves high fat diet (HFD)-induced obesity. The metabolic benefit of intestinal PXR activation is associated with upregulation of β-1,3 galactosyltransferase 5 (B3galt5). Our results reveal that B3galt5 mainly expresses in the intestine and is a direct PXR transcriptional target. B3galt5 knockout exacerbates HFD-induced obesity, insulin resistance and inflammation. Mechanistically, B3galt5 is essential to maintain the integrity of intestinal mucus barrier. B3galt5 ablation impairs the O-glycosylation of mucin2, destabilizes the mucus layer, and increases intestinal permeability. Furthermore, B3galt5 deficiency abolishes the beneficial effect of intestinal PXR activation on metabolic disorders. Our results suggest the intestinal-selective PXR activation regulates B3galt5 expression and maintains metabolic homeostasis, making it a potential therapeutic strategy in obesity.
Collapse
Affiliation(s)
- Jinhang Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ya Huang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Pharmacy, GuiQian International General Hospital, Guiyang, China
| | - Hong Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qinhui Liu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Disease, Kunming, Yunnan Province, China
| | - Zijing Zhang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tong Wu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qin Tang
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qingyi Jia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yan Xia
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiandan Jing
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiahui Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, P.R. China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yanping Li
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
48
|
Lopes EW, Turpin W, Croitoru K, Colombel JF, Torres J. Prediction and Prevention of Inflammatory Bowel Disease. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00597-4. [PMID: 38996831 DOI: 10.1016/j.cgh.2024.05.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 07/14/2024]
Affiliation(s)
- Emily W Lopes
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kenneth Croitoru
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Division of Gastroenterology & Hepatology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Jean-Frederic Colombel
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joana Torres
- Division of Gastrenterology, Hospital da Luz, Lisboa, Portugal; Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal; Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
49
|
Dmytriv TR, Storey KB, Lushchak VI. Intestinal barrier permeability: the influence of gut microbiota, nutrition, and exercise. Front Physiol 2024; 15:1380713. [PMID: 39040079 PMCID: PMC11260943 DOI: 10.3389/fphys.2024.1380713] [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: 02/15/2024] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
The intestinal wall is a selectively permeable barrier between the content of the intestinal lumen and the internal environment of the body. Disturbances of intestinal wall permeability can potentially lead to unwanted activation of the enteric immune system due to excessive contact with gut microbiota and its components, and the development of endotoxemia, when the level of bacterial lipopolysaccharides increases in the blood, causing chronic low-intensity inflammation. In this review, the following aspects are covered: the structure of the intestinal wall barrier; the influence of the gut microbiota on the permeability of the intestinal wall via the regulation of functioning of tight junction proteins, synthesis/degradation of mucus and antioxidant effects; the molecular mechanisms of activation of the pro-inflammatory response caused by bacterial invasion through the TLR4-induced TIRAP/MyD88 and TRAM/TRIF signaling cascades; the influence of nutrition on intestinal permeability, and the influence of exercise with an emphasis on exercise-induced heat stress and hypoxia. Overall, this review provides some insight into how to prevent excessive intestinal barrier permeability and the associated inflammatory processes involved in many if not most pathologies. Some diets and physical exercise are supposed to be non-pharmacological approaches to maintain the integrity of intestinal barrier function and provide its efficient operation. However, at an early age, the increased intestinal permeability has a hormetic effect and contributes to the development of the immune system.
Collapse
Affiliation(s)
- Tetiana R. Dmytriv
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| | | | - Volodymyr I. Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
- Research and Development University, Ivano-Frankivsk, Ukraine
| |
Collapse
|
50
|
Wu Y, Li Y, Zheng Q, Li L. The Efficacy of Probiotics, Prebiotics, Synbiotics, and Fecal Microbiota Transplantation in Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. Nutrients 2024; 16:2114. [PMID: 38999862 PMCID: PMC11243554 DOI: 10.3390/nu16132114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder with gut microbiota imbalance playing a significant role. There are increasing numbers of research studies exploring treatment options involving probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT), but it is still uncertain which treatment option is superior. The research was conducted on various databases and unpublished trial data (up to February 2023). Randomized controlled trials (RCTs) were screened for adult patients with IBS comparing interventions with placebo. Probiotics, prebiotics, synbiotics, and FMT were assessed for their impact using mean difference and Bayesian network meta-analysis. Out of 6528 articles, 54 were included for probiotics, 7 for prebiotics/synbiotics, and 6 for FMT. Probiotics showed improvement in IBS symptoms, particularly with Bifidobacterium and Lactobacillus strains. Prebiotics and synbiotics did not show significant improvement. Network meta-analysis indicated the favorable effects of probiotics (OR = 0.53, 95% CI, 0.48 to 0.59) and FMT (OR = 0.46, 95% CI, 0.33 to 0.64) on IBS, with no serious adverse events reported. In short, probiotics and FMT are effective for managing IBS, with Bifidobacterium and Lactobacillus being dominant strains. However, the most effective probiotic combination or strain remains unclear, while prebiotics and synbiotics did not show significant improvement.
Collapse
Affiliation(s)
- Youhe Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Yuetong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Qi Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou 310003, China
| |
Collapse
|