1
|
Krajewski D, Ranjitkar S, Tedeschi C, Perez NM, Jordan N, Mire M, Schneider SS, Mathias CB. IL-10 Neutralization Attenuates Mast Cell Responses in a Murine Model of Experimental Food Allergy. Immunohorizons 2024; 8:431-441. [PMID: 38888412 PMCID: PMC11220741 DOI: 10.4049/immunohorizons.2400002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/29/2024] [Indexed: 06/20/2024] Open
Abstract
IgE-mediated mast cell (MC) activation is a critical component of allergic responses to oral Ags. Several T cell-derived cytokines have been shown to promote MC reactivity, and we recently demonstrated a critical role for the cytokine IL-10 in mediating MC responses during food allergy. In this study, we further validate the role of IL-10 using Ab-mediated IL-10 depletion. IL-10 neutralization significantly attenuated MC responses, leading to decreased MC accumulation and activation, as well as inhibition of MC-mediated symptoms such as allergic diarrhea. This was accompanied by decreased Th2 cytokine gene expression, attenuated systemic T cell responses, and fewer CD4 T cells, B cells, and MCs in the spleen. Our data further confirm the role of IL-10 in driving MC responses and suggest that IL-10-responsive MCs may constitute an important player in allergic responses.
Collapse
Affiliation(s)
- Dylan Krajewski
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, MA
| | - Saurav Ranjitkar
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT
| | - Caitlin Tedeschi
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT
| | | | - Nathan Jordan
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT
| | - Mohamed Mire
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, MA
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA
| | - Clinton B. Mathias
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT
| |
Collapse
|
2
|
Gustafson KL, McAdams ZL, Russell AL, Dorfmeyer RA, Turner GM, Ericsson AC. Effect size of delayed freezing, diurnal variation, and hindgut location on the mouse fecal microbiome. iScience 2024; 27:109090. [PMID: 38361608 PMCID: PMC10867441 DOI: 10.1016/j.isci.2024.109090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/09/2023] [Accepted: 01/29/2024] [Indexed: 02/17/2024] Open
Abstract
Practical considerations in fecal sample collection for microbiome research include time to sample storage, time of collection, and hindgut position during terminal collections. Here, parallel experiments were performed to investigate the relative effect of these factors on microbiome composition in mice colonized with two different vendor-origin microbiomes. 16S rRNA amplicon sequencing of immediately flash-frozen feces showed no difference in alpha or beta diversity compared to samples incubated up to 9 h at room temperature. Samples collected in the morning showed greater alpha diversity compared to samples collected in the afternoon. While a significant effect of time was detected in all hindgut regions, the effect increased from cecum to distal colon. This study highlights common scenarios in microbiome research that may affect outcome measures of microbial community analysis. However, we demonstrate a relatively low effect size of these technical factors when compared to a primary experimental factor with large intergroup variability.
Collapse
Affiliation(s)
- Kevin L. Gustafson
- University of Missouri (MU) Comparative Medicine Program, Columbia, MO 65201, USA
- Department of Veterinary Pathobiology, MU, Columbia, MO 65201, USA
| | - Zachary L. McAdams
- Molecular Pathogenesis and Therapeutics Program, MU, Columbia, MO 65201, USA
| | - Amber L. Russell
- Department of Veterinary Pathobiology, MU, Columbia, MO 65201, USA
| | - Rebecca A. Dorfmeyer
- MU Metagenomics Center (MUMC), Mutant Mouse Resource and Research Center at the University of Missouri (MU MMRRC), Columbia, MO 65201, USA
| | - Giedre M. Turner
- MU Metagenomics Center (MUMC), Mutant Mouse Resource and Research Center at the University of Missouri (MU MMRRC), Columbia, MO 65201, USA
| | - Aaron C. Ericsson
- University of Missouri (MU) Comparative Medicine Program, Columbia, MO 65201, USA
- Department of Veterinary Pathobiology, MU, Columbia, MO 65201, USA
- Molecular Pathogenesis and Therapeutics Program, MU, Columbia, MO 65201, USA
- MU Metagenomics Center (MUMC), Mutant Mouse Resource and Research Center at the University of Missouri (MU MMRRC), Columbia, MO 65201, USA
| |
Collapse
|
3
|
Peppermüller PP, Gehring J, Zentrich E, Bleich A, Häger C, Buettner M. Grimace scale assessment during Citrobacter rodentium inflammation and colitis development in laboratory mice. Front Vet Sci 2023; 10:1173446. [PMID: 37342621 PMCID: PMC10277495 DOI: 10.3389/fvets.2023.1173446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
Introduction Bacterial infections and chronic intestinal inflammations triggered by genetic susceptibility, environment or an imbalance in the intestinal microbiome are usually long-lasting and painful diseases in which the development and maintenance of these various intestinal inflammations is not yet fully understood, research is still needed. This still requires the use of animal models and is subject to the refinement principle of the 3Rs, to minimize suffering or pain perceived by the animals. With regard to this, the present study aimed at the recognition of pain using the mouse grimace scale (MGS) during chronic intestinal colitis due to dextran sodium sulfate (DSS) treatment or after infection with Citrobacter rodentium. Methods In this study 56 animals were included which were divided into 2 experimental groups: 1. chronic intestinal inflammation (n = 9) and 2. acute intestinal inflammation (with (n = 23) and without (n = 24) C. rodentium infection). Before the induction of intestinal inflammation in one of the animal models, mice underwent an abdominal surgery and the live MGS from the cage side and a clinical score were assessed before (bsl) and after 2, 4, 6, 8, 24, and 48 hours. Results The highest clinical score as well as the highest live MGS was detected 2 hours after surgery and almost no sign of pain or severity were detected after 24 and 48 hours. Eight weeks after abdominal surgery B6-Il4/Il10-/- mice were treated with DSS to trigger chronic intestinal colitis. During the acute phase as well as the chronic phase of the experiment, the live MGS and a clinical score were evaluated. The clinical score increased after DSS administration due to weight loss of the animals but no change of the live MGS was observed. In the second C57BL/6J mouse model, after infection with C. rodentium the clinical score increased but again, no increased score values in the live MGS was detectable. Discussion In conclusion, the live MGS detected post-operative pain, but indicated no pain during DSS-induced colitis or C. rodentium infection. In contrast, clinical scoring and here especially the weight loss revealed a decreased wellbeing due to surgery and intestinal inflammation.
Collapse
|
4
|
Grunz EA, Jones BW, Lateef OM, Sen S, Wilkinson K, Joshi T, Boerman EM. Adventitial macrophage accumulation impairs perivascular nerve function in mesenteric arteries with inflammatory bowel disease. Front Physiol 2023; 14:1198066. [PMID: 37342800 PMCID: PMC10278583 DOI: 10.3389/fphys.2023.1198066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Introduction: Inflammatory bowel disease involves aberrant immune responses and is associated with both cardiovascular disease risk and altered intestinal blood flow. However, little is known about how inflammatory bowel disease affects regulation of perivascular nerves that mediate blood flow. Previous work found perivascular nerve function is impaired in mesenteric arteries with Inflammatory bowel disease. The purpose of this study was to determine the mechanism of impaired perivascular nerve function. Methods: RNA sequencing was performed on mesenteric arteries from IL10-/- mice treated with H. hepaticus to induce disease (inflammatory bowel disease) or left non-gavaged (Control). For all other studies, Control and Inflammatory bowel disease mice received either saline or clodronate liposome injections to study the effect of macrophage depletion. Perivascular nerve function was assessed using pressure myography and electrical field stimulation. Leukocyte populations, and perivascular nerves, and adventitial neurotransmitter receptors were labeled using fluorescent immunolabeling. Results: Inflammatory bowel disease was associated with increases in macrophage-associated gene expression, and immunolabeling showed accumulation of adventitial macrophages. Clodronate liposome injection eliminated adventitial macrophages, which reversed significant attenuation of sensory vasodilation, sympathetic vasoconstriction and sensory inhibition of sympathetic constriction in inflammatory bowel disease. Acetylcholine-mediated dilation was impaired in inflammatory bowel disease and restored after macrophage depletion, but sensory dilation remained nitric oxide independent regardless of disease and/or macrophage presence. Conclusion: Altered neuro-immune signaling between macrophages and perivascular nerves in the arterial adventitia contributes to impaired vasodilation, particularly via dilatory sensory nerves. Targeting the adventitial macrophage population may help preserve intestinal blood flow in Inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Elizabeth A. Grunz
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Benjamin W. Jones
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Olubodun Michael Lateef
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Sidharth Sen
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, United States
| | - Katie Wilkinson
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, United States
| | - Trupti Joshi
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, United States
- Department of Health Management and Informatics and Christopher S Bond Life Science Center, University of Missouri, Columbia, MO, United States
| | | |
Collapse
|
5
|
Grunz EA, Jones BW, Sen S, Wilkenson K, Joshi T, Boerman EM. Adventitial macrophage accumulation impairs perivascular nerve function in mesenteric arteries with inflammatory bowel disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.04.535591. [PMID: 37066314 PMCID: PMC10104036 DOI: 10.1101/2023.04.04.535591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Introduction Inflammatory bowel disease (IBD) involves aberrant immune responses and is associated with both cardiovascular disease risk and altered intestinal blood flow. However, little is known about how IBD affects regulation of perivascular nerves that mediate blood flow. Previous work found perivascular nerve function is impaired in mesenteric arteries with IBD. The purpose of this study was to determine the mechanism of impaired perivascular nerve function. Methods RNA sequencing was performed on mesenteric arteries from IL10 -/- mice treated with H.hepaticus to induce disease (IBD) or left non-gavaged (Control). For all other studies, Control and IBD mice received either saline or clodronate liposome injections to study the effect of macrophage depletion. Perivascular nerve function was assessed using pressure myography and electrical field stimulation. Fluorescent immunolabeling was used to label leukocyte populations and perivascular nerves. Results IBD was associated with increased in macrophage-associated gene expression, and immunolabeling showed accumulation of adventitial macrophages. Clodronate liposome injection eliminated adventitial macrophages, which reversed significant attenuation of sensory vasodilation, sympathetic vasoconstriction and sensory inhibition of sympathetic constriction in IBD. Acetylcholine-mediated dilation was impaired in IBD and restored after macrophage depletion, but sensory dilation remained nitric oxide independent regardless of disease and/or macrophage presence. Conclusion Altered neuro-immune signaling between macrophages and perivascular nerves in the arterial adventitia contributes to impaired vasodilation, particularly via dilatory sensory nerves. Targeting the adventitial macrophage population may help preserve intestinal blood flow in IBD patients.
Collapse
|
6
|
Cheatham CN, Gustafson KL, McAdams ZL, Turner GM, Dorfmeyer RA, Ericsson AC. Standardized Complex Gut Microbiomes Influence Fetal Growth, Food Intake, and Adult Body Weight in Outbred Mice. Microorganisms 2023; 11:484. [PMID: 36838449 PMCID: PMC9961083 DOI: 10.3390/microorganisms11020484] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Obesity places a tremendous burden on individual health and the healthcare system. The gut microbiome (GM) influences host metabolism and behaviors affecting body weight (BW) such as feeding. The GM of mice varies between suppliers and significantly influences BW. We sought to determine whether GM-associated differences in BW are associated with differences in intake, fecal energy loss, or fetal growth. Pair-housed mice colonized with a low or high microbial richness GM were weighed, and the total and BW-adjusted intake were measured at weaning and adulthood. Pups were weighed at birth to determine the effects of the maternal microbiome on fetal growth. Fecal samples were collected to assess the fecal energy loss and to characterize differences in the microbiome. The results showed that supplier-origin microbiomes were associated with profound differences in fetal growth and excessive BW-adjusted differences in intake during adulthood, with no detected difference in fecal energy loss. Agreement between the features of the maternal microbiome associated with increased birth weight here and in recent human studies supports the value of this model to investigate the mechanisms by which the maternal microbiome regulates offspring growth and food intake.
Collapse
Affiliation(s)
- Christa N. Cheatham
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri (MU), Columbia, MO 65201, USA
| | - Kevin L. Gustafson
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri (MU), Columbia, MO 65201, USA
| | - Zachary L. McAdams
- Molecular Pathogenesis and Therapeutics Program, University of Missouri (MU), Columbia, MO 65201, USA
| | - Giedre M. Turner
- Mutant Mouse Resource and Research Center, University of Missouri (MU), Columbia, MO 65201, USA
- University of Missouri Metagenomics Center, University of Missouri (MU), Columbia, MO 65201, USA
| | - Rebecca A. Dorfmeyer
- Mutant Mouse Resource and Research Center, University of Missouri (MU), Columbia, MO 65201, USA
- University of Missouri Metagenomics Center, University of Missouri (MU), Columbia, MO 65201, USA
| | - Aaron C. Ericsson
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri (MU), Columbia, MO 65201, USA
- Molecular Pathogenesis and Therapeutics Program, University of Missouri (MU), Columbia, MO 65201, USA
- Mutant Mouse Resource and Research Center, University of Missouri (MU), Columbia, MO 65201, USA
- University of Missouri Metagenomics Center, University of Missouri (MU), Columbia, MO 65201, USA
| |
Collapse
|
7
|
Transfer efficiency and impact on disease phenotype of differing methods of gut microbiota transfer. Sci Rep 2022; 12:19621. [PMID: 36380056 PMCID: PMC9666633 DOI: 10.1038/s41598-022-24014-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
To test causal relationships between complex gut microbiota (GM) and host outcomes, researchers frequently transfer GM between donor and recipient mice via embryo transfer (ET) rederivation, cross-fostering (CF), and co-housing. In this study, we assess the influence of the transfer method and the differences in baseline donor and recipient microbiota richness, on transfer efficiency. Additionally, recipient mice were subjected to DSS-induced chronic colitis to determine whether disease severity was affected by GM transfer efficiency or features within the GM. We found that the recipient's genetic background, the baseline richness of donor and recipient GM, and the transfer method all influenced the GM transfer efficiency. Recipient genetic background and GM both had significant effects on DSS colitis severity and, unexpectedly, the transfer method was strongly associated with differential disease severity regardless of the other factors.
Collapse
|
8
|
Effects of Dietary Macleaya cordata Extract on Growth Performance, Biochemical Indices, and Intestinal Microbiota of Yellow-Feathered Broilers Subjected to Chronic Heat Stress. Animals (Basel) 2022; 12:ani12172197. [PMID: 36077916 PMCID: PMC9454434 DOI: 10.3390/ani12172197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022] Open
Abstract
This study investigated the effect of dietary Macleaya cordata extract (MCE) supplementation on the growth performance, serum parameters, and intestinal microbiota of yellow-feather broilers under heat stress. A total of 216 yellow-feather broilers (28-days-old) were randomly allotted into three groups. A control group (CON) (24 ± 2 °C) and heat stress group (HS) (35 ± 2 °C) received a basal diet, and heat-stressed plus MCE groups (HS-MCE) (35 ± 2 °C) were fed the basal diet with 1000 mg/kg MCE for 14 consecutive days. The results revealed that MCE supplementation improved the final body weight, average daily feed intake, average daily gain, and spleen index when compared with the HS group (p < 0.05). In addition, MCE supplementation decreased (p < 0.05) the activities of aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, and creatinine, and increased (p < 0.05) the glucose level and alkaline phosphatase activity in heat-stressed yellow-feathered broilers. Moreover, MCE treatment alleviated heat-stress-induced intestinal flora disturbances, decreased the Bacteroidota and Bacteroides relative abundances, and increased Firmicutes. A linear discriminant analysis effect size analysis found five differentially abundant taxa in the HS-MCE group, including Alistipes, Rikenellaceae, Mogibacterium, Butyrivibrio, and Lachnospira. These results suggest that MCE can alleviate HS-induced decline in growth performance by modulating blood biochemical markers and cecal flora composition in broilers.
Collapse
|
9
|
Russell A, Copio JN, Shi Y, Kang S, Franklin CL, Ericsson AC. Reduced housing density improves statistical power of murine gut microbiota studies. Cell Rep 2022; 39:110783. [PMID: 35545042 PMCID: PMC9161176 DOI: 10.1016/j.celrep.2022.110783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/28/2021] [Accepted: 04/13/2022] [Indexed: 11/25/2022] Open
Abstract
The gut microbiome of humans and animals is critical to host health. Mice are used to investigate the microbiome and its influences; however, the predictive value of such studies is hindered by cage effects due to coprophagy. Our objectives were to evaluate the influence of cage density on the statistical power to detect treatment-dependent effects of a selective pressure on microbiome composition. C57BL/6 mice were separated into groups of 2 or 4 mice per cage and then assigned to groups receiving enrofloxacin, broad-spectrum antibiotics, or control. Fecal samples were collected at weeks 0, 1, and 4, along with contents of the jejunum and cecum. Bacterial DNA analysis examined microbiome richness, diversity, and variability within and between cages. Statistical analyses reveal that reduced housing density consistently results in comparable susceptibility to antibiotics, reduced cage effects, and increased statistical power to detect treatment-associated effects, justifying the practice of reduced housing density.
Collapse
Affiliation(s)
- Amber Russell
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65201, USA
| | - Joanna N Copio
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Yushu Shi
- Department of Statistics, College of Arts and Sciences, University of Missouri, Columbia, MO 65201 USA
| | - Sumin Kang
- Department of Statistics, College of Arts and Sciences, University of Missouri, Columbia, MO 65201 USA
| | - Craig L Franklin
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65201, USA; University of Missouri Metagenomics Center, University of Missouri, Columbia, MO 65201 USA; Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO 65201 USA
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO 65201, USA; University of Missouri Metagenomics Center, University of Missouri, Columbia, MO 65201 USA; Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO 65201 USA.
| |
Collapse
|
10
|
Spalinger MR, Schwarzfischer M, Niechcial A, Atrott K, Laimbacher A, Gottier C, Lang S, Scharl M. Loss of PTPN22 Promotes Intestinal Inflammation by Compromising Granulocyte-mediated Antibacterial Defence. J Crohns Colitis 2021; 15:2118-2130. [PMID: 34089589 DOI: 10.1093/ecco-jcc/jjab098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS A single nucleotide polymorphism in protein tyrosine phosphatase non-receptor type 22 [PTPN22] has been associated with the onset of autoimmune disorders, but protects from Crohn's disease. PTPN22 deficiency in mice promotes intestinal inflammation by modulating lymphocyte function. However, the impact of myeloid PTPN22 in colitis development remains unclear. The aim of this study was to investigate the role of PTPN2 in the IL-10 and the T cell transfer colitis models. METHODS PTPN22-deficient mice were crossed with IL-10-/- and RAG2-/- mice. Naïve T cells were injected in RAG-/- mice to induce T-cell transfer colitis. Spontaneous colitis in IL-10-/- mice was monitored for up to 200 days. RESULTS Here, we demonstrate that PTPN22 in non-lymphoid immune cells is required to protect against T cell transfer-mediated and IL-10 knock-out colitis. Analysis of the intestinal immune landscape demonstrated a marked reduction of granulocyte influx into the inflamed colon in PTPN22-deficient mice. On a molecular level, granulocytes were not only reduced by numbers, but also revealed a defective function. In particular, granulocyte activation and granulocyte-mediated bacteria killing was impaired upon loss of PTPN22, resulting in elevated bacterial burden and translocation beyond the intestinal epithelial barrier in PTPN22-deficient mice. Consistently, antibiotic-induced depletion of bacteria reverted the increased colitis susceptibility in PTPN22-deficient mice, whereas granulocyte depletion induced acolitis phenotype in wild-type mice similar to that observed in PTPN22-deficient mice. CONCLUSIONS In conclusion, our data demonstrate that PTPN22 is essential for adequate granulocyte activation and antimicrobial defence to protect the inflamed intestine from bacterial invasion and exacerbated colitis.
Collapse
Affiliation(s)
- Marianne R Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Marlene Schwarzfischer
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Anna Niechcial
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Kirstin Atrott
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Andrea Laimbacher
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Claudia Gottier
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Silvia Lang
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Kidess E, Kleerebezem M, Brugman S. Colonizing Microbes, IL-10 and IL-22: Keeping the Peace at the Mucosal Surface. Front Microbiol 2021; 12:729053. [PMID: 34603258 PMCID: PMC8484919 DOI: 10.3389/fmicb.2021.729053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022] Open
Abstract
Our world is filled with microbes. Each multicellular organism has developed ways to interact with this microbial environment. Microbes do not always pose a threat; they can contribute to many processes that benefit the host. Upon colonization both host and microbes adapt resulting in dynamic ecosystems in different host niches. Regulatory processes develop within the host to prevent overt inflammation to beneficial microbes, yet keeping the possibility to respond when pathogens attempt to adhere and invade tissues. This review will focus on microbial colonization and the early (innate) host immune response, with special emphasis on the microbiota-modifying roles of IL-10 and IL-22 in the intestine. IL-10 knock out mice show an altered microbial composition, and spontaneously develop enterocolitis over time. IL-22 knock out mice, although not developing enterocolitis spontaneously, also have an altered microbial composition and increase of epithelial-adherent bacteria, mainly caused by a decrease in mucin and anti-microbial peptide production. Recently interesting links have been found between the IL-10 and IL-22 pathways. While IL-22 can function as a regulatory cytokine at the mucosal surface, it also has inflammatory roles depending on the context. For example, lack of IL-22 in the IL-10–/– mice model prevents spontaneous colitis development. Additionally, the reduced microbial diversity observed in IL-10–/– mice was also reversed in IL-10/IL-22 double mutant mice (Gunasekera et al., 2020). Since in early life, host immunity develops in parallel and in interaction with colonizing microbes, there is a need for future studies that focus on the effect of the timing of colonization in relation to the developmental phase of the host. To illustrate this, examples from zebrafish research will be compared with studies performed in mammals. Since zebrafish develop from eggs and are directly exposed to the outside microbial world, timing of the development of host immunity and subsequent control of microbial composition, is different from mammals that develop in utero and only get exposed after birth. Likewise, colonization studies using adult germfree mice might yield different results from those using neonatal germfree mice. Lastly, special emphasis will be given to the need for host genotype and environmental (co-housing) control of experiments.
Collapse
Affiliation(s)
- Evelien Kidess
- Animal Sciences Group, Host-Microbe Interactomics, Wageningen University and Research, Wageningen, Netherlands
| | - Michiel Kleerebezem
- Animal Sciences Group, Host-Microbe Interactomics, Wageningen University and Research, Wageningen, Netherlands
| | - Sylvia Brugman
- Animal Sciences Group, Host-Microbe Interactomics, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
12
|
Valle M, Mitchell PL, Pilon G, Varin T, Hénault L, Rolin J, McLeod R, Gill T, Richard D, Vohl MC, Jacques H, Gagnon C, Bazinet L, Marette A. Salmon peptides limit obesity-associated metabolic disorders by modulating a gut-liver axis in vitamin D-deficient mice. Obesity (Silver Spring) 2021; 29:1635-1649. [PMID: 34449134 DOI: 10.1002/oby.23244] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE This study investigated the effects of a low-dose salmon peptide fraction (SPF) and vitamin D3 (VitD3 ) in obese and VitD3 -deficient mice at risk of metabolic syndrome (MetS). METHODS Obese and VitD3 -deficient low-density lipoprotein receptor (LDLr)-/- /apolipoprotein B100 (ApoB)100/100 mice were treated with high-fat high-sucrose diets, with 25% of dietary proteins replaced by SPF or a nonfish protein mix (MP). The SPF and MP groups received a VitD3 -deficient diet or a supplementation of 15,000 IU of VitD3 per kilogram of diet. Glucose homeostasis, atherosclerosis, nonalcoholic fatty liver disease, and gut health were assessed. RESULTS VitD3 supplementation increased plasma 25-hydroxyvitamin D to optimal status whereas the VitD3 -deficient diet maintained moderate deficiency. SPF-treated groups spent more energy and accumulated less visceral fat in association with an improved adipokine profile. SPF lowered homeostatic model assessment of insulin resistance compared with MP, suggesting that SPF can improve insulin sensitivity. SPF alone blunted hepatic and colonic inflammation, whereas VitD3 supplementation attenuated ileal inflammation. These effects were associated with changes in gut microbiota such as increased Mogibacterium and Muribaculaceae. CONCLUSIONS SPF treatment improves MetS by modulating hepatic and gut inflammation along with gut microbiota, suggesting that SPF operates through a gut-liver axis. VitD3 supplementation has limited influence on MetS in this model.
Collapse
Affiliation(s)
- Marion Valle
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
| | - Patricia L Mitchell
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
| | - Geneviève Pilon
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
| | - Thibault Varin
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
| | - Loïc Hénault
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
- Department of Food Sciences, Laboratory of Food Processing and ElectroMembrane Processes, Laval University, Québec City, Québec, Canada
| | - Jonathan Rolin
- Process Engineering and Applied Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Roger McLeod
- Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tom Gill
- Process Engineering and Applied Science, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Denis Richard
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
- School of Nutrition, Laval University, Québec, Québec, Canada
| | - Hélène Jacques
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
- School of Nutrition, Laval University, Québec, Québec, Canada
| | - Claudia Gagnon
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
- Endocrinology and Nephrology Unit, CHU de Québec Research Centre, Québec City, Québec, Canada
| | - Laurent Bazinet
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
- Department of Food Sciences, Laboratory of Food Processing and ElectroMembrane Processes, Laval University, Québec City, Québec, Canada
| | - André Marette
- Faculty of Medicine, Québec Heart and Lung Institute, Laval University, Québec City, Québec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec City, Québec, Canada
| |
Collapse
|
13
|
Supplier-origin mouse microbiomes significantly influence locomotor and anxiety-related behavior, body morphology, and metabolism. Commun Biol 2021; 4:716. [PMID: 34112927 PMCID: PMC8192786 DOI: 10.1038/s42003-021-02249-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/20/2021] [Indexed: 12/26/2022] Open
Abstract
The mouse is the most commonly used model species in biomedical research. Just as human physical and mental health are influenced by the commensal gut bacteria, mouse models of disease are influenced by the fecal microbiome (FM). The source of mice represents one of the strongest influences on the FM and can influence the phenotype of disease models. The FM influences behavior in mice leading to the hypothesis that mice of the same genetic background from different vendors, will have different behavioral phenotypes. To test this hypothesis, colonies of CD-1 mice, rederived via embryo transfer into surrogate dams from four different suppliers, were subjected to phenotyping assays assessing behavior and physiological parameters. Significant differences in behavior, growth rate, metabolism, and hematological parameters were observed. Collectively, these findings show the profound influence of supplier-origin FMs on host behavior and physiology in healthy, genetically similar, wild-type mice maintained in identical environments.
Collapse
|
14
|
Zhang C, Franklin CL, Ericsson AC. Consideration of Gut Microbiome in Murine Models of Diseases. Microorganisms 2021; 9:microorganisms9051062. [PMID: 34068994 PMCID: PMC8156714 DOI: 10.3390/microorganisms9051062] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 04/29/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome (GM), a complex community of bacteria, viruses, protozoa, and fungi located in the gut of humans and animals, plays significant roles in host health and disease. Animal models are widely used to investigate human diseases in biomedical research and the GM within animal models can change due to the impact of many factors, such as the vendor, husbandry, and environment. Notably, variations in GM can contribute to differences in disease model phenotypes, which can result in poor reproducibility in biomedical research. Variation in the gut microbiome can also impact the translatability of animal models. For example, standard lab mice have different pathogen exposure experiences when compared to wild or pet store mice. As humans have antigen experiences that are more similar to the latter, the use of lab mice with more simplified microbiomes may not yield optimally translatable data. Additionally, the literature describes many methods to manipulate the GM and differences between these methods can also result in differing interpretations of outcomes measures. In this review, we focus on the GM as a potential contributor to the poor reproducibility and translatability of mouse models of disease. First, we summarize the important role of GM in host disease and health through different gut–organ axes and the close association between GM and disease susceptibility through colonization resistance, immune response, and metabolic pathways. Then, we focus on the variation in the microbiome in mouse models of disease and address how this variation can potentially impact disease phenotypes and subsequently influence research reproducibility and translatability. We also discuss the variations between genetic substrains as potential factors that cause poor reproducibility via their effects on the microbiome. In addition, we discuss the utility of complex microbiomes in prospective studies and how manipulation of the GM through differing transfer methods can impact model phenotypes. Lastly, we emphasize the need to explore appropriate methods of GM characterization and manipulation.
Collapse
Affiliation(s)
- Chunye Zhang
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA;
| | - Craig L. Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA;
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO 65201, USA
- Metagenomics Center, University of Missouri, Columbia, MO 65201, USA
- Correspondence: (C.L.F.); (A.C.E.)
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65201, USA;
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO 65201, USA
- Metagenomics Center, University of Missouri, Columbia, MO 65201, USA
- Correspondence: (C.L.F.); (A.C.E.)
| |
Collapse
|
15
|
Buchheister S, Bleich A. Health Monitoring of Laboratory Rodent Colonies-Talking about (R)evolution. Animals (Basel) 2021; 11:1410. [PMID: 34069175 PMCID: PMC8155880 DOI: 10.3390/ani11051410] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/02/2021] [Accepted: 05/10/2021] [Indexed: 01/15/2023] Open
Abstract
The health monitoring of laboratory rodents is essential for ensuring animal health and standardization in biomedical research. Progress in housing, gnotobiotic derivation, and hygienic monitoring programs led to enormous improvement of the microbiological quality of laboratory animals. While traditional health monitoring and pathogen detection methods still serve as powerful tools for the diagnostics of common animal diseases, molecular methods develop rapidly and not only improve test sensitivities but also allow high throughput analyses of various sample types. Concurrently, to the progress in pathogen detection and elimination, the research community becomes increasingly aware of the striking influence of microbiome compositions in laboratory animals, affecting disease phenotypes and the scientific value of research data. As repeated re-derivation cycles and strict barrier husbandry of laboratory rodents resulted in a limited diversity of the animals' gut microbiome, future monitoring approaches will have to reform-aiming at enhancing the validity of animal experiments. This review will recapitulate common health monitoring concepts and, moreover, outline strategies and measures on coping with microbiome variation in order to increase reproducibility, replicability and generalizability.
Collapse
Affiliation(s)
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany;
| |
Collapse
|
16
|
Basic M, Peppermüller PP, Bolsega S, Bleich A, Bornemann M, Bode U, Buettner M. Lymph Node Stromal Cells From Different Draining Areas Distinctly Regulate the Development of Chronic Intestinal Inflammation. Front Immunol 2021; 11:549473. [PMID: 33664727 PMCID: PMC7921801 DOI: 10.3389/fimmu.2020.549473] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
The balance between the responsiveness of the intestinal immune system and the gut environment is fundamental for the maintenance of intestinal homeostasis, which is required for an adequate recognition of entering antigens. The disruption of this homeostasis by exaggerated immune response to harmless antigens can lead to the development of intestinal disorders such as inflammatory bowel disease. Stromal cells are sessile non-hematopoietic cells that build the backbone of the lymph node, an important site for the immune response induction, but also contribute to immune response and tolerance induction. However, the knowledge about the role of stromal cells in the regulation of inflammatory responses is still limited. Therefore, in this study we analyzed the influence of stromal cells on the development of chronic intestinal inflammation. Here, we show that intestinal inflammation alters the immune activation of the mesenteric lymph node-derived stromal cells. Podoplanin+ and CD21/35+ stromal cells showed increased expression of MHC class II molecules, but CD106 expression on CD21/35+ cells was reduced. Stromal cells secreted cytokines and chemokines such as CCL7 and CXCL16 influenced the gut-homing phenotype and proliferation of CD4+ and CD8+ T cells. Furthermore, stromal cells of peripheral lymph nodes transplanted into the mesentery attenuated colitis severity in B6-Il10-/- mice. The reduced colitis severity in these mice was associated with increased expression of IL4 and distinct activation pattern of stromal cells derived from transplanted peripheral lymph nodes. Altogether, our results demonstrate that lymph node stromal cells impact development of chronic colitis via T cell induction. Moreover, lymph node stromal cells from different draining area due to neonatally imprinted processes distinctly regulate the induction of immune responses.
Collapse
Affiliation(s)
- Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | | | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Melanie Bornemann
- Institute for Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Ulrike Bode
- Institute for Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| | - Manuela Buettner
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany.,Institute for Functional and Applied Anatomy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Overstreet AMC, Ramer-Tait AE, Suchodolski JS, Hostetter JM, Wang C, Jergens AE, Phillips GJ, Wannemuehler MJ. Temporal Dynamics of Chronic Inflammation on the Cecal Microbiota in IL-10 -/- Mice. Front Immunol 2021; 11:585431. [PMID: 33664728 PMCID: PMC7921487 DOI: 10.3389/fimmu.2020.585431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
The intestinal microbiota is a critical component of mucosal health as evidenced by the fact that alterations in the taxonomic composition of the gastrointestinal microbiota are associated with inflammatory bowel diseases. To better understand how the progression of inflammation impacts the composition of the gastrointestinal microbiota, we used culture independent taxonomic profiling to identify temporal changes in the cecal microbiota of C3Bir IL-10-/- mice concomitantly with the onset and progression of colitis. This analysis revealed that IL-10-/- mice displayed a biphasic progression in disease severity, as evidenced by histopathological scores and cytokine production. Beginning at 4 weeks of age, pro-inflammatory cytokines including TNF-α, IFN-γ, IL-6, G-CSF, and IL-1α as well as chemokines including RANTES and MIP-1α were elevated in the serum of IL-10-/- mice. By 19 weeks of age, the mice developed clinical signs of disease as evidenced by weight loss, which was accompanied by a significant increase in serum levels of KC and IL-17. While the overall diversity of the microbiota of both wild type and IL-10-/- were similar in young mice, the latter failed to increase in complexity as the mice matured and experienced changes in abundance of specific bacterial taxa that are associated with inflammatory bowel disease in humans. Collectively, these results reveal that there is a critical time in young mice between four to six weeks of age when inflammation and the associated immune responses adversely affect maturation of the microbiota.
Collapse
Affiliation(s)
- Anne-Marie C Overstreet
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Amanda E Ramer-Tait
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States
| | - Jan S Suchodolski
- GI Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jesse M Hostetter
- Department of Pathology, University of Georgia, Athens, GA, United States
| | - Chong Wang
- Veterinary Diagnostics and Production Animal Medicine, Iowa State University, Ames, IA, United States
| | - Albert E Jergens
- Veterinary Clinical Science, Iowa State University, Ames, IA, United States
| | - Gregory J Phillips
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Michael J Wannemuehler
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
18
|
Ericsson AC, Franklin CL. The gut microbiome of laboratory mice: considerations and best practices for translational research. Mamm Genome 2021; 32:239-250. [PMID: 33689000 PMCID: PMC8295156 DOI: 10.1007/s00335-021-09863-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
Just as the gut microbiota (GM) is now recognized as an integral mediator of environmental influences on human physiology, susceptibility to disease, and response to pharmacological intervention, so too does the GM of laboratory mice affect the phenotype of research using mouse models. Multiple experimental factors have been shown to affect the composition of the GM in research mice, as well as the model phenotype, suggesting that the GM represents a major component in experimental reproducibility. Moreover, several recent studies suggest that manipulation of the GM of laboratory mice can substantially improve the predictive power or translatability of data generated in mouse models to the human conditions under investigation. This review provides readers with information related to these various factors and practices, and recommendations regarding methods by which issues with poor reproducibility or translatability can be transformed into discoveries.
Collapse
Affiliation(s)
- Aaron C Ericsson
- University of Missouri Metagenomics Center (MUMC), MU Mutant Mouse Resource and Research Center (MU MMRRC), Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA.
| | - Craig L Franklin
- University of Missouri Metagenomics Center (MUMC), MU Mutant Mouse Resource and Research Center (MU MMRRC), Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
19
|
Franklin CL, Ericsson AC. Complex Microbiota in Laboratory Rodents: Management Considerations. ILAR J 2020; 60:289-297. [PMID: 32706377 DOI: 10.1093/ilar/ilaa011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 03/29/2020] [Accepted: 04/21/2020] [Indexed: 02/07/2023] Open
Abstract
Our bodies and those of our animal research subjects are colonized by bacterial communities that occupy virtually every organ system, including many previously considered sterile. These bacteria reside as complex communities that are collectively referred to as microbiota. Prior to the turn of the century, characterization of these communities was limited by a reliance on culture of organisms on a battery of selective media. It was recognized that the vast majority of microbes, especially those occupying unique niches of the body such as the anaerobic environment of the intestinal tract, were uncultivatable. However, with the onset and advancement of next-generation sequencing technology, we are now capable of characterizing these complex communities without the need to cultivate, and this has resulted in an explosion of information and new challenges in interpreting data generated about, and in the context of, these complex communities. We have long known that these microbial communities often exist in an intricate balance that, if disrupted (ie, dysbiosis), can lead to disease or increased susceptibility to disease. Because of many functional redundancies, the makeup of these colonies can vary dramatically within healthy individuals [1]. However, there is growing evidence that subtle differences can alter the phenotype of various animal models, which may translate to the varying susceptibility to disease seen in the human population. In this manuscript, we discuss how to include complex microbiota as a consideration in experimental design and model reproducibility and how to exploit the extensive variation that exists in contemporary rodent research colonies. Our focus will be the intestinal or gut microbiota (GM), but it should be recognized that microbial communities exist in many other body compartments and these too likely influence health and disease [2, 3]. Much like host genetics, can we one day harness the vast genetic capacity of the microbes we live with in ways that will benefit human and animal health?
Collapse
Affiliation(s)
- Craig L Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri.,Mutant Mouse Resource and Research Center, University of Missouri, Columbia, Missouri.,MU Metagenomics Center, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri.,Mutant Mouse Resource and Research Center, University of Missouri, Columbia, Missouri.,MU Metagenomics Center, College of Veterinary Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
20
|
Wolfe AE, Moskowitz JE, Franklin CL, Wiemken TL, Ericsson AC. Interactions of Segmented Filamentous Bacteria (Candidatus Savagella) and bacterial drivers in colitis-associated colorectal cancer development. PLoS One 2020; 15:e0236595. [PMID: 32706816 PMCID: PMC7380633 DOI: 10.1371/journal.pone.0236595] [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: 03/27/2020] [Accepted: 07/08/2020] [Indexed: 01/19/2023] Open
Abstract
Colorectal cancer (CRC) risk is influenced by host genetics, sex, and the gut microbiota. Using a genetically susceptible mouse model of CRC induced via inoculation with pathobiont Helicobacter spp. and demonstrating variable tumor incidence, we tested the ability of the Th17-enhancing commensal Candidatus Savagella, more commonly denoted as Segmented Filamentous Bacteria (SFB), to influence the incidence and severity of colitis-associated CRC in male and female mice. To document the composition of the gut microbiota during CRC development and identify taxa associated with disease, fecal samples were collected before and throughout disease development and characterized via 16S rRNA sequencing. While there were no significant SFB-dependent effects on disease incidence or severity, SFB was found to exert a sex-dependent protective effect in male mice. Furthermore, SFB stabilized the GM against Helicobacter-induced changes post-inoculation, resulting in a shift in disease association from Helicobacter spp. to Escherichia coli. These data support sex-dependent SFB-mediated effects on CRC risk, and highlight the complex community dynamics within the GM during exposure to inflammatory pathobionts.
Collapse
Affiliation(s)
- Annie E. Wolfe
- Molecular Pathogenesis and Therapeutics, University of Missouri, Columbia, Missouri, United States of America
| | - Jacob E. Moskowitz
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Craig L. Franklin
- Molecular Pathogenesis and Therapeutics, University of Missouri, Columbia, Missouri, United States of America
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- University of Missouri Metagenomics Center, Columbia, Missouri, United States of America
- University of Missouri Mutant Mouse Resource and Research Center, Columbia, Missouri, United States of America
- University of Missouri College of Veterinary Medicine, Columbia, Missouri, United States of America
| | - Timothy L. Wiemken
- Saint Louis University Center for Health Outcomes Research, St. Louis, Missouri, United States of America
| | - Aaron C. Ericsson
- Molecular Pathogenesis and Therapeutics, University of Missouri, Columbia, Missouri, United States of America
- Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
- University of Missouri Metagenomics Center, Columbia, Missouri, United States of America
- University of Missouri Mutant Mouse Resource and Research Center, Columbia, Missouri, United States of America
- University of Missouri College of Veterinary Medicine, Columbia, Missouri, United States of America
- * E-mail:
| |
Collapse
|
21
|
Lee NN, Bidot WA, Ericsson AC, Franklin CL. Effects of Giardia lamblia Colonization and Fenbendazole Treatment on Canine Fecal Microbiota. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2020; 59:423-429. [PMID: 32370821 PMCID: PMC7338874 DOI: 10.30802/aalas-jaalas-19-000113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/07/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
The gut microbiota (GM) is the sum of hundreds of distinct microbial species that can equal or outnumber their host's somatic cells. The GM influences a multitude of physiologic and immunologic processes in the host, and changes in the GM have been shown to alter the phenotypes of animal models. Previous studies using rodents have also shown that the composition of the GM is affected by many factors, including diet, husbandry, housing, and the genetic background of the animals. However, limited information exists about factors that may modulate GM in other laboratory species, such as dogs. We sought to eliminate sporadic Giardia colonization of dogs using fenbendazole (FBZ), an antiprotozoal widely used in biomedical research dog colonies. Concerns that FBZ could have inadvertent effects on the canine GM led us to assess GM over the course of treatment. FBZ (50 mg/kg) was given orally to all dogs in 3 different facilities (n = 19 to 25) for 10 consecutive days. Fecal samples were obtained 2 d before the initiation of treatment, on the last day of treatment, and 2 wk after the completion of treatment. Targeted 16S rRNA gene sequencing was used to analyze fecal microbiota. All dogs were clinically normal throughout the sample collection period. Statistical analyses of data showed significant differences between dogs housed in the 3 different facilities, further emphasizing the effect of housing and husbandry factors on the GM. However, negligible differences were seen between time points, indicating that FBZ did not significantly alter the canine GM. Comparison of the GM of Giardia lamblia positive and negative dogs revealed no significant difference between the 2 groups. These findings suggest that FBZ can be used therapeutically in dogs with minimal impact on the GM. Furthermore, the presence of G. lamblia in clinically normal animals may not be sufficient to influence the normal canine microbiota.
Collapse
Affiliation(s)
- Naomi N Lee
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Willie A Bidot
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Aaron C Ericsson
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Craig L Franklin
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri;,
| |
Collapse
|
22
|
|
23
|
Radaelli E, Santagostino SF, Sellers RS, Brayton CF. Immune Relevant and Immune Deficient Mice: Options and Opportunities in Translational Research. ILAR J 2019; 59:211-246. [PMID: 31197363 PMCID: PMC7114723 DOI: 10.1093/ilar/ily026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/03/2018] [Indexed: 12/29/2022] Open
Abstract
In 1989 ILAR published a list and description of immunodeficient rodents used in research. Since then, advances in understanding of molecular mechanisms; recognition of genetic, epigenetic microbial, and other influences on immunity; and capabilities in manipulating genomes and microbiomes have increased options and opportunities for selecting mice and designing studies to answer important mechanistic and therapeutic questions. Despite numerous scientific breakthroughs that have benefitted from research in mice, there is debate about the relevance and predictive or translational value of research in mice. Reproducibility of results obtained from mice and other research models also is a well-publicized concern. This review summarizes resources to inform the selection and use of immune relevant mouse strains and stocks, aiming to improve the utility, validity, and reproducibility of research in mice. Immune sufficient genetic variations, immune relevant spontaneous mutations, immunodeficient and autoimmune phenotypes, and selected induced conditions are emphasized.
Collapse
Affiliation(s)
- Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sara F Santagostino
- Department of Safety Assessment, Genentech, Inc., South San Francisco, California
| | | | - Cory F Brayton
- Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
The gut microbiota modulates differential adenoma suppression by B6/J and B6/N genetic backgrounds in Apc Min mice. Mamm Genome 2019; 30:237-244. [PMID: 31549210 PMCID: PMC6842652 DOI: 10.1007/s00335-019-09814-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/07/2019] [Indexed: 02/06/2023]
Abstract
Tumor multiplicity in the ApcMin (Min) mouse model of CRC is a classic quantitative trait that is subject to complex genetic and environmental factors, and therefore serves as an ideal platform to study modifiers of disease. While disparate inbred genetic backgrounds have well-characterized modifying effects on tumor multiplicity, it is unclear whether more closely related backgrounds such as C57BL/6J and C57BL6/N differentially modify the phenotype. Furthermore, it is unknown whether the complex gut microbiota (GM) influences the effects of these background strains. We assessed tumor multiplicity in F1 mice generated from the original Min colony from the McArdle Laboratory at the University of Wisconsin (C57BL/6JMlcr-ApcMin) crossed with either C57BL/6J or C57BL/6N wild-type mice. We also used complex microbiota targeted rederivation to rederive B6NB6JMF1-ApcMin embryos using surrogate dams harboring complex GMs from two different sources to determine the effects of complex GM. Both B6/J and B6/N backgrounds significantly repressed tumor multiplicity. However, the B6/N background conferred a stronger dominant suppressive effect than B6/J. Moreover, we observed that complex GM likely modulated B6/N-mediated adenoma repression such that two distinct communities conferred differential tumor multiplicity in isogenic B6NB6JMF1-ApcMin mice. Although we cannot rule out possible maternal effects of embryo transfer, we show that B6/J and B6/N have modifier effects on Min, and these effects are further altered by the complex GM. Foremost, strict attention to genetic background and environmental variables influencing the GM is critical to enhance reproducibility in models of complex disease traits.
Collapse
|
25
|
Affiliation(s)
- Axel Kornerup Hansen
- 1 Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Craig Franklin
- 2 Mouse Resource and Research Center, College of Veterinary Medicine, University of Missouri, USA
| |
Collapse
|
26
|
Bolsega S, Basic M, Smoczek A, Buettner M, Eberl C, Ahrens D, Odum KA, Stecher B, Bleich A. Composition of the Intestinal Microbiota Determines the Outcome of Virus-Triggered Colitis in Mice. Front Immunol 2019; 10:1708. [PMID: 31396223 PMCID: PMC6664081 DOI: 10.3389/fimmu.2019.01708] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022] Open
Abstract
The intestinal microbiota is a complex ecosystem implicated in host health and disease. Inflammatory bowel disease (IBD) is a multifactorial chronic disorder of the gastrointestinal mucosa. Even though the exact mechanisms are still unknown, the intestinal microbiota is crucial in IBD development. We previously showed that murine norovirus (MNV) induces colitis in the Il10-deficient (Il10−/−) mouse model of IBD in a microbiota-dependent manner. Thus, in this study we analyzed whether distinct minimal bacterial consortia influence the outcome of MNV-triggered colitis in Il10−/− mice. Gnotobiotic Il10−/− mice associated with Oligo-Mouse-Microbiota 12 (OMM12) or Altered Schaedler Flora (ASF) developed little to no inflammatory lesions in the colon and cecum. MNV infection exacerbated colitis severity only in ASF-colonized mice, but not in those associated with OMM12. Four weeks after MNV infection, inflammatory lesions in ASF-colonized Il10−/− mice were characterized by epithelial hyperplasia, infiltration of inflammatory cells, and increased barrier permeability. Co-colonization of ASF-colonized Il10−/− mice with segmented filamentous bacteria (SFB) abolished MNV-induced colitis, whereas histopathological scores in SFB-OMM12-co-colonized mice stayed unchanged. Moreover, SFB only colonized mice associated with ASF. The SFB-mediated protective effects in ASF-colonized mice involved enhanced activation of intestinal barrier defense mechanisms and mucosal immune responses in the chronic and acute phase of MNV infection. SFB colonization strengthened intestinal barrier function by increasing expression of tight junction proteins, antimicrobial peptides and mucus. Furthermore, SFB colonization enhanced the expression of pro-inflammatory cytokines such as Tnfα, Il1β, and Il12a, as well as the expression of the regulatory cytokine Tgfβ. Altogether, our results showed that MNV-triggered colitis depends on the microbial context.
Collapse
Affiliation(s)
- Silvia Bolsega
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Marijana Basic
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Anna Smoczek
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Manuela Buettner
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Claudia Eberl
- Faculty of Medicine, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, LMU Munich, Munich, Germany
| | - Daniel Ahrens
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Kodwo Appoh Odum
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| | - Bärbel Stecher
- Faculty of Medicine, Max von Pettenkofer Institute of Hygiene and Medical Microbiology, LMU Munich, Munich, Germany.,German Center of Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Andre Bleich
- Hannover Medical School, Institute for Laboratory Animal Science, Hanover, Germany
| |
Collapse
|
27
|
Zheng C, Huang Y, Hu W, Shi J, Ye Z, Qian X, Huang Z, Xue A, Wang Y, Lu J, Tang Z, Wu J, Wang L, Peng K, Zhou Y, Miao S, Sun H. Phenotypic Characterization of Very Early-Onset Inflammatory Bowel Disease with Interleukin-10 Signaling Deficiency: Based on a Large Cohort Study. Inflamm Bowel Dis 2019; 25:756-766. [PMID: 30212871 DOI: 10.1093/ibd/izy289] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Interleukin-10 (IL10)/interleukin-10 receptor (IL10R) deficiency is a rare disease with life-threatening infantile-onset colitis. We sought to accurately phenotype this disorder based on a large cohort of patients with a proven defect of IL10 signaling and to clarify the effects of allogeneic hematopoietic stem cell transplantation (HSCT). METHODS We analyzed the phenotypes of our 61 patients and reviewed 78 other previously reported cases with identified mutations in the genes encoding IL10 or IL10R. We also compared the clinical features of patients with interleukin-10 receptor B (IL10RB), interleukin-10 receptor A (IL10RA), and IL10 mutations. The therapeutic effects of allogeneic HSCT were evaluated. RESULTS We found that the disease onset time was extremely early: 70.3% within 30 days postnatal and 94.9% within the first 6 months of life. In addition, 94.2% of patients typically presented with perianal lesions. Oral ulcers and skin rash were common extra-intestinal manifestations (33.8% and 51.8%, respectively). There was no statistically significant difference in disease onset time, perianal lesion involvement, or mortality rate among patients with IL10RB, IL10RA, or IL10 deficiency. However, the surgery rate was higher in patients with IL10RB mutations than in those with IL10 or IL10RA mutations (P < 0.05). Compared with those with IL10RA deficiency, a higher percentage (32%, 9 of 28) of patients with IL10RB mutations developed B-cell lymphoma (P < 0.01). Compared with other regions, a higher percentage (98.7%) of IL10RA mutations was detected among patients in East Asia countries (P < 0.01), with hot-spot mutation sites of c.C301T and c.G537A. Allogeneic HSCT is efficacious but has a high mortality rate (17.5%, 7 of 40). CONCLUSIONS Our study expands the current knowledge on the genotype-correlated phenotypes with a defect of IL10 signaling. B-cell lymphoma was more frequent than would be expected in patients with IL10RB mutations. There may be a unique genetic architecture among Eastern Asia compared with other populations. Although allogeneic HSCT represents a causal therapeutic approach for IL10-and IL10R-deficient patients, a word of caution is warranted.
Collapse
Affiliation(s)
- Cuifang Zheng
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Huang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Wenhui Hu
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Jieru Shi
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Ziqing Ye
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Xiaowen Qian
- Department of Hematology & Oncology, Children's Hospital of Fudan University, Shanghai, China
| | - Zhiheng Huang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Aijuan Xue
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Yuhuan Wang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Junping Lu
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Zifei Tang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Jie Wu
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Lin Wang
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Kaiyue Peng
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Zhou
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Shijian Miao
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| | - Hua Sun
- Department of Gastroenterology, Pediatric Inflammatory Bowel Disease Research Center, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
28
|
Haney MM, Ericsson AC, Lever TE. Effects of Intraoperative Vagal Nerve Stimulation on the Gastrointestinal Microbiome in a Mouse Model of Amyotrophic Lateral Sclerosis. Comp Med 2018; 68:452-460. [PMID: 30424824 DOI: 10.30802/aalas-cm-18-000039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The gastrointestinal microbiota (GM) plays a fundamental role in health and disease and contributes to the bidirectional signaling between the gastrointestinal system and brain. The direct line of communication between these organ systems is through the vagus nerve. Therefore, vagal nerve stimulation (VNS), a commonly used technique for multiple disorders, has potential to modulate the enteric microbiota, enabling investigation and possibly treatment of numerous neurologic disorders in which the microbiota has been linked with disease. Here we investigate the effect of VNS in a mouse model of amyotrophic lateral sclerosis (ALS). B6SJL-Tg(SOD1*G93A)dl1Gur (SOD1dl) and wildtype mice underwent ventral neck surgery to access the vagus nerve. During surgery, the experimental group received 1 h of VNS, whereas the sham group underwent 1 h of sham treatment. The third (control) group did not undergo any surgical manipulation. Fecal samples were collected before surgery and at 8 d after the initial collection. Microbial DNA was sequenced to determine the GM profiles at both time points. GM profiles did not differ between genotypes at either the initial or end point. In addition, VNS did not alter GM populations, according to the parameters chosen in this study, indicating that this short intraoperative treatment is safe and has no lasting effects on the GM. Future studies are warranted to determine whether different stimulation parameters or chronic use of VNS affect GM profiles.
Collapse
Affiliation(s)
- Megan M Haney
- Metagenomics Center, University of Missouri, Columbia, Missouri, USA.
| | - Aaron C Ericsson
- Metagenomics Center, University of Missouri, Columbia, Missouri, USA
| | - Teresa E Lever
- Department of Otolaryngology-Head and Neck Surgery, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
29
|
Bidot WA, Ericsson AC, Franklin CL. Effects of water decontamination methods and bedding material on the gut microbiota. PLoS One 2018; 13:e0198305. [PMID: 30359379 PMCID: PMC6201873 DOI: 10.1371/journal.pone.0198305] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023] Open
Abstract
Rodent models are invaluable to understanding health and disease in many areas of biomedical research. Unfortunately, many models suffer from lack of phenotype reproducibility. Our laboratory has shown that differences in gut microbiota (GM) can modulate phenotypes of models of colon cancer and inflammatory bowel disease. We and others have also shown that a number of factors associated with rodent research, including vendor, cage system, and bedding can alter GM. The objective of this study was to expand these studies to examine the effect of additional bedding materials and methods of water decontamination on GM diversity and composition. To this end, Crl:CD1 (ICR) mice were housed on corn cob or compressed paper chip bedding and provided water that was decontaminated by four different methods: autoclaving with reverse osmosis, autoclaving with hydrochloric acid, autoclaving with sulfuric acid, and autoclaving alone. Feces was collected at day 0, and at day 28 (endpoint), fecal and cecal samples were collected. DNA was extracted from samples, amplified by PCR using conserved bacterial primer sets and subjected to next generation sequencing. Sequence data were analyzed using Qiime and groups were compared using principal coordinate analysis (PCoA) and permutational multivariate analysis of variance (PERMANOVA). Two factor PERMANOVA of cecal GM data revealed significant changes when comparing bedding and water decontamination methods, while no significant effects were noted in the fecal GM data. Subsequent PERMANOVA and PCoA of cecal data revealed that several combinations of bedding and water decontamination methods resulted in differing GM, highlighting the complexity by which environmental factors interact to modulate GM.
Collapse
Affiliation(s)
- Willie A. Bidot
- Comparative Medicine Program, University of Missouri, Columbia, Missouri, United States of America
| | - Aaron C. Ericsson
- Comparative Medicine Program, University of Missouri, Columbia, Missouri, United States of America
- University of Missouri Metagenomics Center, University of Missouri, Columbia, Missouri, United States of America
- Mutant Mouse Resource & Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Craig L. Franklin
- Comparative Medicine Program, University of Missouri, Columbia, Missouri, United States of America
- University of Missouri Metagenomics Center, University of Missouri, Columbia, Missouri, United States of America
- Mutant Mouse Resource & Research Center, University of Missouri, Columbia, Missouri, United States of America
| |
Collapse
|
30
|
Liu M, Zhao X, Ma Y, Zhou Y, Deng M, Ma Y. Transcription factor c-Maf is essential for IL-10 gene expression in B cells. Scand J Immunol 2018; 88:e12701. [PMID: 29974486 DOI: 10.1111/sji.12701] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 07/02/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Min Liu
- Department of Immunology; School of Medicine; Wuhan University; Wuhan China
| | - Xiaoqi Zhao
- Department of Pharmacology; School of Medicine; Wuhan University; Wuhan China
| | - Yunfeng Ma
- Department of Pathogenic Microbiology and Immunology; School of Basic Medical Sciences; Xi'an Jiaotong University Health Science Center; Xi'an, Shaanxi China
| | - Yan Zhou
- Department of Orthopaedics; Renmin Hospital; Wuhan University; Wuhan City China
| | - Ming Deng
- Department of Orthopaedics; Renmin Hospital; Wuhan University; Wuhan City China
| | - Yonggang Ma
- Department of Orthopaedics; Renmin Hospital; Wuhan University; Wuhan City China
| |
Collapse
|
31
|
Abstract
Mouse colonized with human fecal microbiota is an interesting model concept with pros and cons like any other model system. The concept provides an ecologically relevant context to study food component and drug metabolism, and is an invaluable tool for phenotype transfer studies to prove the role of the gut microbiota in health and disease. The major drawbacks are the difficulties with transferring certain components of the human microbiota to the recipient mice, and immunological abnormalities observed in these mice. There seem to be unexplored opportunities for trying to reduce these limitations, but careful evaluation of pros, cons and possible alternatives is still necessary.
Collapse
Affiliation(s)
- Randi Lundberg
- Internal Research and Development, Taconic Biosciences, Lille Skensved, Denmark
| |
Collapse
|
32
|
Hart ML, Ericsson AC, Lloyd KCK, Grimsrud KN, Rogala AR, Godfrey VL, Nielsen JN, Franklin CL. Development of outbred CD1 mouse colonies with distinct standardized gut microbiota profiles for use in complex microbiota targeted studies. Sci Rep 2018; 8:10107. [PMID: 29973630 PMCID: PMC6031694 DOI: 10.1038/s41598-018-28448-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/22/2018] [Indexed: 02/08/2023] Open
Abstract
Studies indicate that the gut microbiota (GM) can significantly influence both local and systemic host physiologic processes. With rising concern for optimization of experimental reproducibility and translatability, it is essential to consider the GM in study design. However, GM profiles can vary between rodent producers making consistency between models challenging. To circumvent this, we developed outbred CD1 mouse colonies with stable, complex GM profiles that can be used as donors for a variety of GM transfer techniques including rederivation, co-housing, cross-foster, and fecal microbiota transfer (FMT). CD1 embryos were surgically transferred into CD1 or C57BL/6 surrogate dams that varied by GM composition and complexity to establish four separate mouse colonies harboring GM profiles representative of contemporary mouse producers. Using targeted 16S rRNA amplicon sequencing, subsequent female offspring were found to have similar GM profiles to surrogate dams. Furthermore, breeding colonies of CD1 mice with distinct GM profiles were maintained for nine generations, demonstrating GM stability within these colonies. To confirm GM stability, we shipped cohorts of these four colonies to collaborating institutions and found no significant variation in GM composition. These mice are an invaluable experimental resource that can be used to investigate GM effects on mouse model phenotype.
Collapse
Affiliation(s)
- Marcia L Hart
- Comparative Medicine Program, University of Missouri, Columbia, Missouri, United States of America.,University of Missouri Metagenomics Center, University of Missouri, Columbia, Missouri, United States of America.,Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America
| | - Aaron C Ericsson
- Comparative Medicine Program, University of Missouri, Columbia, Missouri, United States of America.,University of Missouri Metagenomics Center, University of Missouri, Columbia, Missouri, United States of America.,Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America.,Mutant Mouse Resource and Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - K C Kent Lloyd
- Mouse Biology Program, University of California, Davis, California, United States of America.,Mutant Mouse Resource and Research Center, University of California, Davis, California, United States of America.,Mouse Metabolic Phenotyping Center, University of California, Davis, California, United States of America
| | - Kristin N Grimsrud
- Mouse Biology Program, University of California, Davis, California, United States of America.,Mutant Mouse Resource and Research Center, University of California, Davis, California, United States of America
| | - Allison R Rogala
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America.,National Gnotobiotic Rodent Resource Center, University of North Carolina, Chapel Hill, North Carolina, United States of America.,Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Virginia L Godfrey
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America.,Mutant Mouse Resource and Research Center, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Judith N Nielsen
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Craig L Franklin
- Comparative Medicine Program, University of Missouri, Columbia, Missouri, United States of America. .,University of Missouri Metagenomics Center, University of Missouri, Columbia, Missouri, United States of America. .,Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri, United States of America. .,Mutant Mouse Resource and Research Center, University of Missouri, Columbia, Missouri, United States of America.
| |
Collapse
|
33
|
Yilmaz B, Spalinger MR, Biedermann L, Franc Y, Fournier N, Rossel JB, Juillerat P, Rogler G, Macpherson AJ, Scharl M. The presence of genetic risk variants within PTPN2 and PTPN22 is associated with intestinal microbiota alterations in Swiss IBD cohort patients. PLoS One 2018; 13:e0199664. [PMID: 29965986 PMCID: PMC6028086 DOI: 10.1371/journal.pone.0199664] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/12/2018] [Indexed: 01/03/2023] Open
Abstract
Background Genetic risk factors, intestinal microbiota and a dysregulated immune system contribute to the pathogenesis of inflammatory bowel disease (IBD). We have previously demonstrated that dysfunction of protein tyrosine phosphatase non-receptor type 2 (PTPN2) and PTPN22 contributes to alterations of intestinal microbiota and the onset of chronic intestinal inflammation in vivo. Here, we investigated the influence of PTPN2 and PTPN22 gene variants on intestinal microbiota composition in IBD patients. Methods Bacterial DNA from mucosa-associated samples of 75 CD and 57 UC patients were sequenced using 16S rRNA sequencing approach. Microbial analysis, including alpha diversity, beta diversity and taxonomical analysis by comparing to PTPN2 (rs1893217) and PTPN22 (rs2476601) genotypes was performed in QIIME, the phyloseq R package and MaAsLin pipeline. Results In PTPN2 variant UC patients, we detected an increase in relative abundance of unassigned genera from Clostridiales and Lachnospiraceae families and reduction of Roseburia when compared to PTPN2 wild-type (WT) patients. Ruminoccocus was increased in PTPN22 variant UC patients. In CD patients with severe disease course, Faecalibacterium, Bilophila, Coprococcus, unclassified Erysipelotrichaeceae, unassigned genera from Clostridiales and Ruminococcaceae families were reduced and Bacteroides were increased in PTPN2 WT carriers, while Faecalibacterium, Bilophila, Coprococcus, and Erysipelotrichaeceae were reduced in PTPN22 WT patients when compared to patients with mild disease. In UC patients with severe disease, relative abundance of Lachnobacterium was reduced in PTPN2 and PTPN22 WT patients, Dorea was increased in samples from PTPN22 WT carriers and an unassigned genus from Ruminococcaceae gen. was increased in patients with PTPN2 variant genotype. Conclusions We identified that IBD-associated genetic risk variants, disease severity and the interaction of these factors are related to significant alterations in intestinal microbiota composition of IBD patients.
Collapse
Affiliation(s)
- Bahtiyar Yilmaz
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marianne R. Spalinger
- Department of Gastroenterology and Hepatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Yannick Franc
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, Switzerland
| | - Nicolas Fournier
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, Switzerland
| | - Jean-Benoit Rossel
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne, Switzerland
| | - Pascal Juillerat
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gerhard Rogler
- Department of Gastroenterology and Hepatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Andrew J. Macpherson
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
34
|
Montonye DR, Ericsson AC, Busi SB, Lutz C, Wardwell K, Franklin CL. Acclimation and Institutionalization of the Mouse Microbiota Following Transportation. Front Microbiol 2018; 9:1085. [PMID: 29892276 PMCID: PMC5985407 DOI: 10.3389/fmicb.2018.01085] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 05/07/2018] [Indexed: 01/02/2023] Open
Abstract
Using animal models, the gut microbiota has been shown to play a critical role in the health and disease of many organ systems. Unfortunately, animal model studies often lack reproducibility when performed at different institutions. Previous studies in our laboratory have shown that the gut microbiota of mice can vary with a number of husbandry factors leading us to speculate that differing environments may alter gut microbiota, which in turn may influence animal model phenotypes. As an extension of these studies, we hypothesized that the shipping of mice from a mouse producer to an institution will result in changes in the type, relative abundance, and functional composition of the gut microbiota. Furthermore, we hypothesized that mice will develop a microbiota unique to the institution and facility in which they are housed. To test these hypotheses, mice of two strains (C57BL/6J and BALB/cJ), two age groups (4 week and 8 week old), and originating from two types of housing (research animal facility under conventional housing and production facilities under maximum barrier housing) were obtained from The Jackson Laboratory. Fecal samples were collected the day prior to shipping, immediately upon arrival, and then on days 2, 5, 7, and weeks 2, 4, and 9 post-arrival. Following the first post-arrival fecal collection, mice were separated into 2 groups and housed at different facilities at our institution while keeping their caging, diet, and husbandry practices the same. DNA was extracted from the collected fecal pellets and 16S rRNA amplicons were sequenced in order to characterize the type and relative abundance of gut bacteria. Principal component analysis (PCA) and permutational multivariate analysis of variance (PERMANOVA) demonstrated that both the shipping and the institution and facility in which mice were housed altered the gut microbiota. Phylogenetic investigation of communities by reconstruction of unobserved states (PICRUSt) predicted differences in functional composition in the gut microbiota of mice based on time of acclimation.
Collapse
Affiliation(s)
- Dan R Montonye
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Aaron C Ericsson
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States.,University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, United States.,University of Missouri Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, United States
| | - Susheel B Busi
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Cathleen Lutz
- The Jackson Laboratory, Bar Harbor, ME, United States
| | | | - Craig L Franklin
- Comparative Medicine Program, Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States.,University of Missouri Metagenomics Center, University of Missouri, Columbia, MO, United States.,University of Missouri Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, United States
| |
Collapse
|
35
|
Korte SW, Franklin CL, Dorfmeyer RA, Ericsson AC. Effects of Fenbendazole-impregnated Feed and Topical Moxidectin during Quarantine on the Gut Microbiota of C57BL/6 Mice. JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2018; 57:229-235. [PMID: 29784074 PMCID: PMC5966229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/09/2017] [Accepted: 12/13/2017] [Indexed: 06/08/2023]
Abstract
To protect the biosecurity of research rodent colonies, research institutions frequently require a quarantine period for live animals transferred into their facilities. Quarantine practices often include antibiotic and antiparasitic treatment with drugs such as fenbendazole and macrolide lactones. The influence of these compounds on the resident gut microbiota of mice is unknown, and any effects might subsequently affect model reproducibility. To test the influence of standard quarantine procedures on the composition of the microbiota, C57BL/6 mice, purchased from 2 different commercial suppliers, were randomly assigned to treatment groups (n = 12) by vendor and treated with fenbendazole-supplemented feed, topical moxidectin, both treatments, or no treatment (control), according to our institution's standard treatment regimen and duration. Feces were collected on arrival, immediately after completing the 8-wk treatment, and at 2 and 4 wk after treatment. Fecal DNA was extracted, sequenced, and analyzed to compare the changes in the microbiota of treated and control groups. Although significant main effects of time and treatment and interactions between those variables were detected in comparisons of richness, α-diversity, and β-diversity, the effect sizes associated with any particular treatment were consistently much smaller than that associated with acclimation to a new facility in the absence of any quarantine treatments. This outcome, along with the visual evaluation of principal coordinate analysis based on multiple similarity indices, suggests that time or institution plays a larger role in alterations of the murine gut microbiota than do quarantine treatments on its composition.
Collapse
Affiliation(s)
- Scott W Korte
- Office of Animal Resources, University of Missouri, Columbia, Missouri
| | - Craig L Franklin
- Metagenomics Center, Mutant Mouse Resource and Research Center, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri
| | - Rebecca A Dorfmeyer
- Mutant Mouse Resource and Research Center, University of Missouri, Columbia, Missouri
| | - Aaron C Ericsson
- Metagenomics Center, Mutant Mouse Resource and Research Center, Department of Veterinary Pathobiology, University of Missouri, Columbia, Missouri;,
| |
Collapse
|
36
|
Yang L, Shen L, Li Y, Li Y, Yu S, Wang S. Hyperoside attenuates dextran sulfate sodium-induced colitis in mice possibly via activation of the Nrf2 signalling pathway. JOURNAL OF INFLAMMATION-LONDON 2017; 14:25. [PMID: 29162986 PMCID: PMC5686943 DOI: 10.1186/s12950-017-0172-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 11/06/2017] [Indexed: 12/28/2022]
Abstract
Background Hyperoside (Hyp) is a flavonoid glycoside compound that has been demonstrated to have anti-inflammatory, anti-apoptotic and antioxidant effects. However, the impact of Hyp on inflammatory bowel disease (IBD) has not been previously explored. Thus, we evaluated the role of Hyp in dextran sodium sulfate (DSS)-induced acute colitis in mice. Methods We established a mouse model of experimental acute colitis by treating mice with drinking water supplemented with 3.0% DSS for 7 days. The disease activity index (DAI), colon length, histological features and colonic malondialdehyde (MDA) levels were examined using appropriate methods, and COX-2 expression was examined by immunohistochemistry. TNF-α, IL-4, IL-6, IL-10, NF-κB p65, Bcl-2, Bax, Caspase-3, nuclear factor-erythroid 2-related factor 2 (Nrf2), hemeoxygenase-1 (HO-1) and superoxide dismutase (SOD) levels in colorectal tissues were detected by RT-PCR and western blotting. Results Hyp significantly attenuated DSS-induced changes in the DAI as well as DSS-induced colonic shortening and histological changes. Hyp also inhibited inflammation, a change reflected by decreases in TNF-α, IL-6, COX-2 and NF-κB p65 expression and increases in IL-10 expression. Hyp suppressed increases in the levels of apoptosis-related proteins, such as Caspase-3 and Bax, but upregulated the level of the anti-apoptotic protein Bcl2. In addition, Hyp also exerted antioxidant effects. The MDA content was decreased, and the expression of Nrf2 and its downstream targets HO-1 and SOD were increased by Hyp. Conclusions Based on these findings, Hyp possesses the ability to attenuate colitis, possibly by mitigating colonic inflammation and apoptosis via activation of the Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Lei Yang
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Lei Shen
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Yue Li
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Yanxia Li
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Shijie Yu
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| | - Shanshan Wang
- Department of Gastroenterology, Key Laboratory of Hubei Province for Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, 430060 China
| |
Collapse
|
37
|
|
38
|
Ericsson AC, Montonye DR, Smith CR, Franklin CL. Modeling a Superorganism - Considerations Regarding the Use of "Dirty" Mice in Biomedical Research
. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2017; 90:361-371. [PMID: 28955177 PMCID: PMC5612181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
An ever-expanding body of evidence in both humans and animal models demonstrates the influence of the resident gut microbiota on host health and disease susceptibility. However, as unwanted bacterial, viral, protozoal, and parasitic agents have gradually been eliminated from colonies of purpose-bred laboratory mice, the resident microbiota has lost richness and complexity. Recent studies have shown that the ultra-hygienic environment of traditional laboratory mice and lack of antigenic exposure during development results in mice with an immune system more akin to that of a neonate than an adult human. In contrast, wild mice or mice purchased from pet stores are exposed to much greater antigen burdens and their immune system reflects this with significantly greater numbers of memory T cells and more robust vaccine responses. The current review explores the use of alternative sources for research rodents, with an emphasis on the differences in resident gut microbiota and pathogen burden between wild mice, pet store-origin mice, and traditional laboratory mice. Specifically, the literature is compared and contrasted to our own data reflecting the endogenous gut microbiota and pathogen load of wild and pet store mice, as well as the changes in both during and after procedures intended to eliminate certain zoonotic agents present in pet store mice. These data demonstrate that, while alternative sources of research rodents will likely provide models that are more translatable to the human condition, there are also several real-world considerations for scientists including contamination of research facilities and human health risks such as zoonotic diseases.
Collapse
Affiliation(s)
- Aaron C. Ericsson
- University of Missouri Metagenomics Center (MUMC), Columbia, MO,University of Missouri Mutant Mouse Resource and Research Center (MMRRC), Columbia, MO,Rat Resource and Research Center (RRRC), Columbia, MO,Department of Veterinary Pathobiology, University of Missouri, Columbia, MO,To whom all correspondence should be addressed: Aaron C. Ericsson, 4011 Discovery Drive, S123B, Columbia, MO 65201, Phone: 573-882-1019, Fax: 573-882-9857,
| | - Daniel R. Montonye
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | - Cierra R. Smith
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| | - Craig L. Franklin
- University of Missouri Metagenomics Center (MUMC), Columbia, MO,University of Missouri Mutant Mouse Resource and Research Center (MMRRC), Columbia, MO,Rat Resource and Research Center (RRRC), Columbia, MO,Department of Veterinary Pathobiology, University of Missouri, Columbia, MO
| |
Collapse
|