51
|
Bernardini I, Matozzo V, Valsecchi S, Peruzza L, Rovere GD, Polesello S, Iori S, Marin MG, Fabrello J, Ciscato M, Masiero L, Bonato M, Santovito G, Boffo L, Bargelloni L, Milan M, Patarnello T. The new PFAS C6O4 and its effects on marine invertebrates: First evidence of transcriptional and microbiota changes in the Manila clam Ruditapes philippinarum. ENVIRONMENT INTERNATIONAL 2021; 152:106484. [PMID: 33740673 DOI: 10.1016/j.envint.2021.106484] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 06/12/2023]
Abstract
There is growing concern for the wide use ofperfluorooctanoic acid (PFOA) because of its toxic effects on the environment and on human health. A new compound - the so called C6O4 (perfluoro ([5-methoxy-1,3-dioxolan-4-yl]oxy) acetic acid) - was recently introduced as one of the alternative to traditional PFOA, however this was done without any scientific evidence of the effects of C6O4 when dispersed into the environment. Recently, the Regional Agency for the Protection of the Environment of Veneto (Italy) detected high levels of C6O4 in groundwater and in the Po river, increasing the alarm for the potential effects of this chemical into the natural environment. The present study investigates for the first time the effects of C6O4 on the Manila clam Ruditapes philippinarum exposed to environmental realistic concentrations of C6O4 (0.1 µg/L and 1 µg/L) for 7 and 21 days. Furthermore, in order to better understand if C6O4 is a valid and less hazardous alternative to its substitute, microbial and transcriptomic alterations were also investigated in clams exposed to 1 µg/L ofPFOA. Results indicate that C6O4 may cause significant perturbations to the digestive gland microbiota, likely determining the impairment of host physiological homeostasis. Despite chemical analyses suggest a 5 times lower accumulation potential of C604 as compared to PFOA in clam soft tissues, transcriptional analyses reveal several alterations of gene expression profile. A large part of the altered pathways, including immune response, apoptosis regulation, nervous system development, lipid metabolism and cell membrane is the same in C6O4 and PFOA exposed clams. In addition, clams exposed to C6O4 showed dose-dependent responses as well as possible narcotic or neurotoxic effects and reduced activation of genes involved in xenobiotic metabolism. Overall, the present study suggests that the potential risks for marine organism following environmental contamination are not reduced by replacing PFOA with C6O4. In addition, the detection of both C6O4 and PFOA into tissues of clams inhabiting the Lagoon of Venice - where there are no point sources of either compounds - recommends a similar capacity to spread throughout the environment. These results prompt the urgent need to re-evaluate the use of C6O4 as it may represent not only an environmental hazard but also a potential risk for human health.
Collapse
Affiliation(s)
- Ilaria Bernardini
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - Valerio Matozzo
- Department of Biology, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - Sara Valsecchi
- Water Research Institute, Italian National Research Council (IRSA-CNR), Via Mulino 19, 20861 Brugherio, MB, Italy
| | - Luca Peruzza
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - Giulia Dalla Rovere
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - Stefano Polesello
- Water Research Institute, Italian National Research Council (IRSA-CNR), Via Mulino 19, 20861 Brugherio, MB, Italy
| | - Silvia Iori
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | | | - Jacopo Fabrello
- Department of Biology, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - Maria Ciscato
- Department of Biology, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - Luciano Masiero
- Department of Biology, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - Marco Bonato
- Department of Biology, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | - Gianfranco Santovito
- Department of Biology, University of Padova, Via Bassi 58/B, 35131 Padova, Italy
| | | | - Luca Bargelloni
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| | - Massimo Milan
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy.
| | - Tomaso Patarnello
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'Università 16, 35020 Legnaro (PD), Italy
| |
Collapse
|
52
|
Wu Y, Zhang X, Pi Y, Han D, Feng C, Zhao J, Chen L, Che D, Bao H, Xie Z, Wang J. Maternal galactooligosaccharides supplementation programmed immune defense, microbial colonization and intestinal development in piglets. Food Funct 2021; 12:7260-7270. [PMID: 34165467 DOI: 10.1039/d1fo00084e] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The benefits of galactooligosaccharides (GOS) in neonates have been confirmed. However, the effects of nutritional programming by maternal GOS intervention on microbial colonization and intestinal development in the offspring remain unclear. In the present study, late gestational sows were fed with GOS (10 g d-1 added into the diet) or not until parturition, and the performances, immune status, microbiota composition and intestinal barriers in their piglets on day 21 were compared. GOS supplementation in pregnant sows improved their litter characteristics and the growth performance of their piglets during the neonatal stage (day 21), and elevated the plasma IgA levels in both sows and their piglets (P < 0.05). GOS intervention enriched fecal Alloprevotella and Ruminoclostridium_1 in gestational sows and vertically increased fecal Alloprevotella and Ruminococcaceae in their piglets (P < 0.05). Moreover, maternal GOS intervention increased fecal acetate (P < 0.05) and improved the intestinal barriers of their piglets by upregulating intestinal tight junctions (Occludin, Claudin-1, ZO-1), the goblet cell number and Mucin-2 (P < 0.05), which correlated positively with the colonized microbiota (P < 0.05). In summary, GOS supplementation for sows during late gestation nutritionally programmed maternal specific microbes and IgA of their offspring. This neonatal programming showed positive potential in promoting the intestinal barriers, immune defense, and growth performance of the piglets. Our findings provide evidence for maternal nutritional programming in neonates and insights for future application of GOS in maternal-neonatal nutrition.
Collapse
Affiliation(s)
- Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
53
|
Peng J, Zhang M, Yao G, Kwok LY, Zhang W. Probiotics as Adjunctive Treatment for Patients Contracted COVID-19: Current Understanding and Future Needs. Front Nutr 2021; 8:669808. [PMID: 34179059 PMCID: PMC8222530 DOI: 10.3389/fnut.2021.669808] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 05/17/2021] [Indexed: 01/07/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) is caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which rages all over the world and seriously threatens human life and health. Currently, there is no optimal treatment for COVID-19, and emerging evidence found that COVID-19 infection results in gut microbiota dysbiosis. The intestinal microbial richness of patients of COVID-19 does not return to normal levels even six months after recovery, but probiotic adjunctive treatment has been found to restore gut homeostasis. An updated PubMed search returned four finished clinical trials that supported the use of probiotics as adjunctive treatment for COVID-19, while at least six clinical trials aiming to investigate beneficial effects of probiotic intake in managing COVID-19 are currently in progress worldwide. Here in we tentatively summarized the understanding of the actions and potential mechanisms of probiotics in the management of COVID-19. We also highlighted some future needs for probiotic researchers in the field. The success in using probiotics as adjunctive treatment for COVID-19 has expanded the scope of application of probiotics, meanwhile deepening our knowledge in the physiological function of probiotics in modulating the gut-lung axis.
Collapse
Affiliation(s)
- Jiangying Peng
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Guoqiang Yao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
| | - Wenyi Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China,Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China,*Correspondence: Wenyi Zhang
| |
Collapse
|
54
|
Guo W, Zhou X, Li X, Zhu Q, Peng J, Zhu B, Zheng X, Lu Y, Yang D, Wang B, Wang J. Depletion of Gut Microbiota Impairs Gut Barrier Function and Antiviral Immune Defense in the Liver. Front Immunol 2021; 12:636803. [PMID: 33841420 PMCID: PMC8027085 DOI: 10.3389/fimmu.2021.636803] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Commensal gut microbiota protects the immune defense of extra-intestinal organs. Gut microbiota depletion by antibiotics can impair host antiviral immune responses and alter hepatitis B virus (HBV) infection outcomes. However, how gut microbiota modulates antiviral immune response in the liver remains unclear. Here, mice were treated with broad-spectrum antibiotics to deplete gut microbiota. Gut integrity was evaluated, and translocation of live commensal gut bacteria and their components into the liver was investigated. An HBV infection model was established to evaluate impairment of antiviral immune response in the liver after gut microbiota depletion. We found that gut microbiota depletion was associated with impairment of colon epithelial integrity, and live commensal gut microbiota could translocate to the liver. Further, T cell antiviral function in the liver was impaired, partially relying on enhanced PD-1 expression, and HBV immune clearance was hampered. In conclusion, gut microbiota depletion by antibiotics can impair gut barrier function and suppress T cell antiviral immune response in the liver.
Collapse
Affiliation(s)
- Weina Guo
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhou
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoran Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingfeng Zhu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Peng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinping Lu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baoju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junzhong Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
55
|
McIntosh AT, Wei R, Ahn J, Aouizerat BE, Kassaye SG, Augenbraun MH, Price JC, French AL, Gange SJ, Anastos KM, Goldman R. A genomic variant of ALPK2 is associated with increased liver fibrosis risk in HIV/HCV coinfected women. PLoS One 2021; 16:e0247277. [PMID: 33705408 PMCID: PMC7951908 DOI: 10.1371/journal.pone.0247277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/03/2021] [Indexed: 01/21/2023] Open
Abstract
HIV coinfection is associated with more rapid liver fibrosis progression in hepatitis C (HCV) infection. Recently, much work has been done to improve outcomes of liver disease and to identify targets for pharmacological intervention in coinfected patients. In this study, we analyzed clinical data of 1,858 participants from the Women's Interagency HIV Study (WIHS) to characterize risk factors associated with changes in the APRI and FIB-4 surrogate measurements for advanced fibrosis. We assessed 887 non-synonymous single nucleotide variants (nsSNV) in a subset of 661 coinfected participants for genetic associations with changes in liver fibrosis risk. The variants utilized produced amino acid substitutions that either altered an N-linked glycosylation (NxS/T) sequon or mapped to a gene related to glycosylation processes. Seven variants were associated with an increased likelihood of liver fibrosis. The most common variant, ALPK2 rs3809973, was associated with liver fibrosis in HIV/HCV coinfected patients; individuals homozygous for the rare C allele displayed elevated APRI (0.61, 95% CI, 0.334 to 0.875) and FIB-4 (0.74, 95% CI, 0.336 to 1.144) relative to those coinfected women without the variant. Although warranting replication, ALPK2 rs3809973 may show utility to detect individuals at increased risk for liver disease progression.
Collapse
Affiliation(s)
- Alec T. McIntosh
- Department of Oncology, Georgetown University, Washington, DC, United States of America
| | - Renhuizi Wei
- Department of Oncology, Georgetown University, Washington, DC, United States of America
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics & Biomathematics, Georgetown University Medical Center, Washington, DC, United States of America
| | - Brad E. Aouizerat
- Bluestone Center for Clinical Research, College of Dentistry, New York University, New York, New York, United States of America
| | - Seble G. Kassaye
- Department of Infectious Diseases, Georgetown University Medical Center, Washington, DC, United States of America
| | - Michael H. Augenbraun
- Division of Infectious Diseases, Department of Medicine, State University of New York, Downstate Medical Center, Brooklyn, New York, United States of America
| | - Jennifer C. Price
- Division of Liver Diseases, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Audrey L. French
- Division of Infectious Disease, Department of Internal Medicine, Stroger Hospital of Cook County, Chicago, Illinois, United States of America
| | - Stephen J. Gange
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kathryn M. Anastos
- Departments of Medicine and Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Radoslav Goldman
- Department of Oncology, Georgetown University, Washington, DC, United States of America
- Department of Biochemistry and Molecular & Cell Biology, Georgetown University Medical Center, Washington, DC, United States of America
- Clinical Translational Glycoscience Research Center, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
56
|
Wensley MR, Tokach MD, Woodworth JC, Goodband RD, Gebhardt JT, DeRouchey JM, McKilligan D. Maintaining continuity of nutrient intake after weaning. I. Review of pre-weaning strategies. Transl Anim Sci 2021; 5:txab021. [PMID: 33750992 PMCID: PMC7963027 DOI: 10.1093/tas/txab021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/02/2021] [Indexed: 12/19/2022] Open
Abstract
Weaning is a crucial phase of swine production marked by a multitude of biological and environmental stressors, which have a significant impact on immediate postweaning behavior and feed intake (FI). During this time, the piglet's gastrointestinal (GI) system is also undergoing extensive epithelial, immune, and nervous system development. In this review, our objective is to describe the different preweaning strategies that can be used to minimize nutrient intake disruption and improve FI in the immediate postweaning period. Reducing nutrient disruption postweaning can be accomplished through the implementation of management and nutritional strategies. Research consistently demonstrates that weaning older, more developmentally mature pigs helps prevent many of the adverse GI effects associated with weaning stress. Providing creep feed to pigs during lactation is another reliable strategy that has been shown to increase immediate postweaning FI by acclimating pigs to solid feed prior to weaning. Likewise, socialization by allowing pigs to mix before weaning improves social skills, minimizing mixing stress, and aggression-related injury immediately postweaning. Supplemental milk replacer has also been shown to elicit a positive response in preweaning growth performance, which may help to reduce preweaning mortality. While socialization and milk replacer are acknowledged to ease the weaning transition, these strategies have not been widely adopted due to labor and application challenges. Additionally, the cost of milk replacer and logistics of retrofitting farrowing houses to accommodate litter socialization have limited adaptation. Further exploration of maternal nutrition strategies, particularly fetal imprinting, is needed to better understand the implications of perinatal learning. Other areas for future research include, combining environmental enrichment with feeding strategies, such as large destructible pellets or play feeders, as well as determining at what time point producers should start socializing pigs before weaning. While more research is needed to develop strategic preweaning management programs, many of the strategies presented in this review provide opportunities for producers to minimize nutrient intake disruption by preventing feed neophobia, reducing stress, and easing the wean pig transition.
Collapse
Affiliation(s)
- Madie R Wensley
- Department of Animal Sciences and Industry, College of Agriculture, Manhattan, KS 66506-0201, USA
| | - Mike D Tokach
- Department of Animal Sciences and Industry, College of Agriculture, Manhattan, KS 66506-0201, USA
| | - Jason C Woodworth
- Department of Animal Sciences and Industry, College of Agriculture, Manhattan, KS 66506-0201, USA
| | - Robert D Goodband
- Department of Animal Sciences and Industry, College of Agriculture, Manhattan, KS 66506-0201, USA
| | - Jordan T Gebhardt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine Kansas State University, Manhattan, KS 66506-0201, USA
| | - Joel M DeRouchey
- Department of Animal Sciences and Industry, College of Agriculture, Manhattan, KS 66506-0201, USA
| | | |
Collapse
|
57
|
Utay NS, Monczor AN, Somasunderam A, Lupo S, Jiang ZD, Alexander AS, Finkelman M, Vigil KJ, Lake JE, Hanson B, DuPont HL, Arduino RC. Evaluation of Six Weekly Oral Fecal Microbiota Transplants in People with HIV. Pathog Immun 2020; 5:364-381. [PMID: 33501400 PMCID: PMC7815055 DOI: 10.20411/pai.v5i1.388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022] Open
Abstract
Background Reduced microbiota diversity (dysbiosis) in people with HIV (PWH) likely contributes to inflammation, a driver of morbidity and mortality. We aimed to evaluate the safety and tolerability of 6 weekly oral fecal microbiota transplants (FMT) administered to reverse this dysbiosis. Methods Six PWH on suppressive antiretroviral therapy (ART) received 6 weekly doses of lyophilized fecal microbiota product from healthy donors. Shotgun sequencing on stool before, after last FMT, and 20 weeks thereafter was performed. Inflammation and gut permeability biomarkers were measured. Results Median age at week 0 was 39 years, CD4+ T cell count 496 cells/mm3, HIV RNA levels <20 copies/mL. FMT was safe and well-tolerated. α diversity increased in 4 participants from weeks 0 to 6, including the 3 with the lowest α diversity at week 0. At week 26, α diversity more closely resembled week 0 than week 6 in these 4 participants. Metagenomic analysis showed no consistent changes across all participants. One participant had high gut permeability and inflammation biomarker levels and low α diversity that improved between weeks 0 and 6 with a shift in distribution. Conclusions Weekly FMT was safe and well-tolerated. α diversity increased in participants with the lowest baseline α diversity during the treatment period. Future randomized, controlled trials of FMT should consider evaluating PWH with greater inflammation, gut damage, or dysbiosis as this population may be most likely to show a significant response.ClinicalTrials.gov Identifier: NCT03329560.
Collapse
Affiliation(s)
- Netanya S Utay
- Division of General Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas.,Kelsey Research Foundation, Houston, Texas
| | - Ana N Monczor
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Anoma Somasunderam
- Division of General Medicine, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Sofia Lupo
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Zhi-Dong Jiang
- School of Public Health at The University of Texas Health Science Center at Houston, Houston, Texas
| | | | | | - Karen J Vigil
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Jordan E Lake
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Blake Hanson
- School of Public Health at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Herbert L DuPont
- Kelsey Research Foundation, Houston, Texas.,School of Public Health at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Roberto C Arduino
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
58
|
Del Bo' C, Bernardi S, Cherubini A, Porrini M, Gargari G, Hidalgo-Liberona N, González-Domínguez R, Zamora-Ros R, Peron G, Marino M, Gigliotti L, Winterbone MS, Kirkup B, Kroon PA, Andres-Lacueva C, Guglielmetti S, Riso P. A polyphenol-rich dietary pattern improves intestinal permeability, evaluated as serum zonulin levels, in older subjects: The MaPLE randomised controlled trial. Clin Nutr 2020; 40:3006-3018. [PMID: 33388204 DOI: 10.1016/j.clnu.2020.12.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 12/02/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIM Increased intestinal permeability (IP) can occur in older people and contribute to the activation of the immune system and inflammation. Dietary interventions may represent a potential strategy to reduce IP. In this regard, specific food bioactives such as polyphenols have been proposed as potential IP modulator due to their ability to affect several critical targets and pathways that control IP. The trial aimed to test the hypothesis that a polyphenol-rich dietary pattern can decrease serum zonulin levels, an IP surrogate marker involved in tight junction modulation, and can beneficially alter the intestinal microbiota, and IP-associated biochemical and clinical markers in older subjects. METHODS A randomised, controlled, cross-over intervention trial was performed. Sixty-six subjects (aged ≥ 60 y) with increased IP based on serum zonulin levels, were randomly allocated to one of the two arms of the intervention consisting of a control diet (C-diet) vs. a polyphenol-rich diet (PR-diet). Each intervention was 8-week long and separated by an 8-week wash out period. At the beginning and at the end of each intervention period, serum samples were collected for the quantification of zonulin and other biological markers. Faecal samples were also collected to investigate the intestinal microbial ecosystem. In addition, anthropometrical/physical/biochemical parameters and food intake were evaluated. RESULTS Fifty-one subjects successfully completed the intervention and a high compliance to the dietary protocols was demonstrated. Overall, polyphenol intake significantly increased from a mean of 812 mg/day in the C diet to 1391 mg/day in the PR-diet. Two-way analysis of variance showed a significant effect of treatment (p = 0.008) and treatment × time interaction (p = 0.025) on serum zonulin levels, which decreased after the 8-week PR-diet. In addition, a treatment × time interaction was observed showing a reduction of diastolic blood pressure (p = 0.028) following the PR-diet, which was strongest in those not using antihypertensive drugs. A decrease in both diastolic (p = 0.043) and systolic blood pressure (p = 0.042) was observed in women. Interestingly, a significant increase in fibre-fermenting and butyrate-producing bacteria such as the family Ruminococcaceae and members of the genus Faecalibacterium was observed following the PR intervention. The efficacy of this dietary intervention was greater in subjects with higher serum zonulin at baseline, who showed more pronounced alterations in the markers under study. Furthermore, zonulin reduction was also stronger among subjects with higher body mass index and with insulin resistance at baseline, thus demonstrating the close interplay between IP and metabolic features. CONCLUSIONS These data show, for the first time, that a PR-diet can reduce serum zonulin levels, an indirect marker of IP. In addition, PR-diet reduced blood pressure and increased fibre-fermenting and butyrate-producing bacteria. These findings may represent an initial breakthrough for further intervention studies evaluating possible dietary treatments for the management of IP, inflammation and gut function in different target populations. THIS STUDY WAS REGISTERED AT WWW.ISRCTN. ORG AS ISRCTN10214981.
Collapse
Affiliation(s)
- Cristian Del Bo'
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Stefano Bernardi
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Antonio Cherubini
- Geriatria, Accettazione Geriatrica e Centro di Ricerca per l'Invecchiamento, IRCCS INRCA, 60127 Ancona, Italy
| | - Marisa Porrini
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Giorgio Gargari
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Nicole Hidalgo-Liberona
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain; CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Raúl González-Domínguez
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain; CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Raul Zamora-Ros
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain; Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), Spain
| | - Gregorio Peron
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain; CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Mirko Marino
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Letizia Gigliotti
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Mark S Winterbone
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Benjamin Kirkup
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Paul A Kroon
- Quadram Institute Bioscience, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain; CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028 Barcelona, Spain
| | - Simone Guglielmetti
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy
| | - Patrizia Riso
- Università, degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milan, Italy.
| |
Collapse
|
59
|
The modulatory effect of plant polysaccharides on gut flora and the implication for neurodegenerative diseases from the perspective of the microbiota-gut-brain axis. Int J Biol Macromol 2020; 164:1484-1492. [DOI: 10.1016/j.ijbiomac.2020.07.208] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/23/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
|
60
|
Lin R, Wang Z, Cao J, Gao T, Dong Y, Chen Y. Role of melatonin in intestinal mucosal injury induced by restraint stress in mice. PHARMACEUTICAL BIOLOGY 2020; 58:342-351. [PMID: 32298156 PMCID: PMC7178821 DOI: 10.1080/13880209.2020.1750659] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/17/2020] [Accepted: 03/28/2020] [Indexed: 06/11/2023]
Abstract
Context: A growing body of evidence demonstrates that gastrointestinal motility disorder (GIMD) and gastric stress ulcers can be induced by restraint stress, while melatonin (MT) elicits anti-inflammation and antioxidant effects.Objective: The present study investigated the mechanisms of MT-mediated protection effects on restraint stress-induced GIMD.Materials and methods: 144 8-week-old male ICR mice were divided into four groups: control, restraint stress, restraint stress + MT and MT (positive control). 20 mg/kg MT or vehicle were intraperitoneally injected 60 min before restraint stress (10 h/day) once daily for 3 days. Biochemical parameters, intestinal mucosal integrity, tissues antioxidant ability and autophagic proteins levels were determined.Results: Mice subjected to restraint stress elevated NE level by 141.41% and decreased MT content by 38.82% in plasma. Consistent with the decrease in MT level, we observed a reduction in the antioxidant ability and an increase in autophagic proteins by 14.29-46.74% in the gut, resulting in injury to intestinal mucosa which was manifested by reductions in villus height and villus height/crypt depth (V/C) ratio, number of goblet and PCAN-positive cells, and expression of tight junction protein (ZO-1, occludin and claudin-1). In contrast, MT reversed these changes caused by restraint stress and improved the intestinal mucosal injury. However, there was no significant difference between MT (positive control) and control group.Discussion and conclusion: Our results suggest that MT effectively mitigates psychological stress-induced injury to intestinal mucosa, providing evidence demonstrating the potential for using MT as therapy against intestinal impairment associated with psychological stress.
Collapse
Affiliation(s)
- Rutao Lin
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Haidian, China
| | - Zixu Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Haidian, China
| | - Jing Cao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Haidian, China
| | - Ting Gao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Haidian, China
| | - Yulan Dong
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Haidian, China
| | - Yaoxing Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Haidian, China
| |
Collapse
|
61
|
Nonlethal Plasmodium yoelii Infection Drives Complex Patterns of Th2-Type Host Immunity and Mast Cell-Dependent Bacteremia. Infect Immun 2020; 88:IAI.00427-20. [PMID: 32958528 PMCID: PMC7671899 DOI: 10.1128/iai.00427-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022] Open
Abstract
Malaria strongly predisposes to bacteremia, which is associated with sequestration of parasitized red blood cells and increased gastrointestinal permeability. The mechanisms underlying this disruption are poorly understood. Here, we evaluated the expression of factors associated with mast cell activation and malaria-associated bacteremia in a rodent model. C57BL/6J mice were infected with Plasmodium yoeliiyoelli 17XNL, and blood and tissues were collected over time to assay for circulating levels of bacterial 16S DNA, IgE, mast cell protease 1 (Mcpt-1) and Mcpt-4, Th1 and Th2 cytokines, and patterns of ileal mastocytosis and intestinal permeability. Malaria strongly predisposes to bacteremia, which is associated with sequestration of parasitized red blood cells and increased gastrointestinal permeability. The mechanisms underlying this disruption are poorly understood. Here, we evaluated the expression of factors associated with mast cell activation and malaria-associated bacteremia in a rodent model. C57BL/6J mice were infected with Plasmodium yoeliiyoelli 17XNL, and blood and tissues were collected over time to assay for circulating levels of bacterial 16S DNA, IgE, mast cell protease 1 (Mcpt-1) and Mcpt-4, Th1 and Th2 cytokines, and patterns of ileal mastocytosis and intestinal permeability. The anti-inflammatory cytokines (interleukin-4 [IL-4], IL-6, and IL-10) and MCP-1/CCL2 were detected early after P. yoeliiyoelii 17XNL infection. This was followed by the appearance of IL-9 and IL-13, cytokines known for their roles in mast cell activation and growth-enhancing activity as well as IgE production. Later increases in circulating IgE, which can induce mast cell degranulation, as well as Mcpt-1 and Mcpt-4, were observed concurrently with bacteremia and increased intestinal permeability. These results suggest that P. yoeliiyoelii 17XNL infection induces the production of early cytokines that activate mast cells and drive IgE production, followed by elevated IgE, IL-9, and IL-13 that maintain and enhance mast cell activation while disrupting the protease/antiprotease balance in the intestine, contributing to epithelial damage and increased permeability.
Collapse
|
62
|
Hidalgo-Liberona N, González-Domínguez R, Vegas E, Riso P, Del Bo' C, Bernardi S, Peron G, Guglielmetti S, Gargari G, Kroon PA, Cherubini A, Andrés-Lacueva C. Increased Intestinal Permeability in Older Subjects Impacts the Beneficial Effects of Dietary Polyphenols by Modulating Their Bioavailability. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12476-12484. [PMID: 33084335 DOI: 10.1021/acs.jafc.0c04976] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Polyphenols have great potential in regulating intestinal health and ameliorating pathological conditions related to increased intestinal permeability (IP). However, the efficacy of dietary interventions with these phytochemicals may significantly be influenced by interindividual variability factors affecting their bioavailability and consequent biological activity. In the present study, urine samples collected from older subjects undergoing a crossover intervention trial with polyphenol-rich foods were subjected to metabolomics analysis for investigating the impact of increased IP on the bioavailability of polyphenols. Interestingly, urinary levels of phase II and microbiota-derived metabolites were significantly different between subjects with healthier intestinal barrier integrity and those with increased IP disruption. Our results support that this IP-dependent impaired bioavailability of polyphenols could be attributed to disturbances in the gut microbial metabolism and phase II methylation processes. Furthermore, we also observed that microbiota-derived metabolites could be largely responsible for the biological activity elicited by dietary polyphenols against age-related disrupted IP.
Collapse
Affiliation(s)
- Nicole Hidalgo-Liberona
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Raúl González-Domínguez
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Esteban Vegas
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029 Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, 08028 Barcelona, Spain
| | - Patrizia Riso
- Università degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milano, Italy
| | - Cristian Del Bo'
- Università degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milano, Italy
| | - Stefano Bernardi
- Università degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milano, Italy
| | - Gregorio Peron
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Simone Guglielmetti
- Università degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milano, Italy
| | - Giorgio Gargari
- Università degli Studi di Milano, Department of Food, Environmental and Nutritional Sciences (DeFENS), 20133 Milano, Italy
| | - Paul Antony Kroon
- Quadram Institute Bioscience, Norwich Research Park, NR4 7UQ Norwich, United Kingdom
| | - Antonio Cherubini
- Geriatria, Accettazione Geriatrica e Centro di Ricerca per l'Invecchiamento, IRCCS INRCA, 60127 Ancona, Italy
| | - Cristina Andrés-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- CIBER Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 28029 Barcelona, Spain
| |
Collapse
|
63
|
Zhou J, Shu R, Yu C, Xiong Z, Xiao Q, Li Z, Xie X, Fu Z. Exposure to low concentration of trifluoromethanesulfonic acid induces the disorders of liver lipid metabolism and gut microbiota in mice. CHEMOSPHERE 2020; 258:127255. [PMID: 32554004 DOI: 10.1016/j.chemosphere.2020.127255] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/09/2020] [Accepted: 05/28/2020] [Indexed: 06/11/2023]
Abstract
Trifluoromethanesulfonic acid (TFMS) is the shortest chain perfluorinated compound. Recently, it has been identified as a persistent and mobile organic chemical with a maximum concentration of 1 μg/L in the environment. However, its toxicological mechanism remains unclear. In this study, to evaluate the liver and intestinal toxicity of TFMS in mammals, male mice were orally exposed to 0, 1, 10 and 100 μg/kg for 12 weeks. Our results showed that TFMS exposure reduced the epididymal fat weight in mice, caused the decrease of serum and liver triglyceride (TG) level and the increase of serum low density lipoprotein (LDL) level. Also, we observed the inflammatory cell infiltration in the liver of mice exposed to 10 μg/kg and 100 μg/kg TFMS, which was coupled with the increased mRNA expression levels of inflammatory factors such as COX2, TNF-α, IL-1β in the liver. In addition, the mRNA expression levels of lipid metabolism-related genes (PPAR-α, ACOX, SCD1, PPAR-γ, etc.) were significantly decreased in the liver of mice after exposure to both doses of TFMS. We also found TFMS exposure caused the imbalance of cecal gut microbiota and change of cecal microbiota diversity. KEGG pathway predictions showed that the exposure of 100 μg/kg TFMS changed the synthesis and degradation of ketone bodies, benzoate degradation and several other metabolic pathways. Our findings indicated that TFMS exposure disturbed the liver lipid metabolism possibly via altering the gut microbiota.
Collapse
Affiliation(s)
- Jiafeng Zhou
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Ruonan Shu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Chunan Yu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Ze Xiong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Qingfeng Xiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Zezhi Li
- Department of Neurology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaoxian Xie
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China.
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China.
| |
Collapse
|
64
|
Skinner C, Thompson AJ, Thursz MR, Marchesi JR, Vergis N. Intestinal permeability and bacterial translocation in patients with liver disease, focusing on alcoholic aetiology: methods of assessment and therapeutic intervention. Therap Adv Gastroenterol 2020; 13:1756284820942616. [PMID: 33149761 PMCID: PMC7580143 DOI: 10.1177/1756284820942616] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/25/2020] [Indexed: 02/04/2023] Open
Abstract
Increased bacterial translocation (BT) across the gut barrier due to greater intestinal permeability (IP) is seen across a range of conditions, including alcohol-related liver disease (ArLD). The phenomenon of BT may contribute to both the pathogenesis and the development of complications in ArLD. There are a number of methods available to assess IP and in this review we look at their various advantages and limitations. The knowledge around BT and IP in ArLD is also reviewed, as well as the therapeutic strategies currently in use and in development.
Collapse
Affiliation(s)
- Charlotte Skinner
- Department of Metabolism, Digestion and Reproduction, St Mary’s Hospital Campus, Imperial College London, London, UK
| | - Alex J. Thompson
- Department of Surgery & Cancer, St. Mary’s Hospital Campus, Imperial College London, London, UK
| | - Mark R. Thursz
- Department of Metabolism, Digestion and Reproduction, St Mary’s Hospital Campus, Imperial College London, London, UK
| | - Julian R. Marchesi
- Department of Metabolism, Digestion and Reproduction, St Mary’s Hospital Campus, Imperial College London, London, UK
| | | |
Collapse
|
65
|
Tan HY, Toh YC. What can microfluidics do for human microbiome research? BIOMICROFLUIDICS 2020; 14:051303. [PMID: 33062112 PMCID: PMC7538166 DOI: 10.1063/5.0012185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/17/2020] [Indexed: 05/13/2023]
Abstract
Dysregulation of the human microbiome has been linked to various disease states, which has galvanized the efforts to modulate human health through microbiomes. Currently, human microbiome research is going through several phases to identify the constituent components of the microbiome, associate microbiome changes with physiological and pathological states, understand causative relationships, and finally translate this knowledge into therapeutics and diagnostics. The convergence of microfluidic technologies with molecular and cell profiling, microbiology, and tissue engineering can potentially be applied to these different phases of microbiome research to overcome the existing challenges faced by conventional approaches. The goal of this paper is to discuss and highlight the opportunities of applying different microfluidic technologies to specific areas of microbiome research as well as unique challenges that microfluidics must overcome when working with microbiome-relevant biological materials, e.g., micro-organisms, host tissues, and fluids. We will discuss the applicability of integrated microfluidic systems for processing biological samples for genomic sequencing analyses. For functional analysis of the microbiota, we will cover state-of-the-art microfluidic devices for microbiota cultivation and functional measurements. Finally, we highlight the use of organs-on-chips to model various microbiome-host tissue interactions. We envision that microfluidic technologies may hold great promise in advancing the knowledge on the interplay between microbiome and human health, as well as its eventual translation into microbiome-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Hsih-Yin Tan
- Institute for Health Innovation and Technology, National University of Singapore, Singapore 117599
| | - Yi-Chin Toh
- Author to whom correspondence should be addressed:
| |
Collapse
|
66
|
Wu Y, Zhang X, Han D, Ye H, Tao S, Pi Y, Zhao J, Chen L, Wang J. Short Administration of Combined Prebiotics Improved Microbial Colonization, Gut Barrier, and Growth Performance of Neonatal Piglets. ACS OMEGA 2020; 5:20506-20516. [PMID: 32832803 PMCID: PMC7439367 DOI: 10.1021/acsomega.0c02667] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/23/2020] [Indexed: 05/04/2023]
Abstract
This study was conducted to investigate the effects of short administration with the combination (GMF) of galactooligosaccharides (GOS), milk fat globule membrane (MFGM), and fructooligosaccharides (FOS) on microbiota, intestinal barriers, and growth performance of neonatal piglets. Sixteen newborn piglets were divided into two groups: GMF group and CON group; GMF solution (5 mL) and saline (5 mL) were, respectively, administered to piglets in the GMF group and CON group once a day during the first week after birth. The results showed that GMF administration improved the growth performance of neonatal piglets on day 8 and day 21, coupled with the enriched genus Lactobacillus on day 8 and the increased genera norank_f__Muribaculaceae, Christensenellaceae_R-7_group, Enterococcus, and Romboutsia on day 21. Additionally, GMF administration increased luminal acetate and propionate levels, upregulated the gene expressions of intestinal tight junctions (Occludin, Claudins, and ZO-1), mucins (Mucin-1, Mucin-2, Mucin-4, and Mucin-20), and cytokines (TNF-α, IL-1β, and IL-22) while decreased the plasma diamine oxidase (DAO) level on day 21. The correlation analysis showed a positive relationship between the colonized beneficial microbiota and the modified intestinal barrier genes. In conclusion, the first week administration of GMF facilitated the colonization of beneficial bacteria, promoted intestinal development by enhancing microbiota-associated intestinal barrier functions, and improved the growth performance of the piglets during the whole neonatal period. Our findings provide guidelines for combined prebiotics application in modulating the microbial colonization and intestinal development of the neonates.
Collapse
Affiliation(s)
- Yujun Wu
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Xiangyu Zhang
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Han
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Hao Ye
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Shiyu Tao
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Yu Pi
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| | - Junying Zhao
- National
Engineering Center of Dairy for Early Life Health, Beijing Sanyuan Foods Co. Ltd, Beijing 100163, China
| | - Lijun Chen
- National
Engineering Center of Dairy for Early Life Health, Beijing Sanyuan Foods Co. Ltd, Beijing 100163, China
| | - Junjun Wang
- State
Key Laboratory of Animal Nutrition, College of Animal Science and
Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
67
|
Fredericks J, Senapati S, Wannemuehler MJ. Cytotoxic effects of manganese oxide nanoparticles in combination with microbial components on intestinal epithelial cells. F1000Res 2020. [DOI: 10.12688/f1000research.25238.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background: Manganese oxide has been shown to cause toxicity and is associated with occupational-related disease (e.g., welders). With the goal to improve several biomedical areas, manganese oxide nanoparticles (MnO NP) are being considered for use in drug delivery and magnetic resonance imaging (MRI) to obtain high resolution anatomical images of tumors and gastrointestinal (GI) inflammation. Regardless of whether it is intentional or unintentional ingestion, the GI tract has been shown to be the primary route of entry for metal nanoparticles including MnO NP. However, studies assessing toxicity of MnO NP for intestinal epithelial cells (IECs) are virtually nonexistent. Methods: Given the proximity to the GI lumen, assessing the effects of nanoparticles on IECs in the presence of bacterial components presents a more holistic model of exposure. Therefore, we examined the effects of MnO NP alone and MnO NP in combination with Escherichia coli LF82 bacterial lysate on selected functions of MODE-K cells, a murine intestinal epithelial cell line. Data were analyzed using one-way ANOVA. Differences with p < 0.05 were considered significant. Results: Results showed MnO NP plus E. coli LF82 lysate added to MODE-K cells severely inhibited monolayer scratch wound healing, enhanced the secretion of interleukin 6 (IL-6), and induced mitochondrial dysfunction. Conclusions: Overall, our findings show that toxicity of MnO NP deleteriously affected MODE-K cells and demonstrated the necessity to integrate other environmental factors, such as microbial components and/or inflammatory cytokines, into studies assessing effects of nanoparticles on mucosal epithelia.
Collapse
|
68
|
Son YS, Ki SJ, Thanavel R, Kim JJ, Lee MO, Kim J, Jung CR, Han TS, Cho HS, Ryu CM, Kim SH, Park DS, Son MY. Maturation of human intestinal organoids in vitro facilitates colonization by commensal lactobacilli by reinforcing the mucus layer. FASEB J 2020; 34:9899-9910. [PMID: 32602623 DOI: 10.1096/fj.202000063r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 12/14/2022]
Abstract
Lactobacilli, which are probiotic commensal bacteria that mainly reside in the human small intestine, have attracted attention for their ability to exert health-promoting effects and beneficially modulate host immunity. However, host epithelial-commensal bacterial interactions are still largely unexplored because of limited access to human small intestinal tissues. Recently, we described an in vitro maturation technique for generating adult-like, mature human intestinal organoids (hIOs) from human pluripotent stem cells (hPSCs) that closely resemble the in vivo tissue structure and cellular diversity. Here, we established an in vitro human model to study the response to colonization by commensal bacteria using luminal microinjection into mature hIOs, allowing for the direct examination of epithelial-bacterial interactions. Lactobacillus reuteri and Lactobacillus plantarum were more likely to survive and colonize when microinjected into the lumen of mature hIOs than when injected into immature hIOs, as determined by scanning electron microscopy, colony formation assay, immunofluorescence, and real-time imaging with L plantarum expressing red fluorescent protein. The improved mature hIO-based host epithelium system resulted from enhanced intestinal epithelial integrity via upregulation of mucus secretion and tight junction proteins. Our study indicates that mature hIOs are a physiologically relevant in vitro model system for studying commensal microorganisms.
Collapse
Affiliation(s)
- Ye Seul Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Soo Jin Ki
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Rajangam Thanavel
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jong-Jin Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Mi-Ok Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Janghwan Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Cho-Rok Jung
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Tae-Su Han
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hyun-Soo Cho
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Choong-Min Ryu
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sang-Heon Kim
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Department of Biomedical Engineering, KIST school, UST, Daejeon, Republic of Korea
| | - Doo-Sang Park
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.,Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Republic of Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
69
|
Tributyrin differentially regulates inflammatory markers and modulates goblet cells number along the intestinal tract segments of weaning pigs. Livest Sci 2020. [DOI: 10.1016/j.livsci.2020.103996] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
70
|
Tindall AM, McLimans CJ, Petersen KS, Kris-Etherton PM, Lamendella R. Walnuts and Vegetable Oils Containing Oleic Acid Differentially Affect the Gut Microbiota and Associations with Cardiovascular Risk Factors: Follow-up of a Randomized, Controlled, Feeding Trial in Adults at Risk for Cardiovascular Disease. J Nutr 2020; 150:806-817. [PMID: 31848609 PMCID: PMC7138683 DOI: 10.1093/jn/nxz289] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/09/2019] [Accepted: 11/04/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND It is unclear whether the favorable effects of walnuts on the gut microbiota are attributable to the fatty acids, including α-linolenic acid (ALA), and/or the bioactive compounds and fiber. OBJECTIVE This study examined between-diet gut bacterial differences in individuals at increased cardiovascular risk following diets that replace SFAs with walnuts or vegetable oils. METHODS Forty-two adults at cardiovascular risk were included in a randomized, crossover, controlled-feeding trial that provided a 2-wk standard Western diet (SWD) run-in and three 6-wk isocaloric study diets: a diet containing whole walnuts (WD; 57-99 g/d walnuts; 2.7% ALA), a fatty acid-matched diet devoid of walnuts (walnut fatty acid-matched diet; WFMD; 2.6% ALA), and a diet replacing ALA with oleic acid without walnuts (oleic acid replaces ALA diet; ORAD; 0.4% ALA). Fecal samples were collected following the run-in and study diets to assess gut microbiota with 16S rRNA sequencing and Qiime2 for amplicon sequence variant picking. RESULTS Subjects had elevated BMI (30 ± 1 kg/m2), blood pressure (121 ± 2/77 ± 1 mmHg), and LDL cholesterol (120 ± 5 mg/dL). Following the WD, Roseburia [relative abundance (RA) = 4.2%, linear discriminant analysis (LDA) = 4], Eubacterium eligensgroup (RA = 1.4%, LDA = 4), LachnospiraceaeUCG001 (RA = 1.2%, LDA = 3.2), Lachnospiraceae UCG004 (RA = 1.0%, LDA = 3), and Leuconostocaceae (RA = 0.03%, LDA = 2.8) were most abundant relative to taxa in the SWD (P ≤ 0.05 for all). The WD was also enriched in Gordonibacter relative to the WFMD. Roseburia (3.6%, LDA = 4) and Eubacterium eligensgroup (RA = 1.5%, LDA = 3.4) were abundant following the WFMD, and Clostridialesvadin BB60group (RA = 0.3%, LDA = 2) and gutmetagenome (RA = 0.2%, LDA = 2) were most abundant following the ORAD relative to the SWD (P ≤ 0.05 for all). Lachnospiraceae were inversely correlated with blood pressure and lipid/lipoprotein measurements following the WD. CONCLUSIONS The results indicate similar enrichment of Roseburia following the WD and WFMD, which could be explained by the fatty acid composition. Gordonibacter enrichment and the inverse association between Lachnospiraceae and cardiovascular risk factors following the WD suggest that the gut microbiota may contribute to the health benefits of walnut consumption in adults at cardiovascular risk. This trial was registered at clinicaltrials.gov as NCT02210767.
Collapse
Affiliation(s)
- Alyssa M Tindall
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | | | - Kristina S Petersen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | | |
Collapse
|
71
|
DuPont HL, Jiang ZD, DuPont AW, Utay NS. Abnormal Intestinal Microbiome in Medical Disorders and Potential Reversibility by Fecal Microbiota Transplantation. Dig Dis Sci 2020; 65:741-756. [PMID: 32008133 DOI: 10.1007/s10620-020-06102-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Reduction in diversity of the intestinal microbiome (dysbiosis) is being identified in many disease states, and studies are showing important biologic contributions of microbiome to health and disease. Fecal microbiota transplantation (FMT) is being evaluated as a way to reverse dysbiosis in diseases and disorders in an attempt to improve health. The published literature was reviewed to determine the value of FMT in the treatment of medical disorders for which clinical trials have recently been conducted. FMT is effective in treating recurrent C. difficile infection in one or two doses, with many healthy donors providing efficacious fecal-derived products. In inflammatory bowel disease (IBD), FMT may lead to remission in approximately one-third of moderate-to-severe illnesses with one study suggesting that more durable FMT responses may be seen when used once medical remissions have been achieved. Donor products differ in their efficacy in treatment of IBD. Combining donor products has been one way to increase the potential value of FMT in treating chronic disorders. FMT is being explored in a variety of clinical settings affecting different organ systems outside CDI, with positive preliminary signals, in treatment of functional constipation, immunotherapy-induced colitis, neurodegenerative disease, as well as prevention of cancer-related disorders like graft versus host disease and decolonization of patients with recurrent urinary tract infection due to antibiotic-resistant bacteria. Currently, intense research is underway to see how the microbiome products like FMT can be harnessed for health benefits.
Collapse
Affiliation(s)
- Herbert L DuPont
- Kelsey Research Foundation, Houston, TX, USA. .,University of Texas School of Public Health, 1200 Pressler St, Houston, TX, 77030, USA. .,University of Texas McGovern Medical School, Houston, USA. .,Baylor College of Medicine, Houston, USA. .,MD Anderson Cancer Center, Houston, USA.
| | - Zhi-Dong Jiang
- University of Texas School of Public Health, 1200 Pressler St, Houston, TX, 77030, USA
| | | | - Netanya S Utay
- Kelsey Research Foundation, Houston, TX, USA.,University of Texas McGovern Medical School, Houston, USA
| |
Collapse
|
72
|
Jiang S, Mohammed AA, Jacobs JA, Cramer TA, Cheng HW. Effect of synbiotics on thyroid hormones, intestinal histomorphology, and heat shock protein 70 expression in broiler chickens reared under cyclic heat stress. Poult Sci 2020; 99:142-150. [PMID: 32416795 PMCID: PMC7587863 DOI: 10.3382/ps/pez571] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 09/15/2019] [Indexed: 01/11/2023] Open
Abstract
This study examined effect of a dietary synbiotic supplement on the concentrations of plasma thyroid hormones, expressions of heat shock protein 70 (HSP70), and intestinal histomorphology in broiler chickens exposed to cyclic heat stress (HS). Three hundred and sixty day old male Ross 708 broiler chicks were randomly distributed among 3 dietary treatments containing a synbiotic (PoultryStar meUS) at 0 (control), 0.5 (0.5×), and 1.0 (1.0×) g/kg. Each treatment contained 8 replicates of 15 birds each housed in floor pens in a temperature and lighting controlled room. Heat stimulation was established from days 15 to 42 at 32°C for 9 h daily. The results indicated that under the HS condition, both synbiotic fed groups had lower liver and hypothalamus HSP70 levels (P < 0.001) compared to control group; however, HSP70 mRNA expression was not different among treatments (P > 0.05). There were no treatment effects on the levels of triiodothyronine (T3) and thyroxine (T4) as well as T3/T4 ratio (P > 0.05). Compared to controls, 1.0× HS broilers had greater villus height in the duodenum (P < 0.01), and greater villus height and villus height:crypt depth ratios in the ileum (P < 0.01). There were no differences among treatments on the measured intestinal parameters in the jejunum (P > 0.05). The results suggest that the synbiotic may ameliorate the negative effects of HS on chicken health as indicated by the changes in the intestinal architecture and the levels of HSP70. Dietary synbiotic supplement could be a feasible nutritive strategy for the poultry industry to improve the health and welfare of chickens when exposed to hot environmental temperature.
Collapse
Affiliation(s)
- S Jiang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - A A Mohammed
- Department of Animal Sciences, Purdue University, 915 West State Street, West Lafayette, IN 47907, USA; Department of Animal Hygiene, Faculty of Veterinary Medicine, Assiut University, Assiut 71526, Egypt
| | - J A Jacobs
- USDA Agricultural Research Service, 125 South Russell Street, West Lafayette, IN 47907, USA
| | - T A Cramer
- Department of Animal and Food Science, Texas Tech University, Lubbock, TX 79409, USA
| | - H W Cheng
- Department of Animal Sciences, Purdue University, 915 West State Street, West Lafayette, IN 47907, USA.
| |
Collapse
|
73
|
DUPONT HERBERTL, JIANG ZHIDONG, DUPONT ANDREWW, UTAY NETANYAS. THE INTESTINAL MICROBIOME IN HUMAN HEALTH AND DISEASE. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2020; 131:178-197. [PMID: 32675857 PMCID: PMC7358474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The Human Microbiome Initiative of NIH, begun in 2007, has opened the door to the power of the intestinal microbiome in health and disease. The 100 trillion gut microbes influence body function through three pathways: (1) via the neural route where 500 million neurons of the enteric nervous system (the body's second brain) connect to the brain and spinal cord, (2) via the immune route where the gut-immune capacity prevents infection and elicits immune response to vaccines, and (3) by the hormonal route wherein biologically active chemicals are released from enteroendocrine cells to control mood and body functions. Through research, the identification of diseases and disorders associated with abnormal microbiome ("dysbiosis") has increased in number with potential for reversibility. Our team has developed an orally administered fecal microbiota transplantation product that is effective in reversing dysbiosis in recurrent Clostridioides difficile (C. difficile) and is being used to reverse abnormal microbiomes in chronic dysbiotic disorders.
Collapse
Affiliation(s)
- HERBERT L. DUPONT
- Correspondence and reprint requests: Herbert L. DuPont, MD, MACP, 1200 Pressler Street, Houston, Texas 77030713-500-9366
| | | | | | | |
Collapse
|
74
|
High-fat diet-induced metabolic syndrome and oxidative stress in obese rats are ameliorated by yogurt supplementation. Sci Rep 2019; 9:20026. [PMID: 31882854 PMCID: PMC6934669 DOI: 10.1038/s41598-019-56538-0] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/13/2019] [Indexed: 12/19/2022] Open
Abstract
The main objective of this experiment was to determine the effects of yogurt supplementation on fat deposition, oxidative stress, inflammation and fibrosis in the liver of rats with high-fat (HF) diet-induced obesity. Male Wistar rats were used in this study and were separated into the following four different groups: the control, control + yogurt, high fat and high fat+ yogurt groups. The high fat groups received a HF diet for eight weeks. A 5% yogurt (w/w) supplement was also provided to rats fed the HF diet. Yogurt supplementation prevented glucose intolerance and normalized liver-specific enzyme activities in the HF diet-fed rats. Yogurt supplementation also significantly reduced the levels of oxidative stress markers in the plasma and liver of HF diet-fed rats. Moreover, inflammatory cell infiltration, collagen deposition and fibrosis in the liver of HF diet-fed rats were also prevented by yogurt supplementation. Furthermore, yogurt supplementation normalized the intestinal lining and brush border in HF diet-fed rats. This study suggests that yogurt supplementation potentially represents an alternative therapy for the prevention of metabolic syndrome in HF diet-fed rats.
Collapse
|
75
|
Barszcz M, Taciak M, Tuśnio A, Święch E, Skomiał J. Dose-dependent effects of two inulin types differing in chain length on the small intestinal morphology, contractility and proinflammatory cytokine gene expression in piglets. Arch Anim Nutr 2019; 74:107-120. [PMID: 31852279 DOI: 10.1080/1745039x.2019.1697140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Inulin is a linear fructose polymer which may affect small intestinal physiology. The effects of dietary level of two inulin types on morphology, contractility and proinflammatory cytokine gene expression in the small intestine of piglets were investigated. Fifty six piglets were divided into seven groups fed diets without inulin addition or with 1%, 2% or 3% of inulin with an average degree of polymerisation of 10 (IN10) or 23 (IN23). All diets were offered from day 10 of life for 40 d. Feeding IN10 diets did not affect villous height to crypt depth ratio in the duodenum, while in the jejunum the 2% IN10 diet increased it as compared to other groups. Jejunal muscle contractions induced by electrical field stimulation were impaired by the 2% and 3% IN10 diets. The ileal expression of interleukin-12p40 was decreased by the 2% IN10 diet. There was no effect of IN23 level on villous height to crypt depth ratio in any segment of the small intestine as well as on jejunal motility. The 2% and 3% IN23 diets decreased the jejunal expression of tumour necrosis factor-α. In conclusion, IN10 is more active in the small intestine than IN23. At the 2% dietary level, it increases absorptive area in the jejunum, but may slightly impair smooth muscle contractions.
Collapse
Affiliation(s)
- Marcin Barszcz
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Marcin Taciak
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Anna Tuśnio
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Ewa Święch
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| | - Jacek Skomiał
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Jabłonna, Poland
| |
Collapse
|
76
|
Lv Z, Hu C, Jiang J, Jin S, Wei Q, Wei X, Yu D, Shi F. Effects of High-Dose Genistein on the Hypothalamic RNA Profile and Intestinal Health of Female Chicks. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:13737-13750. [PMID: 31789024 DOI: 10.1021/acs.jafc.9b05162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Genistein is abundant in animal feed. In this study, the side effects of high-dose genistein on intestinal health and hypothalamic RNA profile were evaluated. Chicks exposed to high-dose genistein by intraperitoneal injection (416 ± 21, 34.5 ± 2.5) and feed supplementation (308 ± 19, 27.2 ± 2.1) both showed a reduced body weight gain and feed intake in comparison with the control group (261 ± 16, 22.7 ± 1.6, P < 0.01). In comparison with the control (22.4 ± 0.5, 33.3 ± 2.4), serum levels of albumin and total protein were decreased after high-dose genistein injection (21.6 ± 0.5, 31.8 ± 1.6) and diet supplementation (20.6 ± 0.9, 29.9 ± 2.5, P < 0.001). Interestingly, the genistein diet presented the chick hypothalamus with downregulated expression of bitter receptors (TAS1R3, P < 0.05). Meanwhile, it upregulated the expressions of TAS2R1 (P < 0.05) and downstream genes (PLCB2 and IP3R3) in the ileum (P < 0.05). Accordingly, high-dose dietary genistein reduced villus height and the abundance of Lactobacillus, along with the increased abundance of pathogenic bacteria in the ileum (P < 0.05). Furthermore, transcriptomic analysis identified 348 differently expressed genes (168 upregulated and 224 downregulated) in the high-dose dietary genistein treated group in comparison with the control (P < 0.05, |log2FoldChange| > 0.585). Therefore, high-dose dietary genistein altered the hypothalamic RNA profile and signal processing. Cluster analysis further revealed that high-dose dietary genistein significantly influenced apoptosis, the immune process, and the whole synthesis of steroid hormones in the hypothalamus (P < 0.05). In conclusion, high-dose dietary genistein altered the hypothalamic RNA profile and intestinal health of female chicks.
Collapse
Affiliation(s)
- Zengpeng Lv
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Chenhui Hu
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Jingle Jiang
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Song Jin
- Changzhou Animal Disease Control Center , Changzhou 213003 , People's Republic of China
| | - Quanwei Wei
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Xihui Wei
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Debing Yu
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| | - Fangxiong Shi
- College of Animal Science and Technology , Nanjing Agricultural University , No. 1 Weigang Road , Nanjing , Jiangsu 210095 , People's Republic of China
| |
Collapse
|
77
|
Iron Transport Tocopheryl Polyethylene Glycol Succinate in Animal Health and Diseases. Molecules 2019; 24:molecules24234289. [PMID: 31775281 PMCID: PMC6930530 DOI: 10.3390/molecules24234289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/18/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022] Open
Abstract
Gut health is the starting place for maintaining the overall health of an animal. Strategies to maintain gut health are, thus, an important part in achieving the goal of improving animal health. A new strategy to do this involves two molecules: the iron transport protein ovotransferrin (IT) and α-tocopheryl polyethylene glycol succinate (TPGS), which result in the novel formulation of ITPGS. These molecules help reduce gut pathogens, while enhancing the absorption and bioavailability of therapeutic drugs, phytomedicines, and nanomedicines. This, in turn, helps to maintain normal health in animals. Maintaining the gastrointestinal tract (GIT) in its normal condition is key for successful absorption and efficacy of any nutrient. A compromised GIT, due to an imbalance (dysbiosis) in the GIT microbiome, can lead to an impaired GI barrier system with impaired absorption and overall health of the animal. The molecules in ITPGS may address the issue of poor absorption by keeping the GI system healthy by maintaining the normal microbiome and improving the absorption of nutrients through multiple mechanisms involving antioxidative, anti-inflammatory, immunomodulatory, and antimicrobial activities. The ITPGS technology can allow the dose of active pharmaceutical or herbal medicine to be significantly reduced in order to attain equal or better efficacy. With complimentary actions between IT and TPGS, ITPGS presents a novel approach to increase the bioavailability of drugs, phytoconstituents, nutrients, and nanomedicines by enhanced transport to the tissues at the site of action, while reducing gut pathogen load. The ITPGS approach appears to be a novel strategy for maintaining the health of animals by manipulation of microbiota.
Collapse
|
78
|
Yin A, Luo Y, Chen W, He M, Deng JH, Zhao N, Cao L, Wang L. FAM96A Protects Mice From Dextran Sulfate Sodium (DSS)-Induced Colitis by Preventing Microbial Dysbiosis. Front Cell Infect Microbiol 2019; 9:381. [PMID: 31803631 PMCID: PMC6876263 DOI: 10.3389/fcimb.2019.00381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/24/2019] [Indexed: 12/19/2022] Open
Abstract
Family with sequence similarity 96 member A (FAM96A) is an evolutionarily conserved intracellular protein that is involved in the maturation of the Fe/S protein, iron regulatory protein 1 (IRP1), and the mitochondria-related apoptosis of gastrointestinal stromal tumor cells. In this study, we used a mouse model of chemically induced colitis to investigate the physiological role of FAM96A in intestinal homeostasis and inflammation. At baseline, colons from Fam96a−/− mice exhibited microbial dysbiosis, dysregulated epithelial cell turnover, an increased number of goblet cells, and disordered tight junctions with functional deficits affecting intestinal permeability. After cohousing, the differences between wild-type and Fam96a−/− colons were abrogated, suggesting that FAM96A affects colonic epithelial cells in a microbiota-dependent manner. Fam96a deficiency in mice resulted in increased susceptibility to dextran sulfate sodium (DSS)-induced colitis. Importantly, the colitogenic activity of Fam96a−/− intestinal microbiota was transferable to wild-type littermate mice via fecal microbial transplantation (FMT), leading to exacerbation of DSS-induced colitis. Taken together, our data indicate that FAM96A helps to maintain colonic homeostasis and protect against DSS-induced colitis by preventing gut microbial dysbiosis. This study used gene knockout animals to help to understand the in vivo effects of the Fam96a gene for the first time and provides new evidence regarding host–microbiota interactions.
Collapse
Affiliation(s)
- Ang Yin
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Yang Luo
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Wei Chen
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Minwei He
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Jin Hai Deng
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Ning Zhao
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Lulu Cao
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| | - Lu Wang
- Department of Immunology, Center for Human Disease Genomics, Health Science Center, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Medical Immunology, School of Basic Medical Science, Peking University, Ministry of Health, Beijing, China
| |
Collapse
|
79
|
Baritaki S, de Bree E, Chatzaki E, Pothoulakis C. Chronic Stress, Inflammation, and Colon Cancer: A CRH System-Driven Molecular Crosstalk. J Clin Med 2019; 8:E1669. [PMID: 31614860 PMCID: PMC6833069 DOI: 10.3390/jcm8101669] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic stress is thought to be involved in the occurrence and progression of multiple diseases, via mechanisms that still remain largely unknown. Interestingly, key regulators of the stress response, such as members of the corticotropin-releasing-hormone (CRH) family of neuropeptides and receptors, are now known to be implicated in the regulation of chronic inflammation, one of the predisposing factors for oncogenesis and disease progression. However, an interrelationship between stress, inflammation, and malignancy, at least at the molecular level, still remains unclear. Here, we attempt to summarize the current knowledge that supports the inseparable link between chronic stress, inflammation, and colorectal cancer (CRC), by modulation of a cascade of molecular signaling pathways, which are under the regulation of CRH-family members expressed in the brain and periphery. The understanding of the molecular basis of the link among these processes may provide a step forward towards personalized medicine in terms of CRC diagnosis, prognosis and therapeutic targeting.
Collapse
Affiliation(s)
- Stavroula Baritaki
- Division of Surgery, School of Medicine, University of Crete, Heraklion, 71500 Crete, Greece.
| | - Eelco de Bree
- Division of Surgery, School of Medicine, University of Crete, Heraklion, 71500 Crete, Greece.
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece.
| | - Charalabos Pothoulakis
- IBD Center, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 10833, USA.
| |
Collapse
|
80
|
Adenovirus infection is associated with altered gut microbial communities in a non-human primate. Sci Rep 2019; 9:13410. [PMID: 31527752 PMCID: PMC6746978 DOI: 10.1038/s41598-019-49829-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
Adenovirus (AdV) infections are one of the main causes of diarrhea in young children. Enteric AdVs probably disrupt gut microbial defences, which can result in diarrhea. To understand the role of the gut microbiome in AdV-induced pathologies, we investigated the gut microbiome of a naturally AdV-infected non-human primate species, the Malagasy mouse lemur (Microcebus griseorufus), which represents an important model in understanding the evolution of diseases. We observed that AdV infection is associated with disruption of the gut microbial community composition. In AdV+ lemurs, several commensal taxa essential for a healthy gut microbiome decreased, whereas genera containing potential pathogens, such as Neisseria, increased in abundance. Microbial co-occurrence networks revealed a loss of important microbial community interactions in AdV+ lemurs and an overrepresentation of Prevotellaceae. The observation of enteric virus-associated loss of commensal bacteria and associated shifts towards pathobionts may represent the missing link for a better understanding of AdV-induced effects in humans, and also for their potential as drivers of co-infections, an area of research that has been largely neglected so far.
Collapse
|
81
|
Duan Q, Xia P, Nandre R, Zhang W, Zhu G. Review of Newly Identified Functions Associated With the Heat-Labile Toxin of Enterotoxigenic Escherichia coli. Front Cell Infect Microbiol 2019; 9:292. [PMID: 31456954 PMCID: PMC6700299 DOI: 10.3389/fcimb.2019.00292] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Heat-labile toxin (LT) is a well-characterized powerful enterotoxin produced by enterotoxigenic Escherichia coli (ETEC). This toxin is known to contribute to diarrhea in young children in developing countries, international travelers, as well as many different species of young animals. Interestingly, it has also been revealed that LT is involved in other activities in addition to its role in enterotoxicity. Recent studies have indicated that LT toxin enhances enteric pathogen adherence and subsequent intestinal colonization. LT has also been shown to act as a powerful adjuvant capable of upregulating vaccine antigenicity; it also serves as a protein or antigenic peptide display platform for new vaccine development, and can be used as a naturally derived cell targeting and protein delivery tool. This review summarizes the epidemiology, secretion, delivery, and mechanisms of action of LT, while also highlighting new functions revealed by recent studies.
Collapse
Affiliation(s)
- Qiangde Duan
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Pengpeng Xia
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Rahul Nandre
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Guoqiang Zhu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
82
|
Yuan Q, Jiang Y, Fan Y, Ma Y, Lei H, Su J. Fumonisin B 1 Induces Oxidative Stress and Breaks Barrier Functions in Pig Iliac Endothelium Cells. Toxins (Basel) 2019; 11:toxins11070387. [PMID: 31269688 PMCID: PMC6669581 DOI: 10.3390/toxins11070387] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/26/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
Fumonisins (Fums) are types of mycotoxin that widely contaminante feed material crops, and can trigger potential biological toxicities to humans and various animals. However, the toxicity of Fums on porcine blood vessels has not been fully explored. Fumonisin B1 (FB1) is the main component of Fums. Therefore, the aim of this study was to explore the effects of FB1 on the oxidative stress and tight junctions of the pig iliac endothelial cells (PIECs) in vitro. The results showed that FB1 reduced the viability of PIECs, increased the contents of lipid peroxidation product malondialdehyde (MDA), decreased the activities of antioxidant enzymes superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT) and thioredoxin reductase (TrxR), and decreased the level of glutathione (GSH). In addition, the barrier functions were destroyed, along with the down-regulations on Claudin 1, Occludin and ZO-1 and the increase of paracellular permeability. Thus, this research indicates that FB1 facilitates oxidative stress and breaks barrier functions to damage pig iliac endothelium cells.
Collapse
Affiliation(s)
- Qiaoling Yuan
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yancheng Jiang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Ying Fan
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yingfeng Ma
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Hongyu Lei
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Jianming Su
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
83
|
Nay K, Jollet M, Goustard B, Baati N, Vernus B, Pontones M, Lefeuvre-Orfila L, Bendavid C, Rué O, Mariadassou M, Bonnieu A, Ollendorff V, Lepage P, Derbré F, Koechlin-Ramonatxo C. Gut bacteria are critical for optimal muscle function: a potential link with glucose homeostasis. Am J Physiol Endocrinol Metab 2019; 317:E158-E171. [PMID: 31039010 DOI: 10.1152/ajpendo.00521.2018] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Gut microbiota is involved in the development of several chronic diseases, including diabetes, obesity, and cancer, through its interactions with the host organs. It has been suggested that the cross talk between gut microbiota and skeletal muscle plays a role in different pathological conditions, such as intestinal chronic inflammation and cachexia. However, it remains unclear whether gut microbiota directly influences skeletal muscle function. In this work, we studied the impact of gut microbiota modulation on mice skeletal muscle function and investigated the underlying mechanisms. We determined the consequences of gut microbiota depletion after treatment with a mixture of a broad spectrum of antibiotics for 21 days and after 10 days of natural reseeding. We found that, in gut microbiota-depleted mice, running endurance was decreased, as well as the extensor digitorum longus muscle fatigue index in an ex vivo contractile test. Importantly, the muscle endurance capacity was efficiently normalized by natural reseeding. These endurance changes were not related to variation in muscle mass, fiber typology, or mitochondrial function. However, several pertinent glucose metabolism markers, such as ileum gene expression of short fatty acid chain and glucose transporters G protein-coupled receptor 41 and sodium-glucose cotransporter 1 and muscle glycogen level, paralleled the muscle endurance changes observed after treatment with antibiotics for 21 days and reseeding. Because glycogen is a key energetic substrate for prolonged exercise, modulating its muscle availability via gut microbiota represents one potent mechanism that can contribute to the gut microbiota-skeletal muscle axis. Taken together, our results strongly support the hypothesis that gut bacteria are required for host optimal skeletal muscle function.
Collapse
Affiliation(s)
- Kevin Nay
- DMEM, University of Montpellier, INRA, Montpellier , France
- Laboratory "Movement Sport and Health Sciences" EA7470, University of Rennes/ENS Rennes , France
| | - Maxence Jollet
- DMEM, University of Montpellier, INRA, Montpellier , France
| | | | - Narjes Baati
- DMEM, University of Montpellier, INRA, Montpellier , France
| | - Barbara Vernus
- DMEM, University of Montpellier, INRA, Montpellier , France
| | - Maria Pontones
- DMEM, University of Montpellier, INRA, Montpellier , France
| | - Luz Lefeuvre-Orfila
- Laboratory "Movement Sport and Health Sciences" EA7470, University of Rennes/ENS Rennes , France
| | - Claude Bendavid
- Institut NuMeCan, Inserm U1241/CHU Rennes/INRA, Université de Rennes , Rennes , France
| | - Olivier Rué
- MaIAGE, INRA, Université Paris-Saclay , Jouy-en-Josas , France
| | | | - Anne Bonnieu
- DMEM, University of Montpellier, INRA, Montpellier , France
| | | | - Patricia Lepage
- MICALIS, AgroParisTech, INRA, Université Paris-Saclay , Jouy-en-Josas , France
| | - Frédéric Derbré
- Laboratory "Movement Sport and Health Sciences" EA7470, University of Rennes/ENS Rennes , France
| | | |
Collapse
|
84
|
Vissenaekens H, Grootaert C, Rajkovic A, Van De Wiele T, Calatayud M. The response of five intestinal cell lines to anoxic conditionsin vitro. Biol Cell 2019; 111:232-244. [DOI: 10.1111/boc.201800076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/22/2019] [Accepted: 05/19/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Hanne Vissenaekens
- Department of Food technologySafety and HealthFaculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Charlotte Grootaert
- Department of Food technologySafety and HealthFaculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Andreja Rajkovic
- Department of Food technologySafety and HealthFaculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Tom Van De Wiele
- Center for Microbial Ecology and Technology (CMET)Faculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| | - Marta Calatayud
- Center for Microbial Ecology and Technology (CMET)Faculty of Bioscience EngineeringGhent University Ghent 9000 Belgium
| |
Collapse
|
85
|
Asao K, Hashida N, Ando S, Motooka D, Kurakami H, Nakamura S, Yamashita D, Maruyama K, Kawasaki S, Yamada T, Iida T, Nishida K. Conjunctival dysbiosis in mucosa-associated lymphoid tissue lymphoma. Sci Rep 2019; 9:8424. [PMID: 31182732 PMCID: PMC6557838 DOI: 10.1038/s41598-019-44861-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 03/05/2019] [Indexed: 12/21/2022] Open
Abstract
To investigate the conjunctival microbiota and the association between the development of conjunctival mucosa-associated lymphoid tissue (MALT) lymphoma and dysbiosis, DNA samples were collected from 25 conjunctival MALT lymphoma patients and 25 healthy controls. To compare the microbiota, samples were collected from the following four body locations: conjunctiva, meibomian gland, periocular skin and hand. Extracted DNA was analyzed by 16S rRNA sequences, and libraries were sequenced on an Illumina MiSeq sequencer. The differences in bacteria were characterized by using principal coordinate analysis of metagenomics data, and the differences in bacterial compositions were evaluated by linear discriminant analysis effect size. The conjunctival microbiota of MALT lymphoma patients was compositionally different from that of healthy controls. For the conjunctival MALT lymphoma patients, alterations in the microbial composition were detected, and a remarkable change was detected at the conjunctiva. Detailed analysis showed that a specific population of the microbiota, the genus Delftia, was significantly more abundant in conjunctival MALT lymphoma patients, and the genera Bacteroides and Clostridium were less abundant in the MALT lymphoma patients. A specific microbiota on the ocular surface in conjunctival MALT lymphoma patients was detected, and dysbiosis may play an important role in the pathophysiology of conjunctival MALT lymphoma.
Collapse
Affiliation(s)
- Kazunobu Asao
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Noriyasu Hashida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Satoru Ando
- Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan.,Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Ako, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Research Institute for Microbial Disease, Osaka University, Osaka, Japan
| | - Hiroyuki Kurakami
- Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Shota Nakamura
- Department of Infection Metagenomics, Research Institute for Microbial Disease, Osaka University, Osaka, Japan
| | - Daisuke Yamashita
- Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan.,Ako Research Institute, Otsuka Pharmaceutical Co., Ltd., Ako, Japan
| | - Kazuichi Maruyama
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Satoshi Kawasaki
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Ocular Immunology and Regenerative Medicine, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tomomi Yamada
- Department of Medical Innovation, Osaka University Hospital, Osaka, Japan
| | - Tetsuya Iida
- Department of Infection Metagenomics, Research Institute for Microbial Disease, Osaka University, Osaka, Japan
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
86
|
Maynou G, Chester-Jones H, Bach A, Terré M. Feeding Pasteurized Waste Milk to Preweaned Dairy Calves Changes Fecal and Upper Respiratory Tract Microbiota. Front Vet Sci 2019; 6:159. [PMID: 31245388 PMCID: PMC6562338 DOI: 10.3389/fvets.2019.00159] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/10/2019] [Indexed: 12/25/2022] Open
Abstract
In the present study bacterial communities from both, the gastrointestinal and respiratory tract of pre-weaned dairy calves fed two different milk-feeding programs were characterized using 16S rRNA gene sequencing. Twenty female Holstein calves (38.8 ± 1.40 kg of BW) were fed pasteurized waste milk (pWM) containing residues of various antimicrobials. Twenty additional calves (38.1 ± 1.19 kg of BW) were fed milk replacer (MR) with similar nutrient composition (27.5% crude protein, 32.1% fat) compared to waste milk (28.6% crude protein, 30.0% fat) from day 1 to weaning at day 49 of study. Fecal samples and nasal swabs were collected on day 42 only from calves that were not treated with therapeutic antibiotics throughout the study, which were 8 MR and 10 pWM calves. To assess the impact of the two feeding regimes on the fecal and nasal microbiota, α and β-diversity measures were calculated, and the relative abundance of operational taxonomic units (OTUs) at different taxonomic levels was determined for each sample. In general, Chao1, PD Whole Tree, and Shannon diversity indices were similar for the fecal and nasal bacterial communities of calves regardless of the feeding regime. However, principal coordinate analysis based on unweighted Unifrac distances indicated differences in the structure of bacterial communities of calves fed milk replacer compared with those from calves fed pasteurized waste milk. The relative abundance of the Streptococcaceae family and the genus Histophilus was greater (P < 0.05) in the nasal microbiota of calves fed milk replacer than in those fed pasteurized waste milk. However, the genus Prevotella tended (P = 0.06) to be more relatively abundant in the respiratory tract of calves fed pasteurized waste milk than in those fed milk replacer. Differences in relative abundances of bacterial taxa in gut microbiota were only observed at the phylum level, suggesting that antimicrobial residues present in waste milk have a non-specific influence at a lower taxonomical level.
Collapse
Affiliation(s)
- Georgina Maynou
- Department of Ruminant Production, Institute of Agrifood Research and Technology (IRTA), Caldes de Montbui, Spain
| | - Hugh Chester-Jones
- Department of Animal Science, Southern Research and Outreach Center (SROC), Waseca, MN, United States
| | - Alex Bach
- Department of Ruminant Production, Institute of Agrifood Research and Technology (IRTA), Caldes de Montbui, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Marta Terré
- Department of Ruminant Production, Institute of Agrifood Research and Technology (IRTA), Caldes de Montbui, Spain
| |
Collapse
|
87
|
Neubauer V, Humer E, Mann E, Kröger I, Reisinger N, Wagner M, Zebeli Q, Petri RM. Effects of clay mineral supplementation on particle-associated and epimural microbiota, and gene expression in the rumen of cows fed high-concentrate diet. Anaerobe 2019; 59:38-48. [PMID: 31102775 DOI: 10.1016/j.anaerobe.2019.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/29/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023]
Abstract
The increased concentrate amounts in cow diets may initiate changes in both particle-associated (PaM) and epimural microbiota (EpM) with the potential for promoting the establishment of pathogens. Clay minerals have shown promising potentials in binding harmful microorganisms and metabolites due to their high adsorption capacity. This study evaluated the effects of a clay-mineral based product (CM) on PaM, EpM, fermentation parameters, and epithelial gene expression in cows fed a high-concentrate diet. Eight rumen-cannulated non-lactating Holstein cows received a concentrate mix supplemented with CM or not (CON) in a change-over design with an initial 100% roughage diet phase (RD, 1 week), followed by intermittent 65%-high-concentrate-diet phases (HC1, HC2; 1 and 2 week duration, respectively), interrupted by 1 week roughage only. Rumen samples for short-chain fatty acids, ammonia, and lactate quantification, as well as PaM, and epithelial biopsies for EpM examination and epithelial gene expression were collected via the cannula once during each feeding phase. Phylogenetic distance analysis of Illumina MiSeq sequencing of the 16S rRNA gene region V345 showed a clear clustering of RD microbiota compared to HC in PaM, showing the impact of the high-concentrate diet on the bacterial community. In the EpM this effect was less pronounced, due to higher variability in RD. In the PaM, a decrease (P < 0.01) of community diversity occurred with the onset of HC feeding, while in the EpM there was an increase in diversity (P < 0.05). In the PaM, CM increased the relative abundance of genus Butyrivibrio (P < 0.01), a commensal bacterium of the rumen, which was, with 6.4%, the second most abundant genus. There, the CM supplementation decreased the genera Lactobacillus, Fusobacterium, and Treponema (P = 0.05), which are potentially either lactate producing or opportunistic pathogens. In the EpM, CM decreased the relative abundance of Succiniclasticum genus (P < 0.01), a possible endotoxin producer, and increased bacteria that are associated with a normobiotic rumen, such as Campylobacter (P = 0.06). Barrier function genes were upregulated in HC2 and nutrient transport genes downregulated in HC1 (P < 0.05); however, there was little effect on pro-inflammatory genes at the epithelium. The CM showed a significant decreasing effect on the cellular metabolism genes HMGCS1 (P = 0.04). Our results suggest that CM supplementation can increase the relative abundance of commensal microbiota and decrease bacteria that could negatively impact the rumen milieu and health during high-concentrate feeding.
Collapse
Affiliation(s)
- Viktoria Neubauer
- Institute of Animal Nutrition and Functional Plant Compounds, Austria; Institute for Milk Hygiene, Milk Technology and Food Science, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, 1210, Vienna, Austria; FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety & Innovation, 3430, Tulln, Austria
| | - Elke Humer
- Institute of Animal Nutrition and Functional Plant Compounds, Austria
| | - Evelyne Mann
- Institute for Milk Hygiene, Milk Technology and Food Science, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, 1210, Vienna, Austria
| | - Iris Kröger
- Institute of Animal Nutrition and Functional Plant Compounds, Austria
| | - Nicole Reisinger
- BIOMIN Research Center, BIOMIN Holding GmbH, 3430, Tulln, Austria
| | - Martin Wagner
- Institute for Milk Hygiene, Milk Technology and Food Science, Department for Farm Animals and Veterinary Public Health, University of Veterinary Medicine, 1210, Vienna, Austria; FFoQSI GmbH - Austrian Competence Centre for Feed and Food Quality, Safety & Innovation, 3430, Tulln, Austria
| | - Qendrim Zebeli
- Institute of Animal Nutrition and Functional Plant Compounds, Austria
| | - Renee M Petri
- Institute of Animal Nutrition and Functional Plant Compounds, Austria.
| |
Collapse
|
88
|
Kahlert S, Renner L, Kluess J, Frahm J, Tesch T, Bannert E, Kersten S, Dänicke S, Rothkötter HJ. Effects of deoxynivalenol-feed contamination on circulating LPS in pigs. Innate Immun 2019; 25:168-175. [PMID: 30760085 PMCID: PMC6830939 DOI: 10.1177/1753425919829552] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Low concentration of LPS can be detected in healthy mammals without triggering
systemic inflammation. Here we analysed the influence of the mycotoxin
deoxynivalenol (DON) on very low LPS concentrations and the role of DON in the
physiology of pigs challenged with high artificial LPS dosage mimicking septic
shock. Pigs were fed for 29 d with DON-contaminated (4.59 mg/kg feed) or control
feed. Samples of control animals showed 6.6 ± 13.5 pg/ml LPS in portal and
3.1 ± 7.6 pg/ml LPS in jugular serum samples. In the DON fed group,
3.4 ± 7.2 pg/ml and 0.6 ± 0.8 pg/ml were detected. The differences were
statistically not significant, indicating that DON is not a trigger for enhanced
LPS transfer into the blood circulation. Next, pigs were challenged with 7.5 µg
LPS/kg body mass via portal or jugular route. The application route did not
significantly influence the LPS concentration. We expected higher circulating
LPS concentrations in the presence of DON due to the additional stress of liver
metabolism and reduced liver capacity to remove LPS from circulation. This
scenario is supported by tendency. In summary, we found that DON is unlikely to
influence LPS transfer in the gut; DON likely reduces the capacity for LPS
removal in septic shock conditions.
Collapse
Affiliation(s)
- Stefan Kahlert
- 1 Institute of Anatomy, Otto von Guericke University Magdeburg, Germany
| | - Lydia Renner
- 1 Institute of Anatomy, Otto von Guericke University Magdeburg, Germany
| | - Jeannette Kluess
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Jana Frahm
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Tanja Tesch
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Erik Bannert
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Susanne Kersten
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | - Sven Dänicke
- 2 Institute of Animal Nutrition, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Braunschweig, Germany
| | | |
Collapse
|
89
|
Dong X, Xu Q, Wang C, Zou X, Lu J. Supplemental-coated zinc oxide relieves diarrhoea by decreasing intestinal permeability in weanling pigs. JOURNAL OF APPLIED ANIMAL RESEARCH 2019. [DOI: 10.1080/09712119.2019.1645673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Xinyang Dong
- College of Animal Sciences, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, People’s Republic of China
| | - Qianqian Xu
- College of Animal Sciences, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, People’s Republic of China
| | - Chao Wang
- College of Animal Sciences & Technology, Nanjing Agricultural University, Nanjing, People’s Republic of China
| | - Xiaoting Zou
- College of Animal Sciences, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, People’s Republic of China
| | - Jianjun Lu
- College of Animal Sciences, Zhejiang University, Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Hangzhou, People’s Republic of China
| |
Collapse
|
90
|
Ebersole JL, Orraca L, Novak MJ, Kirakodu S, Gonzalez-Martinez J, Gonzalez OA. Comparative Analysis of Gene Expression Patterns for Oral Epithelium-Related Functions with Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1197:143-163. [PMID: 31732940 DOI: 10.1007/978-3-030-28524-1_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epithelial cells and functions of the epithelium are critical to the health of the oral cavity. We used a nonhuman primate model to profile the transcriptome of gingival tissues in health across the lifespan and hypothesized that in older animals, epithelial-related transcriptome patterns would reflect epithelial cells that are aggressively responsive to the surrounding environment and less able to modulate and resolve the noxious challenge from the bacteria. Rhesus monkeys (n = 34) with a healthy periodontium were distributed into four groups: ≤3 years (young), 3-7 years (adolescent), 12-16 years (adult), and 18-23 years (aged), and a buccal gingival sample from the premolar/molar region of each animal was obtained. RNA was subjected to a microarray analysis (GeneChip® Rhesus Macaque Genome Array, Affymetrix), and 336 genes examined that are linked to epithelium and epithelial cell functions categorized into 9 broad functional groups: extracellular matrix and cell structure; extracellular matrix remodeling enzymes; cell adhesion molecules, cytoskeleton regulation; inflammatory response; growth factors; kinases/cell signaling; cell surface receptors; junction associated molecules; autophagy/apoptosis; antimicrobial peptides; and transcription factors. Total of 255 genes displayed a normalized signal >100, and differences across the age groups were observed primarily in extracellular matrix and cell structure, cell adhesion molecules, and cell surface receptor gene categories with elevations in the aged tissues. Keratins 2, 5, 6B, 13, 16, 17 were all significantly increased in healthy-aged tissues versus adults, and keratins 1 and 2 were significantly decreased in young animals. Approximately 15 integrins are highly expressed in the gingival tissues across the age groups with only ITGA8, ITGAM (CD11b), and ITGB2 significantly increased in the aged tissues. Little impact of aging on desmosomal/hemidesmosomal genes was noted. These results suggest that healthy gingival aging has a relatively limited impact on the broader functions of the epithelium and epithelial cells, with some effects on genes for extracellular matrix and cell adhesion molecules (e.g., integrins). Thus, while there is a substantial impact of aging on immune system targets even in healthy gingiva, it appears that the epithelial barrier remains reasonably molecularly intact in this model system.
Collapse
Affiliation(s)
- J L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, USA.
| | - L Orraca
- School of Dental Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - M J Novak
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA.,Division of Periodontology, University College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - S Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA.,Division of Periodontology, University College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - J Gonzalez-Martinez
- Caribbean Primate Research Center, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - O A Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA.,Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
91
|
Yu LCH. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J Biomed Sci 2018; 25:79. [PMID: 30413188 PMCID: PMC6234774 DOI: 10.1186/s12929-018-0483-8] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a multifactorial disease which arises as a result of the interaction of genetic, environmental, barrier and microbial factors leading to chronic inflammation in the intestine. Patients with IBD had a higher risk of developing colorectal carcinoma (CRC), of which the subset was classified as colitis-associated cancers. Genetic polymorphism of innate immune receptors had long been considered a major risk factor for IBD, and the mutations were also recently observed in CRC. Altered microbial composition (termed microbiota dybiosis) and dysfunctional gut barrier manifested by epithelial hyperpermeability and high amount of mucosa-associated bacteria were observed in IBD and CRC patients. The findings suggested that aberrant immune responses to penetrating commensal microbes may play key roles in fueling disease progression. Accumulative evidence demonstrated that mucosa-associated bacteria harbored colitogenic and protumoral properties in experimental models, supporting an active role of bacteria as pathobionts (commensal-derived opportunistic pathogens). Nevertheless, the host factors involved in bacterial dysbiosis and conversion mechanisms from lumen-dwelling commensals to mucosal pathobionts remain unclear. Based on the observation of gut leakiness in patients and the evidence of epithelial hyperpermeability prior to the onset of mucosal histopathology in colitic animals, it was postulated that the epithelial barrier dysfunction associated with mucosal enrichment of specific bacterial strains may predispose the shift to disease-associated microbiota. The speculation of leaky gut as an initiating factor for microbiota dysbiosis that eventually led to pathological consequences was proposed as the "common ground hypothesis", which will be highlighted in this review. Overall, the understanding of the core interplay between gut microbiota and epithelial barriers at early subclinical phases will shed light to novel therapeutic strategies to manage chronic inflammatory disorders and colitis-associated cancers.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Suite 1020, #1 Jen-Ai Rd. Sec. 1, Taipei, 100, Taiwan, Republic of China.
| |
Collapse
|
92
|
Mustfa SA, Singh M, Suhail A, Mohapatra G, Verma S, Chakravorty D, Rana S, Rampal R, Dhar A, Saha S, Ahuja V, Srikanth CV. SUMOylation pathway alteration coupled with downregulation of SUMO E2 enzyme at mucosal epithelium modulates inflammation in inflammatory bowel disease. Open Biol 2018; 7:rsob.170024. [PMID: 28659381 PMCID: PMC5493774 DOI: 10.1098/rsob.170024] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/25/2017] [Indexed: 02/06/2023] Open
Abstract
Post-translational modification pathways such as SUMOylation are integral to all cellular processes and tissue homeostasis. We investigated the possible involvement of SUMOylation in the epithelial signalling in Crohn's disease (CD) and ulcerative colitis (UC), the two major forms of inflammatory bowel disease (IBD). Initially in a murine model of IBD, induced by dextran–sulfate–sodium (DSS mice), we observed inflammation accompanied by a lowering of global SUMOylation of colonic epithelium. The observed SUMOylation alteration was due to a decrease in the sole SUMO E2 enzyme (Ubc9). Mass-spectrometric analysis revealed the existence of a distinct SUMOylome (SUMO-conjugated proteome) in DSS mice with alteration of key cellular regulators, including master kinase Akt1. Knocking-down of Ubc9 in epithelial cells resulted in dramatic activation of inflammatory gene expression, a phenomenon that acted via reduction in Akt1 and its SUMOylated form. Importantly, a strong decrease in Ubc9 and Akt1 was also seen in endoscopic biopsy samples (N = 66) of human CD and UC patients. Furthermore, patients with maximum disease indices were always accompanied by severely lowered Ubc9 or SUMOylated-Akt1. Mucosal tissues with severely compromised Ubc9 function displayed higher levels of pro-inflammatory cytokines and compromised wound-healing markers. Thus, our results reveal an important and previously undescribed role for the SUMOylation pathway involving Ubc9 and Akt1 in modulation of epithelial inflammatory signalling in IBD.
Collapse
Affiliation(s)
- Salman Ahmad Mustfa
- Laboratory of gut inflammation and infection biology (LGIIB), Regional Centre for Biotechnology, 3rd milestone Gurgaon Faridabad Expressway, Faridabad, India.,Department of Gastroenterology, Manipal University, Manipal, Karnataka, India
| | - Mukesh Singh
- Laboratory of gut inflammation and infection biology (LGIIB), Regional Centre for Biotechnology, 3rd milestone Gurgaon Faridabad Expressway, Faridabad, India
| | - Aamir Suhail
- Laboratory of gut inflammation and infection biology (LGIIB), Regional Centre for Biotechnology, 3rd milestone Gurgaon Faridabad Expressway, Faridabad, India.,Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, India
| | - Gayatree Mohapatra
- Laboratory of gut inflammation and infection biology (LGIIB), Regional Centre for Biotechnology, 3rd milestone Gurgaon Faridabad Expressway, Faridabad, India.,Department of Gastroenterology, Manipal University, Manipal, Karnataka, India
| | - Smriti Verma
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Charlestown, Boston, MA, USA
| | - Debangana Chakravorty
- Functional interactomics laboratory, Bose Institute Kolkata, P 1/12, C.I.T Road, Scheme VII M, Kolkata 700054, India
| | - Sarika Rana
- Laboratory of gut inflammation and infection biology (LGIIB), Regional Centre for Biotechnology, 3rd milestone Gurgaon Faridabad Expressway, Faridabad, India.,Department of Gastroenterology, Manipal University, Manipal, Karnataka, India
| | - Ritika Rampal
- All India Institute of Medical Sciences, Ansari Nagar East, New Delhi, India
| | - Atika Dhar
- National Institute of Immunology, New Delhi, India
| | - Sudipto Saha
- Functional interactomics laboratory, Bose Institute Kolkata, P 1/12, C.I.T Road, Scheme VII M, Kolkata 700054, India
| | - Vineet Ahuja
- All India Institute of Medical Sciences, Ansari Nagar East, New Delhi, India
| | - C V Srikanth
- Laboratory of gut inflammation and infection biology (LGIIB), Regional Centre for Biotechnology, 3rd milestone Gurgaon Faridabad Expressway, Faridabad, India
| |
Collapse
|
93
|
Orlando A, Linsalata M, Bianco G, Notarnicola M, D'Attoma B, Scavo MP, Tafaro A, Russo F. Lactobacillus rhamnosus GG Protects the Epithelial Barrier of Wistar Rats from the Pepsin-Trypsin-Digested Gliadin (PTG)-Induced Enteropathy. Nutrients 2018; 10:nu10111698. [PMID: 30405050 PMCID: PMC6265991 DOI: 10.3390/nu10111698] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/25/2018] [Accepted: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CD) is a chronic immune-mediated disorder, characterized by enhanced paracellular permeability across the intestinal epithelium. The complex system of intercellular junctions, including tight junctions (TJs) and adherens junctions (AJs), seals together the epithelial cells to form a continuous layer. The improvements in barrier integrity have been related to modifications in intercellular junction protein expression. Polyamines (spermidine, spermine, and putrescine) actively participate in the modulation of the AJ expression. Both in vitro and in vivo studies have demonstrated that also probiotics can promote the integrity and the function of the intestinal barrier. On these bases, the present work investigated the protective effects exerted by Lactobacillus rhamnosus GG (L.GG) against the pepsin-trypsin-digested gliadin (PTG)-induced enteropathy in jejunal tissue samples of Wistar rats. In particular, the probiotic effects have been evaluated on the intestinal mucosal architecture, polyamine metabolism and intercellular junction protein expression (ZO-1, Occludin, Claudin-1, β-catenin and E-cadherin). The results from this study indicate that L.GG protects the intestinal mucosa of rats from PTG-induced damage, by preventing the reduction of the expression of the intercellular junction proteins. Consequently, a role for L.GG in the therapeutic management of the gluten-related disorders in humans could be hypothesized.
Collapse
Affiliation(s)
- Antonella Orlando
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Michele Linsalata
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Giusy Bianco
- Animal Facility, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Maria Notarnicola
- Laboratory of Nutritional Biochemistry, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Benedetta D'Attoma
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Maria Principia Scavo
- National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Angela Tafaro
- Animal Facility, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| | - Francesco Russo
- Laboratory of Nutritional Pathophysiology, National Institute of Gastroenterology "S. de Bellis" Research Hospital, 70013 Castellana Grotte (Ba), Italy.
| |
Collapse
|
94
|
Akagi K, Wilson KA, Katewa SD, Ortega M, Simons J, Hilsabeck TA, Kapuria S, Sharma A, Jasper H, Kapahi P. Dietary restriction improves intestinal cellular fitness to enhance gut barrier function and lifespan in D. melanogaster. PLoS Genet 2018; 14:e1007777. [PMID: 30383748 PMCID: PMC6233930 DOI: 10.1371/journal.pgen.1007777] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/13/2018] [Accepted: 10/18/2018] [Indexed: 12/12/2022] Open
Abstract
Loss of gut integrity is linked to various human diseases including inflammatory bowel disease. However, the mechanisms that lead to loss of barrier function remain poorly understood. Using D. melanogaster, we demonstrate that dietary restriction (DR) slows the age-related decline in intestinal integrity by enhancing enterocyte cellular fitness through up-regulation of dMyc in the intestinal epithelium. Reduction of dMyc in enterocytes induced cell death, which leads to increased gut permeability and reduced lifespan upon DR. Genetic mosaic and epistasis analyses suggest that cell competition, whereby neighboring cells eliminate unfit cells by apoptosis, mediates cell death in enterocytes with reduced levels of dMyc. We observed that enterocyte apoptosis was necessary for the increased gut permeability and shortened lifespan upon loss of dMyc. Furthermore, moderate activation of dMyc in the post-mitotic enteroblasts and enterocytes was sufficient to extend health-span on rich nutrient diets. We propose that dMyc acts as a barometer of enterocyte cell fitness impacting intestinal barrier function in response to changes in diet and age.
Collapse
Affiliation(s)
- Kazutaka Akagi
- Aging Homeostasis Research Project Team, National Center for Geriatrics and Gerontology, Obu, Aichi, Japan
| | - Kenneth A. Wilson
- Buck Institute for Research on Aging, Novato, California, United States of America
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Subhash D. Katewa
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Mauricio Ortega
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Jesse Simons
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Tyler A. Hilsabeck
- Buck Institute for Research on Aging, Novato, California, United States of America
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, United States of America
| | - Subir Kapuria
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Amit Sharma
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Heinrich Jasper
- Buck Institute for Research on Aging, Novato, California, United States of America
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, California, United States of America
| |
Collapse
|
95
|
Wang H, Zhai N, Chen Y, Fu C, Huang K. OTA induces intestinal epithelial barrier dysfunction and tight junction disruption in IPEC-J2 cells through ROS/Ca 2+-mediated MLCK activation. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:106-112. [PMID: 29966834 DOI: 10.1016/j.envpol.2018.06.062] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 05/23/2018] [Accepted: 06/20/2018] [Indexed: 06/08/2023]
Abstract
Ochratoxin A (OTA) is a frequent contaminant of feed and food worldwide. The toxicity of OTA on intestinal barrier was investigated in porcine intestinal epithelial cells (IPEC-J2). We observed that OTA induced intestinal barrier dysfunction as indicated by the reduction in transepithelial electrical resistance (TEER) and elevation in paracellular permeability to 4 kDa dextran. The barrier dysfunction was accompanied with tight junction disruption including a down-regulation in ZO-1 expression and redistribution of Occludin and ZO-1. Moreover, OTA exposure increased reactive oxygen species (ROS) generation, elevated the intracellular calcium level ([Ca2+]c) and activated myosin light chain kinase (MLCK). Simultaneously, NAC, a ROS scavenger, blocked OTA-induced ROS generation, [Ca2+]c elevation, barrier dysfunction and tight junction disruption, suggesting that OTA-induced ROS generation may act as a trigger. Next, we found that OTA-induced MLCK activation was inhibited by BAPTA-AM, the cytosolic Ca2+ chelator, demonstrating that OTA-induced MLCK activation is dependent on [Ca2+]c elevation. Furthermore, inhibition of MLCK with ML-7 or inhibition of [Ca2+]c elevation with BAPTA-AM markedly prevented OTA-induced barrier dysfunction and tight junction disruption. Taken together, our results indicated that OTA induces ROS generation, and then elevates the [Ca2+]c and MLCK activity in turn, which finally induces barrier dysfunction and disrupts tight junction in IPEC-J2 cell monolayers.
Collapse
Affiliation(s)
- Hong Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Nianhui Zhai
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Ying Chen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Chongyang Fu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China
| | - Kehe Huang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China; Institute of Nutritional and Metabolic Disorders in Domestic Animals and Fowls, Nanjing Agricultural University, Nanjing, 210095, Jiangsu Province, China.
| |
Collapse
|
96
|
Stefanaki C, Bacopoulou F, Michos A. The impact of probiotics' administration on glycemic control, body composition, gut microbiome, mitochondria, and other hormonal signals in adolescents with prediabetes - A randomized, controlled trial study protocol. Contemp Clin Trials Commun 2018; 11:55-62. [PMID: 30003169 PMCID: PMC6041374 DOI: 10.1016/j.conctc.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/20/2018] [Accepted: 06/01/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Recent studies have demonstrated that a significant proportion of adolescents exhibit abdominal obesity in early-middle adolescence, and impaired glucose metabolism. Dysregulation of glucose metabolism is aggravated by the existing osteosarcopenia not only in obese but also in overweight youth. Biochemical inflammation, derived from glucose metabolism dysregulation, in combination with increased stress levels lead to the accumulation of reactive oxygen species, also known as ROS, which seem to afflict the integrity of the gastrointestinal wall, gut mucosa, and commensal, intestinal gut microflora. The current scientific protocol aims to assess the administration of probiotics in prediabetic adolescents in relation with their glycemic control, body composition, and intestinal microbiome. METHODS/DESIGN This is a study protocol of a two-armed RCT, that recruits adolescents with prediabetes, who will receive either a 4-month, life-style intervention, or a life-style intervention along with a probiotic supplement. The primary outcome is the differences in gut microbiome synthesis, body composition analysis parameters, and concentrations of hormones, before and after the intervention. DISCUSSION This study aims to halt the progression of obesity and diabetes and aspires to contribute new evidence for upgraded treatment of obesity and diabetes. TRIAL REGISTRATION Australian New Zealand Clinical Trial Registry (ACTRN12615000470594).
Collapse
Affiliation(s)
- Charikleia Stefanaki
- Choremeion Research Laboratory, First Department of Pediatrics, Faculty of Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| | - Flora Bacopoulou
- Choremeion Research Laboratory, First Department of Pediatrics, Faculty of Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, Faculty of Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| | - Athanasios Michos
- Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, First Department of Pediatrics, Faculty of Medicine, National and Kapodistrian University of Athens, “Aghia Sophia” Children's Hospital, Athens, Greece
| |
Collapse
|
97
|
Lee T, Huang Y, Lu Y, Yeh Y, Yu LC. Hypoxia-induced intestinal barrier changes in balloon-assisted enteroscopy. J Physiol 2018; 596:3411-3424. [PMID: 29178568 PMCID: PMC6068115 DOI: 10.1113/jp275277] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/20/2017] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Balloon-assisted enteroscopy (BAE) is an emerging standard procedure by utilizing distensible balloons to facilitate deep endoscopy in the small and large intestine. Sporadic cases of bacteraemia were found after BAE. Balloon distension by BAE caused gut tissue hypoxia. The impact of balloon distension-induced hypoxia on intestinal barriers remains unclear. Murine models of BAE by colonic balloon distension showed that short- and long-term hypoxia evoked opposite effects on epithelial tight junctions (TJs). Short-term hypoxia fortified TJ integrity, whereas long-term hypoxia caused damage to barrier function. Our data showed for the first time the molecular mechanisms and signalling pathways of epithelial barrier fortification and TJ reorganization by short-term hypoxia for the maintenance of gut homeostasis. The findings suggest avoiding prolonged balloon distension during BAE to reduce the risk of hypoxia-induced gut barrier dysfunction. ABSTRACT Balloon-assisted enteroscopy (BAE) is an emerging standard procedure that uses distensible balloons to facilitate deep endoscopy. Intestines are known to harbour an abundant microflora. Whether balloon distension causes perturbation of blood flow and gut barrier dysfunction, and elicits risk of bacterial translocation remains unknown. Our aims were to (1) conduct a prospective study to gather microbiological and molecular evidence of bacterial translocation by BAE in patients, (2) establish a murine model of colonic balloon distension to investigate tissue hypoxia and intestinal barrier, and (3) assess the effect of short- and long-term hypoxia on epithelial permeability using cell lines. Thirteen patients were enrolled for BAE procedures, and blood samples were obtained before and after BAE for paired comparison. Four of the 13 patients (30.8%) had positive bacterial DNA in blood after BAE. Post-BAE endotoxaemia was higher than the pre-BAE level. Nevertheless, no clinical symptom of sepsis or fever was reported. To mimic clinical BAE, mice were subjected to colonic balloon distension. Local tissue hypoxia was observed during balloon inflation, and reoxygenation after deflation. A trend of increased gut permeability was seen after long-term distension, whereas a significant reduction of permeability was observed by short-term distension in the proximal colon. Human colonic epithelial Caco-2 cells exposed to hypoxia for 5-20 min exhibited increased tight junctional assembly, while those exposed to longer hypoxia displayed barrier disruption. In conclusion, sporadic cases of bacteraemia were found after BAE, without septic symptoms. Short-term hypoxia by balloon distension yielded a protective effect whereas long-term hypoxia caused damage to the gut barrier.
Collapse
Affiliation(s)
- Tsung‐Chun Lee
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwanROC
- Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwanROC
| | - Yi‐Chen Huang
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwanROC
| | - Yen‐Zhen Lu
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwanROC
| | - Yu‐Chang Yeh
- Department of AnesthesiologyNational Taiwan University HospitalTaipeiTaiwanROC
| | - Linda Chia‐Hui Yu
- Graduate Institute of PhysiologyNational Taiwan University College of MedicineTaipeiTaiwanROC
| |
Collapse
|
98
|
Hiippala K, Jouhten H, Ronkainen A, Hartikainen A, Kainulainen V, Jalanka J, Satokari R. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients 2018; 10:nu10080988. [PMID: 30060606 PMCID: PMC6116138 DOI: 10.3390/nu10080988] [Citation(s) in RCA: 412] [Impact Index Per Article: 58.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 07/19/2018] [Accepted: 07/27/2018] [Indexed: 02/06/2023] Open
Abstract
The intestinal microbiota, composed of pro- and anti-inflammatory microbes, has an essential role in maintaining gut homeostasis and functionality. An overly hygienic lifestyle, consumption of processed and fiber-poor foods, or antibiotics are major factors modulating the microbiota and possibly leading to longstanding dysbiosis. Dysbiotic microbiota is characterized to have altered composition, reduced diversity and stability, as well as increased levels of lipopolysaccharide-containing, proinflammatory bacteria. Specific commensal species as novel probiotics, so-called next-generation probiotics, could restore the intestinal health by means of attenuating inflammation and strengthening the epithelial barrier. In this review we summarize the latest findings considering the beneficial effects of the promising commensals across all major intestinal phyla. These include the already well-known bifidobacteria, which use extracellular structures or secreted substances to promote intestinal health. Faecalibacterium prausnitzii, Roseburia intestinalis, and Eubacterium hallii metabolize dietary fibers as major short-chain fatty acid producers providing energy sources for enterocytes and achieving anti-inflammatory effects in the gut. Akkermansia muciniphila exerts beneficial action in metabolic diseases and fortifies the barrier function. The health-promoting effects of Bacteroides species are relatively recently discovered with the findings of excreted immunomodulatory molecules. These promising, unconventional probiotics could be a part of biotherapeutic strategies in the future.
Collapse
Affiliation(s)
- Kaisa Hiippala
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Hanne Jouhten
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Aki Ronkainen
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Anna Hartikainen
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Veera Kainulainen
- Pharmacology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Jonna Jalanka
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| | - Reetta Satokari
- Immunobiology Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
99
|
Yu LCH, Wei SC, Ni YH. Impact of microbiota in colorectal carcinogenesis: lessons from experimental models. Intest Res 2018; 16:346-357. [PMID: 30090033 PMCID: PMC6077307 DOI: 10.5217/ir.2018.16.3.346] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
A role of gut microbiota in colorectal cancer (CRC) growth was first suggested in germ-free rats almost 50 years ago, and the existence of disease-associated bacteria (termed pathobionts) had becoming increasingly evident from experimental data of fecal transplantation, and microbial gavage or monoassociation. Altered bacterial compositions in fecal and mucosal specimens were observed in CRC patients compared to healthy subjects. Microbial fluctuations were found at various cancer stages; an increase of bacterial diversity was noted in the adenoma specimens, while a reduction of bacterial richness was documented in CRC samples. The bacterial species enriched in the human cancerous tissues included Escherichia coli, Fusobacterium nucleatum, and enterotoxigenic Bacteroides fragilis. The causal relationship of gut bacteria in tumorigenesis was established by introducing particular bacterial strains in in situ mouse CRC models. Detailed experimental protocols of bacterial gavage and the advantages and caveats of different experimental models are summarized in this review. The microbial genotoxins, enterotoxins, and virulence factors implicated in the mechanisms of bacteria-driven tumorigenesis are described. In conclusion, intestinal microbiota is involved in colon tumorigenesis. Bacteria-targeting intervention would be the next challenge for CRC.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hsuan Ni
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
100
|
Montalban-Arques A, Chaparro M, Gisbert JP, Bernardo D. The Innate Immune System in the Gastrointestinal Tract: Role of Intraepithelial Lymphocytes and Lamina Propria Innate Lymphoid Cells in Intestinal Inflammation. Inflamm Bowel Dis 2018; 24:1649-1659. [PMID: 29788271 DOI: 10.1093/ibd/izy177] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The gastrointestinal tract harbors the largest microbiota load in the human body, hence maintaining a delicate balance between immunity against invading pathogens and tolerance toward commensal. Such immune equilibrium, or intestinal homeostasis, is conducted by a tight regulation and cooperation of the different branches of the immune system, including the innate and the adaptive immune system. However, several factors affect this delicate equilibrium, ultimately leading to gastrointestinal disorders including inflammatory bowel disease. Therefore, here we decided to review the currently available information about innate immunity lymphocyte subsets playing a role in intestinal inflammation. RESULTS Intestinal innate lymphocytes are composed of intraepithelial lymphocytes (IELs) and lamina propria innate lymphoid cells (ILCs). While IELs can be divided into natural or induced, ILCs can be classified into type 1, 2, or 3, resembling, respectively, the properties of TH1, TH2, or TH17 adaptive lymphocytes. Noteworthy, the phenotype and function of both IELs and ILCs are disrupted under inflammatory conditions, where they help to exacerbate intestinal immune responses. CONCLUSIONS The modulation of both IELs and ILCs to control intestinal inflammatory responses represents a major challenge, as they provide tight regulation among the epithelium, the microbiota, and the adaptive immune system. An improved understanding of the innate immunity mechanisms involved in gastrointestinal inflammation would therefore aid in the diagnosis and further treatment of gastrointestinal inflammatory disorders.
Collapse
Affiliation(s)
- A Montalban-Arques
- Servicio de Aparato Digestivo. Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - M Chaparro
- Servicio de Aparato Digestivo. Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Javier P Gisbert
- Servicio de Aparato Digestivo. Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - D Bernardo
- Servicio de Aparato Digestivo. Hospital Universitario de La Princesa e Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| |
Collapse
|