130 results on '"Rheinallt M. Jones"'
Search Results
2. Intestinal barrier dysfunction in murine sickle cell disease is associated with small intestine neutrophilic inflammation, oxidative stress, and dysbiosis
- Author
-
Caitlin V. Lewis, Hassan Sellak, Mariem A. Sawan, Giji Joseph, Trevor M. Darby, David VanInsberghe, Crystal R. Naudin, David R. Archer, Rheinallt M. Jones, and W. Robert Taylor
- Subjects
gut microbiome ,intestinal permeability ,neutrophils ,oxidative stress ,sickle cell disease ,Biology (General) ,QH301-705.5 - Abstract
Abstract The intestinal microbiome has emerged as a potential contributor to the severity of sickle cell disease (SCD). We sought to determine whether SCD mice exhibit intestinal barrier dysfunction, inflammation, and dysbiosis. Using the Townes humanized sickle cell mouse model, we found a 3‐fold increase in intestinal permeability as assessed via FITC‐dextran (4 kDa) assay in SS (SCD) mice compared to AA (wild type) mice (n = 4, p
- Published
- 2023
- Full Text
- View/download PDF
3. Callus γδ T cells and microbe-induced intestinal Th17 cells improve fracture healing in mice
- Author
-
Hamid Y. Dar, Daniel S. Perrien, Subhashis Pal, Andreea Stoica, Sasidhar Uppuganti, Jeffry S. Nyman, Rheinallt M. Jones, M. Neale Weitzmann, and Roberto Pacifici
- Subjects
Bone Biology ,Microbiology ,Medicine - Abstract
IL-17A (IL-17), a driver of the inflammatory phase of fracture repair, is produced locally by several cell lineages including γδ T cells and Th17 cells. However, the origin of these T cells and their relevance for fracture repair are unknown. Here, we show that fractures rapidly expanded callus γδ T cells, which led to increased gut permeability by promoting systemic inflammation. When the microbiota contained the Th17 cell–inducing taxon segmented filamentous bacteria (SFB), activation of γδ T cells was followed by expansion of intestinal Th17 cells, their migration to the callus, and improved fracture repair. Mechanistically, fractures increased the S1P receptor 1–mediated (S1PR1-mediated) egress of Th17 cells from the intestine and enhanced their homing to the callus through a CCL20-mediated mechanism. Fracture repair was impaired by deletion of γδ T cells, depletion of the microbiome by antibiotics (Abx), blockade of Th17 cell egress from the gut, or Ab neutralization of Th17 cell influx into the callus. These findings demonstrate the relevance of the microbiome and T cell trafficking for fracture repair. Modifications of microbiome composition via Th17 cell–inducing bacteriotherapy and avoidance of broad-spectrum Abx may represent novel therapeutic strategies to optimize fracture healing.
- Published
- 2023
- Full Text
- View/download PDF
4. The microbiome restrains melanoma bone growth by promoting intestinal NK and Th1 cell homing to bone
- Author
-
Subhashis Pal, Daniel S. Perrien, Tetsuya Yumoto, Roberta Faccio, Andreea Stoica, Jonathan Adams, Craig M. Coopersmith, Rheinallt M. Jones, M. Neale Weitzmann, and Roberto Pacifici
- Subjects
Bone Biology ,Medicine - Abstract
Bone metastases are frequent complications of malignant melanoma leading to reduced quality of life and significant morbidity. Regulation of immune cells by the gut microbiome influences cancer progression, but the role of the microbiome in tumor growth in bone is unknown. Using intracardiac or intratibial injections of B16-F10 melanoma cells into mice, we showed that gut microbiome depletion by broad-spectrum antibiotics accelerated intraosseous tumor growth and osteolysis. Microbiome depletion blunted melanoma-induced expansion of intestinal NK cells and Th1 cells and their migration from the gut to tumor-bearing bones. Demonstrating the functional relevance of immune cell trafficking from the gut to the bone marrow (BM) in bone metastasis, blockade of S1P-mediated intestinal egress of NK and Th1 cells, or inhibition of their CXCR3/CXCL9-mediated influx into the BM, prevented the expansion of BM NK and Th1 cells and accelerated tumor growth and osteolysis. Using a mouse model, this study revealed mechanisms of microbiota-mediated gut-bone crosstalk that are relevant to the immunological restraint of melanoma metastasis and tumor growth in bone. Microbiome modifications induced by antibiotics might have negative clinical consequences in patients with melanoma.
- Published
- 2022
- Full Text
- View/download PDF
5. Lactobacillus rhamnosus GG Orchestrates an Antitumor Immune ResponseSummary
- Author
-
Joshua A. Owens, Bejan J. Saeedi, Crystal R. Naudin, Sarah Hunter-Chang, Maria E. Barbian, Richard U. Eboka, Lauren Askew, Trevor M. Darby, Brian S. Robinson, and Rheinallt M. Jones, PhD
- Subjects
Probiotics ,Microbiome ,LGG ,Cancer ,CD8 T Cells ,Dendritic Cells ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: In colorectal cancer, approximately 95% of patients are refractory to immunotherapy because of low antitumor immune responses. Therefore, there is an exigent need to develop treatments that increase antitumor immune responses and decrease tumor burden to enhance immunotherapy. Methods: The gut microbiome has been described as a master modulator of immune responses. We administered the human commensal, Lactobacillus rhamnosus GG (LGG), to mice and characterized the changes in the gut immune landscape. Because the presence of lactobacilli in the gut microbiome has been linked with decreased tumor burden and antitumor immune responses, we also supplemented a genetic and a chemical model of murine intestinal cancer with LGG. For clinical relevance, we therapeutically administered LGG after tumors had formed. We also tested for the requirement of CD8 T cells in LGG-mediated modulation of gut tumor burden. Results: We detected increased colonic CD8 T-cell responses specifically in LGG-supplemented mice. The CD8 T-cell induction was dependent on dendritic cell activation mediated via Toll-like receptor-2, thereby describing a novel mechanism in which a member of the human microbiome induces an intestinal CD8 T-cell response. We also show that LGG decreased tumor burden in the murine gut cancer models by a CD8 T-cell–dependent manner. Conclusions: These data support the potential use of LGG to augment antitumor immune responses in colorectal cancer patients and ultimately for increasing the breadth and efficacy of immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
6. PTH induces bone loss via microbial-dependent expansion of intestinal TNF+ T cells and Th17 cells
- Author
-
Mingcan Yu, Abdul Malik Tyagi, Jau-Yi Li, Jonathan Adams, Timothy L. Denning, M. Neale Weitzmann, Rheinallt M. Jones, and Roberto Pacifici
- Subjects
Science - Abstract
T cells are involved in the bone loss induced by parathyroid hormone (PTH), but their origin is unknown. Here, the authors show that the intestinal microbiota is required for PTH to induce bone loss and describes mechanisms for microbiota-mediated gut–bone crosstalk in mouse models of hyperparathyroidism.
- Published
- 2020
- Full Text
- View/download PDF
7. Lactobacillus rhamnosus GG–induced Expression of Leptin in the Intestine Orchestrates Epithelial Cell ProliferationSummary
- Author
-
Trevor M. Darby, Crystal R. Naudin, Liping Luo, and Rheinallt M. Jones
- Subjects
Leptin ,Nox1 ,Probiotic ,Lactobacillus ,Probiotics ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: Identifying the functional elements that mediate efficient gut epithelial growth and homeostasis is essential for understanding intestinal health and disease. Many of these processes involve the Lactobacillus-induced generation of reactive oxygen species by NADPH oxidase (Nox1). However, the downstream signaling pathways that respond to Nox1-generated reactive oxygen species and mediate these events have not been described. Methods: Wild-type and knockout mice were fed Lactobacillus rhamnosus GG and the transcriptional and cell signaling pathway responses in the colon measured. Corroboration of data generated in mice was done using in organoid tissue culture and in vivo gut injury models. Results: Ingestion of L rhamnosus GG induces elevated levels of leptin in the gut epithelia, which as well as functioning in the context of metabolism, has pleiotropic activity as a chemokine that triggers cell proliferation. Consistently, using gut epithelial-specific knockout mice, we show that L rhamnosus GG–induced elevated levels of leptin is dependent on a functional Nox1 protein in the colonic epithelium, and that L rhamnosus GG–induced cell proliferation is dependent on Nox1, leptin, and leptin receptor. We also show that L rhamnosus GG induces the JAK-STAT signaling pathway in the gut in a Nox1, leptin, and leptin receptor–dependent manner. Conclusions: These results demonstrate a novel role for leptin in the response to colonization by lactobacilli, where leptin functions in the transduction of signals from symbiotic bacteria to subepithelial compartments, where it modulates intestinal growth and homeostasis.
- Published
- 2020
- Full Text
- View/download PDF
8. Proline-Rich Acidic Protein 1 (PRAP1) Protects the Gastrointestinal Epithelium From Irradiation-Induced ApoptosisSummary
- Author
-
Alexandra A. Wolfarth, Xu Liu, Trevor M. Darby, Darra J. Boyer, Jocelyn B. Spizman, Joshua A. Owens, Bindu Chandrasekharan, Crystal R. Naudin, Krisztina Z. Hanley, Brian S. Robinson, Eric A. Ortlund, Rheinallt M. Jones, and Andrew S. Neish
- Subjects
Oxidative Stress ,Intrinsically Disordered Proteins ,p21 ,Small Intestine ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: The intestinal epithelium must be resilient to physiochemical stress to uphold the physiological barrier separating the systemic compartment from the microbial and antigenic components of the gut lumen. Identifying proteins that mediate protection and enhancing their expression is therefore a clear approach to promote intestinal health. We previously reported that oral ingestion of the probiotic Lactobacillus rhamnosus GG not only induced the expression of several recognized cytoprotective factors in the murine colon, but also many genes with no previously described function, including the gene encoding proline-rich acidic protein 1 (PRAP1). PRAP1 is a highly expressed protein in the epithelium of the gastrointestinal tract and we sought to define its function in this tissue. Methods: Purified preparations of recombinant PRAP1 were analyzed biochemically and PRAP1 antisera were used to visualize localization in tissues. Prap1-/- mice were characterized at baseline and challenged with total body irradiation, then enteroids were generated to recapitulate the irradiation challenge ex vivo. Results: PRAP1 is a 17-kilodalton intrinsically disordered protein with no recognizable sequence homology. PRAP1 expression levels were high in the epithelia of the small intestine. Although Prap1-/- mice presented only mild phenotypes at baseline, they were highly susceptible to intestinal injury upon challenge. After irradiation, the Prap1-/- mice showed accelerated death with a significant increase in apoptosis and p21 expression in the small intestinal epithelium. Conclusions: PRAP1 is an intrinsically disordered protein highly expressed by the gastrointestinal epithelium and functions at exposed surfaces to protect the barrier from oxidative insult.
- Published
- 2020
- Full Text
- View/download PDF
9. Lactococcus Lactis Subsp. cremoris Is an Efficacious Beneficial Bacterium that Limits Tissue Injury in the Intestine
- Author
-
Trevor M. Darby, Joshua A. Owens, Bejan J. Saeedi, Liping Luo, Jason D. Matthews, Brian S. Robinson, Crystal R. Naudin, and Rheinallt M. Jones
- Subjects
Science - Abstract
Summary: The use of beneficial bacteria to promote health is widely practiced. However, experimental evidence corroborating the efficacy of bacteria promoted with such claims remains limited. We address this gap by identifying a beneficial bacterium that protects against tissue damage and injury-induced inflammation in the gut. We first employed the Drosophila animal model to screen for the capacity of candidate beneficial bacteria to protect the fly gut against injury. From this screen, we identified Lactococcus lactis subsp. cremoris as a bacterium that elicited potent cytoprotective activity. Then, in a murine model, we demonstrated that the same strain confers powerful cytoprotective influences against radiological damage, as well as anti-inflammatory activity in a gut colitis model. In summary, we demonstrate the positive salutary effects of a beneficial bacterium, namely, L. lactis subsp. cremoris on intestinal tissue and propose the use of this strain as a therapeutic to promote intestinal health. : Biological Sciences; Microbiology; Cell Biology Subject Areas: Biological Sciences, Microbiology, Cell Biology
- Published
- 2019
- Full Text
- View/download PDF
10. Proteomic analysis of microbial induced redox-dependent intestinal signaling
- Author
-
Jason D. Matthews, April R. Reedy, Huixia Wu, Benjamin H. Hinrichs, Trevor M. Darby, Caroline Addis, Brian S. Robinson, Young-Mi Go, Dean P. Jones, Rheinallt M. Jones, and Andrew S. Neish
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Intestinal homeostasis is regulated in-part by reactive oxygen species (ROS) that are generated in the colonic mucosa following contact with certain lactobacilli. Mechanistically, ROS can modulate protein function through the oxidation of cysteine residues within proteins. Recent advances in cysteine labeling by the Isotope Coded Affinity Tags (ICATs) technique has facilitated the identification of cysteine thiol modifications in response to stimuli. Here, we used ICATs to map the redox protein network oxidized upon initial contact of the colonic mucosa with Lactobacillus rhamnosus GG (LGG). We detected significant LGG-specific redox changes in over 450 proteins, many of which are implicated to function in cellular processes such as endosomal trafficking, epithelial cell junctions, barrier integrity, and cytoskeleton maintenance and formation. We particularly noted the LGG-specific oxidation of Rac1, which is a pleiotropic regulator of many cellular processes. Together, these data reveal new insights into lactobacilli-induced and redox-dependent networks involved in intestinal homeostasis.
- Published
- 2019
- Full Text
- View/download PDF
11. Cruciferous vegetables (Brassica oleracea) confer cytoprotective effects in Drosophila intestines
- Author
-
James T. Lyles, Liping Luo, Ken Liu, Dean P. Jones, Rheinallt M. Jones, and Cassandra L. Quave
- Subjects
indole ,brassica ,drosophila ,nrf2 ,leaky-gut ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Varieties and cultivars of the cruciferous vegetable Brassica oleracea are widely presumed to elicit positive influences on mammalian health and disease, particularly related to their indole and sulforaphane content. However, there is a considerable gap in knowledge regarding the mechanisms whereby these plant-derived molecules elicit their beneficial effects on the host. In this study, we examined the chemical variation between B. oleracea varieties and evaluated their capacity to both activate Nrf2 in the Drosophila intestine and elicit cytoprotection. Ten types of edible B. oleracea were purchased and B. macrocarpa was wild collected. Fresh material was dried, extracted by double maceration and green kale was also subjected to anaerobic fermentation before processing. Untargeted metabolomics was used to perform Principal Component Analysis. Targeted mass spectral analysis determined the presence of six indole species and quantified indole. Extracts were tested for their capacity to activate Nrf2 in the Drosophila intestine in third instar Drosophila larvae. Cytoprotective effects were evaluated using a paraquat-induced oxidative stress gut injury model. A “Smurf” assay was used to determine protective capacity against a chemically induced leaky gut. Extracts of Brussels sprouts and broccoli activated Nrf2 and protected against paraquat-induced damage and leaky gut. Lacto-fermented kale showed a cytoprotective effect, increasing survival by 20% over the non-fermented extract, but did not protect against leaky gut. The protective effects observed do not directly correlate with indole content, suggesting involvement of multiple compounds and a synergistic mechanism.
- Published
- 2021
- Full Text
- View/download PDF
12. At the heart of microbial conversations: endocannabinoids and the microbiome in cardiometabolic risk
- Author
-
Ramsha Nabihah Khan, Kristal Maner-Smith, Joshua A. Owens, Maria Estefania Barbian, Rheinallt M. Jones, and Crystal R. Naudin
- Subjects
endocannabinoid ,microbiome ,metabolic syndrome ,cardiovascular ,probiotic ,sexual dimorphism ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Cardiometabolic syndrome encompasses intertwined risk factors such as hypertension, dyslipidemia, elevated triglycerides, abdominal obesity, and other maladaptive metabolic and inflammatory aberrations. As the molecular mechanisms linking cardiovascular disease and metabolic disorders are investigated, endocannabinoids have emerged as molecules of interest. The endocannabinoid system (ECS) of biologically active lipids has been implicated in several conditions, including chronic liver disease, osteoporosis, and more recently in cardiovascular diseases. The gut microbiome is a major regulator of inflammatory and metabolic signaling in the host, and if disrupted, has the potential to drive metabolic and cardiovascular diseases. Extensive studies have unraveled the impact of the gut microbiome on host physiology, with recent reports showing that gut microbes exquisitely control the ECS, with significant influences on host metabolic and cardiac health. In this review, we outline how modulation of the gut microbiome affects host metabolism and cardiovascular health via the ECS, and how these findings could be exploited as novel therapeutic targets for various metabolic and cardiac diseases.
- Published
- 2021
- Full Text
- View/download PDF
13. Bifidobacterium adolescentis supplementation attenuates fracture-induced systemic sequelae
- Author
-
Joseph L. Roberts, Guanglu Liu, Trevor M. Darby, Lorenzo M. Fernandes, Martha E. Diaz-Hernandez, Rheinallt M. Jones, and Hicham Drissi
- Subjects
Probiotic ,Intestinal permeability ,Trauma ,Inflammation ,Microbiome ,Dietary supplement ,Therapeutics. Pharmacology ,RM1-950 - Abstract
The gut microbiota is an important contributor to both health and disease. While previous studies have reported on the beneficial influences of the gut microbiota and probiotic supplementation on bone health, their role in recovery from skeletal injury and resultant systemic sequelae remains unexplored. This study aimed to determine the extent to which probiotics could modulate bone repair by dampening fracture-induced systemic inflammation. Our findings demonstrate that femur fracture induced an increase in gut permeability lasting up to 7 days after trauma before returning to basal levels. Strikingly, dietary supplementation with Bifidobacterium adolescentis augmented the tightening of the intestinal barrier, dampened the systemic inflammatory response to fracture, accelerated fracture callus cartilage remodeling, and elicited enhanced protection of the intact skeleton following fracture. Together, these data outline a mechanism whereby dietary supplementation with beneficial bacteria can be therapeutically targeted to prevent the systemic pathologies induced by femur fracture.
- Published
- 2020
- Full Text
- View/download PDF
14. A Human Microbiota-Associated Murine Model for Assessing the Impact of the Vaginal Microbiota on Pregnancy Outcomes
- Author
-
Alexandra A. Wolfarth, Taylor M. Smith, David VanInsberghe, Anne Lang Dunlop, Andrew S. Neish, Elizabeth J. Corwin, and Rheinallt M. Jones
- Subjects
vaginal microbiota ,bacterial vaginosis (BV) ,humanization ,pregnancy ,inflammation ,Microbiology ,QR1-502 - Abstract
Disease states are often linked to large scale changes in microbial community structure that obscure the contributions of individual microbes to disease. Establishing a mechanistic understanding of how microbial community structure contribute to certain diseases, however, remains elusive thereby limiting our ability to develop successful microbiome-based therapeutics. Human microbiota-associated (HMA) mice have emerged as a powerful approach for directly testing the influence of microbial communities on host health and disease, with the transfer of disease phenotypes from humans to germ-free recipient mice widely reported. We developed a HMA mouse model of the human vaginal microbiota to interrogate the effects of Bacterial Vaginosis (BV) on pregnancy outcomes. We collected vaginal swabs from 19 pregnant African American women with and without BV (diagnosed per Nugent score) to colonize female germ-free mice and measure its impact on birth outcomes. There was considerable variability in the microbes that colonized each mouse, with no association to the BV status of the microbiota donor. Although some of the women in the study had adverse birth outcomes, the vaginal microbiota was not predictive of adverse birth outcomes in mice. However, elevated levels of pro-inflammatory cytokines in the uterus of HMA mice were detected during pregnancy. Together, these data outline the potential uses and limitations of HMA mice to elucidate the influence of the vaginal microbiota on health and disease.
- Published
- 2020
- Full Text
- View/download PDF
15. Organic Solute Transporter α-β Protects Ileal Enterocytes From Bile Acid–Induced InjurySummary
- Author
-
Courtney B. Ferrebee, Jianing Li, Jamie Haywood, Kimberly Pachura, Brian S. Robinson, Benjamin H. Hinrichs, Rheinallt M. Jones, Anuradha Rao, and Paul A. Dawson
- Subjects
Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: Ileal bile acid absorption is mediated by uptake via the apical sodium-dependent bile acid transporter (ASBT), and export via the basolateral heteromeric organic solute transporter α-β (OSTα-OSTβ). In this study, we investigated the cytotoxic effects of enterocyte bile acid stasis in Ostα-/- mice, including the temporal relationship between intestinal injury and initiation of the enterohepatic circulation of bile acids. Methods: Ileal tissue morphometry, histology, markers of cell proliferation, gene, and protein expression were analyzed in male and female wild-type and Ostα-/- mice at postnatal days 5, 10, 15, 20, and 30. Ostα-/-Asbt-/- mice were generated and analyzed. Bile acid activation of intestinal Nrf2-activated pathways was investigated in Drosophila. Results: As early as day 5, Ostα-/- mice showed significantly increased ileal weight per length, decreased villus height, and increased epithelial cell proliferation. This correlated with premature expression of the Asbt and induction of bile acid–activated farnesoid X receptor target genes in neonatal Ostα-/- mice. Expression of reduced nicotinamide adenine dinucleotide phosphate oxidase-1 and Nrf2–anti-oxidant responsive genes were increased significantly in neonatal Ostα-/- mice at these postnatal time points. Bile acids also activated Nrf2 in Drosophila enterocytes and enterocyte-specific knockdown of Nrf2 increased sensitivity of flies to bile acid–induced toxicity. Inactivation of the Asbt prevented the changes in ileal morphology and induction of anti-oxidant response genes in Ostα-/- mice. Conclusions: Early in postnatal development, loss of Ostα leads to bile acid accumulation, oxidative stress, and a restitution response in ileum. In addition to its essential role in maintaining bile acid homeostasis, Ostα-Ostβ functions to protect the ileal epithelium against bile acid–induced injury. NCBI Gene Expression Omnibus: GSE99579. Keywords: Ileum, Reactive Oxygen Species, Nuclear Factor Erythroid-Derived 2-Like 2, Neonate, Drosophila
- Published
- 2018
- Full Text
- View/download PDF
16. Lactobacilli Modulate Epithelial Cytoprotection through the Nrf2 Pathway
- Author
-
Rheinallt M. Jones, Chirayu Desai, Trevor M. Darby, Liping Luo, Alexandra A. Wolfarth, Christopher D. Scharer, Courtney S. Ardita, April R. Reedy, Erin S. Keebaugh, and Andrew S. Neish
- Subjects
Biology (General) ,QH301-705.5 - Abstract
An optimal gut microbiota influences many beneficial processes in the metazoan host. However, the molecular mechanisms that mediate and function in symbiont-induced host responses have not yet been fully characterized. Here, we report that cellular ROS enzymatically generated in response to contact with lactobacilli in both mice and Drosophila has salutary effects against exogenous insults to the intestinal epithelium via the activation of Nrf2 responsive cytoprotective genes. These data show that the xenobiotic-inducible Nrf2 pathway participates as a signaling conduit between the prokaryotic symbiont and the eukaryotic host. Indeed, our data imply that the capacity of lactobacilli to induce redox signaling in epithelial cells is a highly conserved hormetic adaptation to impel cellular conditioning to exogenous biotic stimuli. These data also highlight the role the microbiota plays in eukaryotic cytoprotective pathways and may have significant implications in the characterization of a eubiotic microbiota.
- Published
- 2015
- Full Text
- View/download PDF
17. The gut microbiota is a transmissible determinant of skeletal maturation
- Author
-
Abdul Malik Tyagi, Trevor M Darby, Emory Hsu, Mingcan Yu, Subhashis Pal, Hamid Dar, Jau-Yi Li, Jonathan Adams, Rheinallt M Jones, and Roberto Pacifici
- Subjects
microbiome ,bone ,T cells ,bone structure ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Genetic factors account for the majority of the variance of human bone mass, but the contribution of non-genetic factors remains largely unknown. By utilizing maternal/offspring transmission, cohabitation, or fecal material transplantation (FMT) studies, we investigated the influence of the gut microbiome on skeletal maturation. We show that the gut microbiome is a communicable regulator of bone structure and turnover in mice. In addition, we found that the acquisition of a specific bacterial strain, segmented filamentous bacteria (SFB), a gut microbe that induces intestinal Th17 cell expansion, was sufficient to negatively impact skeletal maturation. These findings have significant translational implications, as the identification of methods or timing of microbiome transfer may lead to the development of bacteriotherapeutic interventions to optimize skeletal maturation in humans. Moreover, the transfer of SFB-like microbes capable of triggering the expansion of human Th17 cells during therapeutic FMT procedures could lead to significant bone loss in fecal material recipients.
- Published
- 2021
- Full Text
- View/download PDF
18. Lactobacillus rhamnosus GG Orchestrates an Antitumor Immune Response
- Author
-
Rheinallt M. Jones, Bejan Saeedi, Richard U. Eboka, Trevor Darby, Joshua A. Owens, Crystal Naudin, Sarah Hunter-Chang, Maria E. Barbian, Lauren Askew, and Brian S. Robinson
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Cell Communication ,RC799-869 ,CD8-Positive T-Lymphocytes ,LGG, Lactobacillus rhamnosus GG ,Mice ,DC, dendritic cell ,PCR, polymerase chain reaction ,0302 clinical medicine ,Neoplasms ,Medicine ,Intestinal Mucosa ,Original Research ,Cancer ,PD-1, anti programmed cell death protein 1 ,biology ,Lacticaseibacillus rhamnosus ,LGG ,Gastroenterology ,Human microbiome ,CFSE, carboxyfluorescein succinimidyl ester ,HBSS, Hank’s buffered salt solution ,MSI, microsatellite instability ,Diseases of the digestive system. Gastroenterology ,FCM, flow cytometry ,Tumor Burden ,CRC ,mLn, mesenteric lymph node ,CRC, colorectal cancer ,MSH, MutS Homolog ,030211 gastroenterology & hepatology ,Signal Transduction ,TLR, Toll-like receptor ,Colon ,BMDC, bone-marrow–derived dendritic cell ,PBS, phosphate-buffered saline ,DSS-AOM, dextran sulfate sodium–azoxymethane ,Immunomodulation ,03 medical and health sciences ,Immune system ,FBS, fetal bovine serum ,Lactobacillus rhamnosus ,Animals ,Humans ,Microbiome ,CXCL, chemokine ligands ,Host Microbial Interactions ,Hepatology ,CD8 T Cells ,business.industry ,Probiotics ,Immunity ,Dendritic Cells ,Immunotherapy ,Dendritic cell ,biology.organism_classification ,medicine.disease ,WT, wild-type ,Toll-Like Receptor 2 ,Gastrointestinal Microbiome ,IL, interleukin ,Disease Models, Animal ,TLR2 ,030104 developmental biology ,Immunology ,business ,BC, Bacillus cereus - Abstract
Background & Aims In colorectal cancer, approximately 95% of patients are refractory to immunotherapy because of low antitumor immune responses. Therefore, there is an exigent need to develop treatments that increase antitumor immune responses and decrease tumor burden to enhance immunotherapy. Methods The gut microbiome has been described as a master modulator of immune responses. We administered the human commensal, Lactobacillus rhamnosus GG (LGG), to mice and characterized the changes in the gut immune landscape. Because the presence of lactobacilli in the gut microbiome has been linked with decreased tumor burden and antitumor immune responses, we also supplemented a genetic and a chemical model of murine intestinal cancer with LGG. For clinical relevance, we therapeutically administered LGG after tumors had formed. We also tested for the requirement of CD8 T cells in LGG-mediated modulation of gut tumor burden. Results We detected increased colonic CD8 T-cell responses specifically in LGG-supplemented mice. The CD8 T-cell induction was dependent on dendritic cell activation mediated via Toll-like receptor-2, thereby describing a novel mechanism in which a member of the human microbiome induces an intestinal CD8 T-cell response. We also show that LGG decreased tumor burden in the murine gut cancer models by a CD8 T-cell–dependent manner. Conclusions These data support the potential use of LGG to augment antitumor immune responses in colorectal cancer patients and ultimately for increasing the breadth and efficacy of immunotherapy., Graphical abstract
- Published
- 2021
19. Parathyroid hormone–dependent bone formation requires butyrate production by intestinal microbiota
- Author
-
Rheinallt M. Jones, Jau-Yi Li, Subhashis Pal, Mingcan Yu, Roberto Pacifici, M. Neale Weitzmann, Jonathan Adams, Abdul Malik Tyagi, and Hamid Y. Dar
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Bone disease ,Anabolism ,Parathyroid hormone ,Butyrate ,CD8-Positive T-Lymphocytes ,T-Lymphocytes, Regulatory ,Bone resorption ,Receptors, G-Protein-Coupled ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,Internal medicine ,medicine ,Animals ,Mice, Knockout ,Chemistry ,Wnt signaling pathway ,General Medicine ,medicine.disease ,Gastrointestinal Microbiome ,Wnt Proteins ,Butyrates ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Parathyroid Hormone ,030220 oncology & carcinogenesis ,Bone marrow ,CD8 - Abstract
Parathyroid hormone (PTH) is a critical regulator of skeletal development that promotes both bone formation and bone resorption. Using microbiota depletion by wide-spectrum antibiotics and germ-free (GF) female mice, we showed that the microbiota was required for PTH to stimulate bone formation and increase bone mass. Microbiota depletion lowered butyrate levels, a metabolite responsible for gut-bone communication, while reestablishment of physiologic levels of butyrate restored PTH-induced anabolism. The permissive activity of butyrate was mediated by GPR43 signaling in dendritic cells and by GPR43-independent signaling in T cells. Butyrate was required for PTH to increase the number of bone marrow (BM) regulatory T cells (Tregs). Tregs stimulated production of the osteogenic Wnt ligand Wnt10b by BM CD8+ T cells, which activated Wnt-dependent bone formation. Together, these data highlight the role that butyrate produced by gut luminal microbiota plays in triggering regulatory pathways, which are critical for the anabolic action of PTH in bone.
- Published
- 2020
20. Lactococcus lactis sb. cremoris orchestrates signal events in the gut epithelium via TLR2 to promote tissue restitution
- Author
-
Crystal R. Naudin, Joshua A. Owens, Lauren C. Askew, Ramsha Nabihah Khan, Christopher D. Scharer, Jason D. Matthews, Liping Luo, Jiyoung Kim, April R. Reedy, Maria E. Barbian, and Rheinallt M. Jones
- Abstract
The use of beneficial bacteria to promote gastrointestinal heath is widely practiced, however, the mechanisms whereby many of these microbes elicit their beneficial effects remain elusive. Previously, we conducted a screen for the discovery of novel beneficial microbes and identified the potent cytoprotective effects of a strain of Lactococcus lactis subsp. cremoris. Here, we show that dietary supplementation with L. lactis subsp. cremoris induced transcript enrichment of a set of genes within the colon whose functions are associated with host cell and microbe interactions. Specifically, L. lactis subsp. cremoris induced the expression of tlr2, which we show was required for L. lactis subsp. cremoris to elicit its beneficial effects on the intestine. L. lactis subsp. cremoris did not confer beneficial effects in mice deficient in TLR-2, or deficient in its adaptor protein Myd88 in chronic gut injury models. In addition to cytoprotection, culture supernatant from L. lactis subsp. cremoris accelerated epithelial migration in a cultured epithelial cell scratch wound assay; and effect that was abrogated by a TLR-2 antagonist. Furthermore, L. lactis subsp. cremoris accelerated epithelial tissue restitution following the infliction of a colonic wound biopsy in a TLR-2 and Myd88-dependent manner. Within colonic wounds, L. lactis subsp. cremoris induced the activation of signaling pathways that function in tissue restitution following injury, including the ERK signaling pathway, and of focal adhesion complex (FAC) proteins. Together, these data demonstrate that L. lactis subsp. cremoris signals via the TLR2/MyD88-axis to confer cytoprotection and accelarated tissue restituion in the gut epithelium. These data point to evolving adaptations where beneficial gut microbes moduate innate immune signaling to excert positive influnces on host physiology.
- Published
- 2021
21. The gut-bone axis: how bacterial metabolites bridge the distance
- Author
-
Rheinallt M. Jones, Mario M. Zaiss, Roberto Pacifici, and Georg Schett
- Subjects
0301 basic medicine ,Review ,Gut flora ,digestive system ,Systemic circulation ,Bone health ,03 medical and health sciences ,0302 clinical medicine ,Bone cell ,Animals ,Humans ,Involution (medicine) ,biology ,Probiotics ,Gastrointestinal Microbiome ,General Medicine ,Fatty Acids, Volatile ,biology.organism_classification ,Gut microbiome ,Cell biology ,Bone Diseases, Metabolic ,Prebiotics ,030104 developmental biology ,030220 oncology & carcinogenesis ,Microbiota composition - Abstract
The gut microbiome is a key regulator of bone health that affects postnatal skeletal development and skeletal involution. Alterations in microbiota composition and host responses to the microbiota contribute to pathological bone loss, while changes in microbiota composition that prevent, or reverse, bone loss may be achieved by nutritional supplements with prebiotics and probiotics. One mechanism whereby microbes influence organs of the body is through the production of metabolites that diffuse from the gut into the systemic circulation. Recently, short-chain fatty acids (SCFAs), which are generated by fermentation of complex carbohydrates, have emerged as key regulatory metabolites produced by the gut microbiota. This Review will focus on the effects of SCFAs on the musculoskeletal system and discuss the mechanisms whereby SCFAs regulate bone cells.
- Published
- 2019
22. Antenatal Dietary Butyrate Supplementation Reduces Postnatal Gastrointestinal Injury in a Murine Model of Colitis
- Author
-
Rheinallt M. Jones, Maria E. Barbian, Patricia W. Denning, Ravi Mangal Patel, and Joshua A. Owens
- Subjects
chemistry.chemical_classification ,medicine.medical_specialty ,Offspring ,business.industry ,Fatty acid ,Gastrointestinal Injury ,Butyrate ,medicine.disease ,Endocrinology ,chemistry ,Murine model ,Internal medicine ,medicine ,Ingestion ,Colitis ,business ,Barrier function - Abstract
Purpose: Determine whether optimizing the antenatal diet, with either direct or indirect butyrate supplementation, reduces the severity of intestinal injury in offspring with colitis. Butyrate is a short-chain fatty acid that has been shown to enhance intestinal barrier function, regulate intestinal mucosal immunity and reduce intestinal inflammation. Butyrate levels can be increased by ingestion of butyrate, a HF diet or certain probiotics. Therefore, we hypothesize that consuming a diet that increases butyrate levels prenatally will reduce the severity of intestinal injury in offspring with colitis. Methods: Mating pairs of C57BL/6 mice were fed either 1% butyrate, high fiber (HF) diet or probiotics with or without a …
- Published
- 2021
23. The gut microbiota is a transmissible determinant of skeletal maturation
- Author
-
Rheinallt M. Jones, Hamid Y. Dar, Subhashis Pal, Emory Hsu, Roberto Pacifici, Jau-Yi Li, Jonathan Adams, Mingcan Yu, Trevor Darby, and Abdul Malik Tyagi
- Subjects
0301 basic medicine ,bone structure ,Mouse ,QH301-705.5 ,Offspring ,Science ,Segmented filamentous bacteria ,Regulator ,T cells ,microbiome ,Biology ,Gut flora ,bone ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Feces ,Mice ,0302 clinical medicine ,Animals ,Microbiome ,Biology (General) ,Skeleton ,General Immunology and Microbiology ,General Neuroscience ,General Medicine ,Fecal Microbiota Transplantation ,biology.organism_classification ,Cell biology ,Gastrointestinal Microbiome ,030104 developmental biology ,Skeletal maturation ,Medicine ,Female ,030217 neurology & neurosurgery ,Bone structure ,Research Article - Abstract
Genetic factors account for the majority of the variance of human bone mass, but the contribution of non-genetic factors remains largely unknown. By utilizing maternal/offspring transmission, cohabitation, or fecal material transplantation (FMT) studies, we investigated the influence of the gut microbiome on skeletal maturation. We show that the gut microbiome is a communicable regulator of bone structure and turnover in mice. In addition, we found that the acquisition of a specific bacterial strain, segmented filamentous bacteria (SFB), a gut microbe that induces intestinal Th17 cell expansion, was sufficient to negatively impact skeletal maturation. These findings have significant translational implications, as the identification of methods or timing of microbiome transfer may lead to the development of bacteriotherapeutic interventions to optimize skeletal maturation in humans. Moreover, the transfer of SFB-like microbes capable of triggering the expansion of human Th17 cells during therapeutic FMT procedures could lead to significant bone loss in fecal material recipients.
- Published
- 2021
24. Cruciferous vegetables (Brassica oleracea) confer cytoprotective effects in Drosophila intestines
- Author
-
Rheinallt M. Jones, Dean P. Jones, Liping Luo, Cassandra L. Quave, James T. Lyles, and Ken Liu
- Subjects
0301 basic medicine ,Microbiology (medical) ,NF-E2-Related Factor 2 ,brassica ,Brassica ,RC799-869 ,Protective Agents ,medicine.disease_cause ,Microbiology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Vegetables ,medicine ,Animals ,Drosophila Proteins ,Food science ,nrf2 ,Drosophila ,Indole test ,biology ,Plant Extracts ,Cruciferous vegetables ,Brief Report ,Gastroenterology ,drosophila ,Diseases of the digestive system. Gastroenterology ,biology.organism_classification ,Cytoprotection ,Intestines ,Oxidative Stress ,030104 developmental biology ,Infectious Diseases ,indole ,chemistry ,Larva ,Brassica oleracea ,030211 gastroenterology & hepatology ,leaky-gut ,Oxidative stress ,Sulforaphane - Abstract
Varieties and cultivars of the cruciferous vegetable Brassica oleracea are widely presumed to elicit positive influences on mammalian health and disease, particularly related to their indole and sulforaphane content. However, there is a considerable gap in knowledge regarding the mechanisms whereby these plant-derived molecules elicit their beneficial effects on the host. In this study, we examined the chemical variation between B. oleracea varieties and evaluated their capacity to both activate Nrf2 in the Drosophila intestine and elicit cytoprotection. Ten types of edible B. oleracea were purchased and B. macrocarpa was wild collected. Fresh material was dried, extracted by double maceration and green kale was also subjected to anaerobic fermentation before processing. Untargeted metabolomics was used to perform Principal Component Analysis. Targeted mass spectral analysis determined the presence of six indole species and quantified indole. Extracts were tested for their capacity to activate Nrf2 in the Drosophila intestine in third instar Drosophila larvae. Cytoprotective effects were evaluated using a paraquat-induced oxidative stress gut injury model. A “Smurf” assay was used to determine protective capacity against a chemically induced leaky gut. Extracts of Brussels sprouts and broccoli activated Nrf2 and protected against paraquat-induced damage and leaky gut. Lacto-fermented kale showed a cytoprotective effect, increasing survival by 20% over the non-fermented extract, but did not protect against leaky gut. The protective effects observed do not directly correlate with indole content, suggesting involvement of multiple compounds and a synergistic mechanism.
- Published
- 2021
25. At the heart of microbial conversations: endocannabinoids and the microbiome in cardiometabolic risk
- Author
-
Joshua A. Owens, Rheinallt M. Jones, Crystal Naudin, Maria E. Barbian, Ramsha Khan, and Kristal M. Maner-Smith
- Subjects
0301 basic medicine ,Microbiology (medical) ,microbiome ,Review ,RC799-869 ,Biology ,Bioinformatics ,Microbiology ,metabolic syndrome ,03 medical and health sciences ,0302 clinical medicine ,Metabolic Diseases ,Risk Factors ,medicine ,Animals ,Humans ,Microbiome ,Receptors, Cannabinoid ,Abdominal obesity ,Cardiometabolic risk ,Host Microbial Interactions ,Probiotics ,cardiovascular ,Gastroenterology ,endocannabinoid ,Diseases of the digestive system. Gastroenterology ,medicine.disease ,Endocannabinoid system ,Gastrointestinal Microbiome ,030104 developmental biology ,Infectious Diseases ,sexual dimorphism ,030211 gastroenterology & hepatology ,Metabolic syndrome ,medicine.symptom ,Dyslipidemia ,probiotic ,Endocannabinoids - Abstract
Cardiometabolic syndrome encompasses intertwined risk factors such as hypertension, dyslipidemia, elevated triglycerides, abdominal obesity, and other maladaptive metabolic and inflammatory aberrations. As the molecular mechanisms linking cardiovascular disease and metabolic disorders are investigated, endocannabinoids have emerged as molecules of interest. The endocannabinoid system (ECS) of biologically active lipids has been implicated in several conditions, including chronic liver disease, osteoporosis, and more recently in cardiovascular diseases. The gut microbiome is a major regulator of inflammatory and metabolic signaling in the host, and if disrupted, has the potential to drive metabolic and cardiovascular diseases. Extensive studies have unraveled the impact of the gut microbiome on host physiology, with recent reports showing that gut microbes exquisitely control the ECS, with significant influences on host metabolic and cardiac health. In this review, we outline how modulation of the gut microbiome affects host metabolism and cardiovascular health via the ECS, and how these findings could be exploited as novel therapeutic targets for various metabolic and cardiac diseases.
- Published
- 2021
26. Author response: The gut microbiota is a transmissible determinant of skeletal maturation
- Author
-
Jonathan Adams, Abdul Malik Tyagi, Rheinallt M. Jones, Jau-Yi Li, Emory Hsu, Trevor Darby, Mingcan Yu, Hamid Y. Dar, Roberto Pacifici, and Subhashis Pal
- Subjects
Skeletal maturation ,biology ,Gut flora ,biology.organism_classification ,Cell biology - Published
- 2020
27. Immune Cell Isolation from Murine Intestine for Antibody Array Analysis
- Author
-
Rheinallt M. Jones and Joshua A. Owens
- Subjects
0301 basic medicine ,Antibody microarray ,Host (biology) ,Peyer's patch ,Pathogenic bacteria ,Disease ,Biology ,medicine.disease_cause ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Antigen ,030220 oncology & carcinogenesis ,medicine ,Intraepithelial lymphocyte - Abstract
Gut mucosal immune cells play an essential role in health due to their ability to orchestrate host signaling events in response to exogenous antigens. These antigens may originate from microorganisms including viruses, commensal or pathogenic bacteria, or single-celled eukaryotes, as well as from dietary foodstuff-derived proteins or products. A critical technological capacity to understand host responses to antigens is the ability to efficiently isolate and functionally characterize immune cells from intestinal tissues. Additionally, after characterization, it is of paramount importance to understand the exact functions of these immune cells under different disease states or genetic variables. Here, we outline methods for immune cell isolation from murine small and large intestines with the goal of undertaking a functional analysis of isolated cell types using antibody array platforms.
- Published
- 2020
28. Immune Cell Isolation from Murine Intestine for Antibody Array Analysis
- Author
-
Joshua A, Owens and Rheinallt M, Jones
- Subjects
Immunoassay ,Mice ,Protein Array Analysis ,Animals ,Lymphocytes ,Intestinal Mucosa ,Flow Cytometry - Abstract
Gut mucosal immune cells play an essential role in health due to their ability to orchestrate host signaling events in response to exogenous antigens. These antigens may originate from microorganisms including viruses, commensal or pathogenic bacteria, or single-celled eukaryotes, as well as from dietary foodstuff-derived proteins or products. A critical technological capacity to understand host responses to antigens is the ability to efficiently isolate and functionally characterize immune cells from intestinal tissues. Additionally, after characterization, it is of paramount importance to understand the exact functions of these immune cells under different disease states or genetic variables. Here, we outline methods for immune cell isolation from murine small and large intestines with the goal of undertaking a functional analysis of isolated cell types using antibody array platforms.
- Published
- 2020
29. A Human Microbiota-Associated Murine Model for Assessing the Impact of the Vaginal Microbiota on Pregnancy Outcomes
- Author
-
David VanInsberghe, Anne L. Dunlop, Andrew S. Neish, Rheinallt M. Jones, Alexandra A. Wolfarth, Elizabeth J. Corwin, and Taylor M Smith
- Subjects
0301 basic medicine ,Microbiology (medical) ,humanization ,030106 microbiology ,Immunology ,lcsh:QR1-502 ,Disease ,bacterial vaginosis (BV) ,Microbiology ,lcsh:Microbiology ,03 medical and health sciences ,Mice ,Cellular and Infection Microbiology ,Pregnancy ,medicine ,Animals ,Humans ,Microbiome ,Pregnancy outcomes ,Original Research ,vaginal microbiota ,business.industry ,Microbiota ,Human microbiome ,Pregnancy Outcome ,Vaginosis, Bacterial ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Infectious Diseases ,Murine model ,inflammation ,Vagina ,Female ,Nugent score ,Bacterial vaginosis ,business - Abstract
Disease states are often linked to large scale changes in microbial community structure that obscure the contributions of individual microbes to disease. Establishing a mechanistic understanding of how microbial community structure contribute to certain diseases, however, remains elusive thereby limiting our ability to develop successful microbiome-based therapeutics. Human microbiota-associated (HMA) mice have emerged as a powerful approach for directly testing the influence of microbial communities on host health and disease, with the transfer of disease phenotypes from humans to germ-free recipient mice widely reported. We developed a HMA mouse model of the human vaginal microbiota to interrogate the effects of Bacterial Vaginosis (BV) on pregnancy outcomes. We collected vaginal swabs from 19 pregnant African American women with and without BV (diagnosed per Nugent score) to colonize female germ-free mice and measure its impact on birth outcomes. There was considerable variability in the microbes that colonized each mouse, with no association to the BV status of the microbiota donor. Although some of the women in the study had adverse birth outcomes, the vaginal microbiota was not predictive of adverse birth outcomes in mice. However, elevated levels of pro-inflammatory cytokines in the uterus of HMA mice were detected during pregnancy. Together, these data outline the potential uses and limitations of HMA mice to elucidate the influence of the vaginal microbiota on health and disease.
- Published
- 2020
- Full Text
- View/download PDF
30. Indoles from the commensal microbiota act via the AHR and IL-10 to tune the cellular composition of the colonic epithelium during aging
- Author
-
Domonica N. Powell, Alexis Bretin, Rheinallt M. Jones, Daniel Kalman, Alyson Swimm, Andrew T. Gewirtz, and Robert Sonowal
- Subjects
0301 basic medicine ,Male ,Aging ,Indoles ,medicine.medical_treatment ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Secretion ,Mice, Knockout ,Goblet cell ,Mice, Inbred BALB C ,Multidisciplinary ,biology ,Bacteria ,Chemistry ,Interleukins ,Microbiota ,Cell Differentiation ,Epithelial Cells ,Biological Sciences ,Aryl hydrocarbon receptor ,Intestinal epithelium ,Mucus ,Epithelium ,Cell biology ,Interleukin-10 ,Mice, Inbred C57BL ,Interleukin 10 ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Receptors, Aryl Hydrocarbon ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Goblet Cells ,Signal Transduction - Abstract
The intestinal epithelium is a highly dynamic structure that rejuvenates in response to acute stressors and can undergo alterations in cellular composition as animals age. The microbiota, acting via secreted factors related to indole, appear to regulate the sensitivity of the epithelium to stressors and promote epithelial repair via IL-22 and type I IFN signaling. As animals age, the cellular composition of the intestinal epithelium changes, resulting in a decreased proportion of goblet cells in the colon. We show that colonization of young or geriatric mice with bacteria that secrete indoles and various derivatives or administration of the indole derivative indole-3 aldehyde increases proliferation of epithelial cells and promotes goblet cell differentiation, reversing an effect of aging. To induce goblet cell differentiation, indole acts via the xenobiotic aryl hydrocarbon receptor to increase expression of the cytokine IL-10. However, the effects of indoles on goblet cells do not depend on type I IFN or on IL-22 signaling, pathways responsible for protection against acute stressors. Thus, indoles derived from the commensal microbiota regulate intestinal homeostasis, especially during aging, via mechanisms distinct from those used during responses to acute stressors. Indoles may have utility as an intervention to limit the decline of barrier integrity and the resulting systemic inflammation that occurs with aging.
- Published
- 2020
31. Galectin-9 Is a Novel Regulator of Epithelial Restitution
- Author
-
Nourine Ahmed, Rheinallt M. Jones, Sean R. Stowell, Andrew S. Neish, Bejan Saeedi, Connie M. Arthur, Josh Owens, Liping Luo, Crystal Naudin, and Brian S. Robinson
- Subjects
0301 basic medicine ,Colon ,Galectins ,Regulator ,Context (language use) ,Endogeny ,Biology ,Inflammatory bowel disease ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Cell Movement ,medicine ,Animals ,Humans ,Intestinal Mucosa ,Galectin ,Cell Proliferation ,Mice, Knockout ,Cell migration ,medicine.disease ,Colitis ,Cell biology ,030104 developmental biology ,030220 oncology & carcinogenesis ,Homeostasis - Abstract
Increasingly, the s-galactoside binding lectins, termed galectins, are being recognized as critical regulators of cell function and organismal homeostasis. Within the context of the mucosal surface, galectins are established regulators of innate and adaptive immune responses, microbial populations, and several critical epithelial functions, including cell migration, proliferation, and response to injury. However, given their complex tissue distribution and expression patterns, the role within specific processes remains poorly understood. We took a genetic approach to understand the role of endogenous galectin-9 (Gal-9), a mucosal galectin that has been linked to inflammatory bowel disease, within the context of the murine intestine. We found that Gal-9–deficient (Gal9−/−) animals show increased sensitivity to chemically induced colitis and impaired proliferation in the setting of acute injury. Moreover, our results show that Gal9−/−-derived enteroids show impaired growth ex vivo. Consistent with a model in which endogenous Gal-9 controls epithelial growth and repair, we found that Gal9−/− animals showed increased sensitivity to intestinal challenge in multiple models of epithelial injury, including acute irradiation injury and ectopic wound biopsies. Finally, our results show that regenerating crypts from patient biopsy specimens show increased expression of Gal-9, indicating these processes may be conserved in humans. Taken together, these studies implicate Gal-9 in the regulation of cellular proliferation and epithelial restitution after intestinal epithelial injury.
- Published
- 2020
32. Microbial metabolite delta-valerobetaine is a diet-dependent obesogen
- Author
-
Ken H. Liu, Joshua A. Owens, Bejan Saeedi, Catherine E. Cohen, Moriah P. Bellissimo, Crystal Naudin, Trevor Darby, Samuel Druzak, Kristal Maner-Smith, Michael Orr, Xin Hu, Jolyn Fernandes, Mary Catherine Camacho, Sarah Hunter-Chang, David VanInsberghe, Chunyu Ma, Thota Ganesh, Samantha M. Yeligar, Karan Uppal, Young-Mi Go, Jessica A. Alvarez, Miriam B. Vos, Thomas R. Ziegler, Michael H. Woodworth, Colleen S. Kraft, Rheinallt M. Jones, Eric Ortlund, Andrew S. Neish, and Dean P. Jones
- Subjects
Host Microbial Interactions ,Endocrinology, Diabetes and Metabolism ,Microbiota ,Fatty Acids ,Cell Biology ,Lipid Metabolism ,Gastrointestinal Microbiome ,Mitochondria ,Mice ,Liver ,Diet, Western ,Physiology (medical) ,Internal Medicine ,Animals ,Humans ,Obesity ,Energy Metabolism ,Oxidation-Reduction ,Adiposity - Abstract
Obesity and obesity-related metabolic disorders are linked to the intestinal microbiome. However, the causality of changes in the microbiome-host interaction affecting energy metabolism remains controversial. Here, we show the microbiome-derived metabolite δ-valerobetaine (VB) is a diet-dependent obesogen that is increased with phenotypic obesity and is correlated with visceral adipose tissue mass in humans. VB is absent in germ-free mice and their mitochondria but present in ex-germ-free conventionalized mice and their mitochondria. Mechanistic studies in vivo and in vitro show VB is produced by diverse bacterial species and inhibits mitochondrial fatty acid oxidation through decreasing cellular carnitine and mitochondrial long-chain acyl-coenzyme As. VB administration to germ-free and conventional mice increases visceral fat mass and exacerbates hepatic steatosis with a western diet but not control diet. Thus, VB provides a molecular target to understand and potentially manage microbiome-host symbiosis or dysbiosis in diet-dependent obesity.
- Published
- 2020
33. PTH induces bone loss via microbial-dependent expansion of intestinal TNF+ T cells and Th17 cells
- Author
-
Abdul Malik Tyagi, Rheinallt M. Jones, Jonathan Adams, Jau-Yi Li, Mingcan Yu, M. Neale Weitzmann, Roberto Pacifici, and Timothy L. Denning
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Science ,T cell ,General Physics and Astronomy ,Parathyroid hormone ,030209 endocrinology & metabolism ,Biology ,Gut flora ,digestive system ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,lcsh:Science ,Hyperparathyroidism ,Multidisciplinary ,General Chemistry ,medicine.disease ,biology.organism_classification ,CCL20 ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,lcsh:Q ,Tumor necrosis factor alpha ,Bone marrow ,hormones, hormone substitutes, and hormone antagonists ,Homing (hematopoietic) - Abstract
Bone loss is a frequent but not universal complication of hyperparathyroidism. Using antibiotic-treated or germ-free mice, we show that parathyroid hormone (PTH) only caused bone loss in mice whose microbiota was enriched by the Th17 cell-inducing taxa segmented filamentous bacteria (SFB). SFB+ microbiota enabled PTH to expand intestinal TNF+ T and Th17 cells and increase their S1P-receptor-1 mediated egress from the intestine and recruitment to the bone marrow (BM) that causes bone loss. CXCR3-mediated TNF+ T cell homing to the BM upregulated the Th17 chemoattractant CCL20, which recruited Th17 cells to the BM. This study reveals mechanisms for microbiota-mediated gut–bone crosstalk in mice models of hyperparathyroidism that may help predict its clinical course. Targeting the gut microbiota or T cell migration may represent therapeutic strategies for hyperparathyroidism. T cells are involved in the bone loss induced by parathyroid hormone (PTH), but their origin is unknown. Here, the authors show that the intestinal microbiota is required for PTH to induce bone loss and describes mechanisms for microbiota-mediated gut–bone crosstalk in mouse models of hyperparathyroidism.
- Published
- 2020
34. Proteomic analysis of microbial induced redox-dependent intestinal signaling
- Author
-
Jason D. Matthews, Benjamin H. Hinrichs, Caroline Addis, Huixia Wu, Rheinallt M. Jones, Trevor Darby, Brian S. Robinson, Andrew S. Neish, Dean P. Jones, April R. Reedy, and Young-Mi Go
- Subjects
0301 basic medicine ,Proteomics ,Proteome ,Clinical Biochemistry ,RAC1 ,Review Article ,medicine.disease_cause ,Biochemistry ,Models, Biological ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Tandem Mass Spectrometry ,medicine ,Intestinal Mucosa ,Cytoskeleton ,lcsh:QH301-705.5 ,chemistry.chemical_classification ,lcsh:R5-920 ,Reactive oxygen species ,Organic Chemistry ,Computational Biology ,Cell biology ,Oxidative Stress ,030104 developmental biology ,lcsh:Biology (General) ,chemistry ,Signal transduction ,lcsh:Medicine (General) ,Reactive Oxygen Species ,Oxidation-Reduction ,030217 neurology & neurosurgery ,Oxidative stress ,Cysteine ,Signal Transduction - Abstract
Intestinal homeostasis is regulated in-part by reactive oxygen species (ROS) that are generated in the colonic mucosa following contact with certain lactobacilli. Mechanistically, ROS can modulate protein function through the oxidation of cysteine residues within proteins. Recent advances in cysteine labeling by the Isotope Coded Affinity Tags (ICATs) technique has facilitated the identification of cysteine thiol modifications in response to stimuli. Here, we used ICATs to map the redox protein network oxidized upon initial contact of the colonic mucosa with Lactobacillus rhamnosus GG (LGG). We detected significant LGG-specific redox changes in over 450 proteins, many of which are implicated to function in cellular processes such as endosomal trafficking, epithelial cell junctions, barrier integrity, and cytoskeleton maintenance and formation. We particularly noted the LGG-specific oxidation of Rac1, which is a pleiotropic regulator of many cellular processes. Together, these data reveal new insights into lactobacilli-induced and redox-dependent networks involved in intestinal homeostasis.
- Published
- 2018
35. Osteomicrobiology: The influence of gut microbiota on bone in health and disease
- Author
-
Roberto Pacifici, Rheinallt M. Jones, and Jennifer G. Mulle
- Subjects
0301 basic medicine ,Histology ,Physiology ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,Disease ,Biology ,Gut flora ,biology.organism_classification ,Bone and Bones ,Gastrointestinal Microbiome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Estrogen ,030220 oncology & carcinogenesis ,Immunology ,medicine ,Animals ,Humans - Published
- 2018
36. Organic Solute Transporter α-β Protects Ileal Enterocytes From Bile Acid–Induced Injury
- Author
-
Anuradha Rao, Rheinallt M. Jones, Courtney B. Ferrebee, Benjamin H. Hinrichs, Jianing Li, Brian S. Robinson, Paul A. Dawson, Kimberly Pachura, and Jamie Haywood
- Subjects
0301 basic medicine ,Gene knockdown ,Hepatology ,Bile acid ,Chemistry ,medicine.drug_class ,Enterocyte ,Gastroenterology ,Ileum ,Transporter ,digestive system ,Molecular biology ,Epithelium ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,lcsh:Diseases of the digestive system. Gastroenterology ,Farnesoid X receptor ,lcsh:RC799-869 ,Enterohepatic circulation - Abstract
Background & Aims: Ileal bile acid absorption is mediated by uptake via the apical sodium-dependent bile acid transporter (ASBT), and export via the basolateral heteromeric organic solute transporter α-β (OSTα-OSTβ). In this study, we investigated the cytotoxic effects of enterocyte bile acid stasis in Ostα-/- mice, including the temporal relationship between intestinal injury and initiation of the enterohepatic circulation of bile acids. Methods: Ileal tissue morphometry, histology, markers of cell proliferation, gene, and protein expression were analyzed in male and female wild-type and Ostα-/- mice at postnatal days 5, 10, 15, 20, and 30. Ostα-/-Asbt-/- mice were generated and analyzed. Bile acid activation of intestinal Nrf2-activated pathways was investigated in Drosophila. Results: As early as day 5, Ostα-/- mice showed significantly increased ileal weight per length, decreased villus height, and increased epithelial cell proliferation. This correlated with premature expression of the Asbt and induction of bile acid–activated farnesoid X receptor target genes in neonatal Ostα-/- mice. Expression of reduced nicotinamide adenine dinucleotide phosphate oxidase-1 and Nrf2–anti-oxidant responsive genes were increased significantly in neonatal Ostα-/- mice at these postnatal time points. Bile acids also activated Nrf2 in Drosophila enterocytes and enterocyte-specific knockdown of Nrf2 increased sensitivity of flies to bile acid–induced toxicity. Inactivation of the Asbt prevented the changes in ileal morphology and induction of anti-oxidant response genes in Ostα-/- mice. Conclusions: Early in postnatal development, loss of Ostα leads to bile acid accumulation, oxidative stress, and a restitution response in ileum. In addition to its essential role in maintaining bile acid homeostasis, Ostα-Ostβ functions to protect the ileal epithelium against bile acid–induced injury. NCBI Gene Expression Omnibus: GSE99579. Keywords: Ileum, Reactive Oxygen Species, Nuclear Factor Erythroid-Derived 2-Like 2, Neonate, Drosophila
- Published
- 2018
37. sal Genes determining the catabolism of salicylate esters are part of a supraoperonic cluster of catabolic genes in acinetobacter sp. strain ADP1
- Author
-
Jones, Rheinallt M. Jones, Pagmantidis, Vassilis, and Williams, Peter A.
- Subjects
Aromatic compounds -- Physiological aspects ,Metabolism -- Physiological aspects ,Operons -- Genetic aspects ,Bacteria -- Physiological aspects ,Biological sciences - Abstract
Research demonstrates that a 5 kilobasepair of the ben-cat supraoperonic cluster past the are genes include genes involved in the catabolism of alkyl salicylates. Data further show that ethyl salicylate and salicylate are processed by the beta-ketoadipate pathway.
- Published
- 2000
38. Probiotic Supplements Accelerate Bone Repair and Prevent Systemic Bone Loss Following Femoral Fracture (FS09-04-19)
- Author
-
Rheinallt M. Jones, Lorenzo M. Fernandes, Joseph L. Roberts, Hicham Drissi, and Trevor Darby
- Subjects
Osteoplasty ,medicine.medical_specialty ,Nutrition and Dietetics ,Aging and Chronic Disease ,business.industry ,Medicine (miscellaneous) ,Bifidobacterium adolescentis ,Bone healing ,Femoral fracture ,medicine.disease ,Surgery ,law.invention ,Osteopenia ,Probiotic ,Trabecular bone ,law ,medicine ,Intestinal bacteria ,business ,Food Science - Abstract
OBJECTIVES: Fracture is a traumatic event that can initiate systemic bone loss and lead to increased future risk of fracture, especially in the elderly. We sought to determine whether modulation of the gut microbiome using a daily probiotic supplement would dampen systemic inflammatory responses to fracture to accelerate bone repair and preserve distant bone sites. METHODS: Twelve-week-old male C57BL/6 J mice were randomly assigned to receive the probiotic Bifidobacterium adolescentis (1 × 10(8) CFU) via a daily oral gavage or PBS vehicle control. After two weeks, experimental mid-diaphyseal femoral fractures were created. Bones were analyzed using micro-CT at 10 and 18-days post-fracture. Midsagittal histological sections of fractured femora were stained with TRAP to quantify osteoclast number within the callus. RNA isolated from the small intestines was analyzed for gene expression of tight junction proteins. Serum levels of 20 cytokines were measured and analyzed using principal components analysis (PCA). RESULTS: Micro-CT of fractured limbs revealed a significant increase in callus bone volume (+28%) at day 18 post-fracture in mice receiving the B. adolescentis supplement. The total number of osteoclasts within the fracture callus at day 18 was also significantly increased (+158%) in the probiotic group suggesting more advanced remodeling of the callus. Micro-CT analyses of the L3 vertebral body, a distant bone site, revealed significantly higher trabecular bone volume at day 10 (+ 22%) and 18 (+11%) post-fracture in the probiotic group. PCA of serum cytokines revealed distinct clustering of the B. adolescentis and PBS groups indicating an anti-inflammatory effect by B. adolescentis. In the small intestines, probiotic supplementation significantly increased gene expression of tight junction proteins occludin, Jam3, ZO-1, and claudin-15 at both day 10 and day 18 post-fracture. CONCLUSIONS: Daily B. adolescentis supplementation not only accelerated fracture healing but also protected against vertebral body bone loss following fracture. These effects are likely a consequence of tightened intestinal barrier integrity, thus preventing systemic inflammation stemming from fracture-induced gut permeability. FUNDING SOURCES: Funding provided by Emory University and the Burroughs Wellcome Fund.
- Published
- 2019
39. Lactobacillus rhamnosus GG-induced Expression of Leptin in the Intestine Orchestrates Epithelial Cell Proliferation
- Author
-
Crystal Naudin, Rheinallt M. Jones, Liping Luo, and Trevor Darby
- Subjects
0301 basic medicine ,Leptin ,Probiotic ,LGG, Lactobacillus rhamnosus GG ,Mice ,0302 clinical medicine ,PCR, polymerase chain reaction ,Intestinal Mucosa ,Cells, Cultured ,Original Research ,Mice, Knockout ,NADPH oxidase ,biology ,Lacticaseibacillus rhamnosus ,Nox, NADPH oxidase ,digestive, oral, and skin physiology ,Gastroenterology ,food and beverages ,ELISA, enzyme-linked immunosorbent assay ,Cell biology ,STAT Transcription Factors ,NOX1 ,Knockout mouse ,Models, Animal ,NADPH Oxidase 1 ,Receptors, Leptin ,030211 gastroenterology & hepatology ,Signal transduction ,Signal Transduction ,Colon ,Primary Cell Culture ,03 medical and health sciences ,ROS, reactive oxygen species ,Lactobacillus rhamnosus ,PBS, phosphate buffered saline ,Nox1 ,Animals ,lcsh:RC799-869 ,LEP-R, leptin receptor ,Cell Proliferation ,Janus Kinases ,Leptin receptor ,Hepatology ,Cell growth ,Probiotics ,Epithelial Cells ,biology.organism_classification ,WT, wild-type ,Lactobacillus ,030104 developmental biology ,biology.protein ,lcsh:Diseases of the digestive system. Gastroenterology ,Reactive Oxygen Species - Abstract
Background & Aims Identifying the functional elements that mediate efficient gut epithelial growth and homeostasis is essential for understanding intestinal health and disease. Many of these processes involve the Lactobacillus-induced generation of reactive oxygen species by NADPH oxidase (Nox1). However, the downstream signaling pathways that respond to Nox1-generated reactive oxygen species and mediate these events have not been described. Methods Wild-type and knockout mice were fed Lactobacillus rhamnosus GG and the transcriptional and cell signaling pathway responses in the colon measured. Corroboration of data generated in mice was done using in organoid tissue culture and in vivo gut injury models. Results Ingestion of L rhamnosus GG induces elevated levels of leptin in the gut epithelia, which as well as functioning in the context of metabolism, has pleiotropic activity as a chemokine that triggers cell proliferation. Consistently, using gut epithelial-specific knockout mice, we show that L rhamnosus GG–induced elevated levels of leptin is dependent on a functional Nox1 protein in the colonic epithelium, and that L rhamnosus GG–induced cell proliferation is dependent on Nox1, leptin, and leptin receptor. We also show that L rhamnosus GG induces the JAK-STAT signaling pathway in the gut in a Nox1, leptin, and leptin receptor–dependent manner. Conclusions These results demonstrate a novel role for leptin in the response to colonization by lactobacilli, where leptin functions in the transduction of signals from symbiotic bacteria to subepithelial compartments, where it modulates intestinal growth and homeostasis., Graphical abstract
- Published
- 2019
40. Neutrophil-Derived Reactive Oxygen Orchestrates Epithelial Cell Signaling Events during Intestinal Repair
- Author
-
Ronen Sumagin, Crystal Naudin, Joshua A. Owens, Benjamin H. Hinrichs, Ashfaqul Alam, Jason D. Matthews, Rheinallt M. Jones, Andrew S. Neish, Bejan Saeedi, and April R. Reedy
- Subjects
0301 basic medicine ,Enzyme complex ,Neutrophils ,Cell junction ,Article ,Pathology and Forensic Medicine ,Focal adhesion ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Animals ,Regeneration ,Intestinal Mucosa ,p21-activated kinases ,Cells, Cultured ,chemistry.chemical_classification ,Mice, Knockout ,Reactive oxygen species ,Wound Healing ,NADPH oxidase ,biology ,Chemistry ,Epithelial Cells ,Cell biology ,Intestines ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,NADPH Oxidase 2 ,biology.protein ,Phosphorylation ,Signal transduction ,Reactive Oxygen Species ,Signal Transduction - Abstract
Recent evidence has demonstrated that reactive oxygen (eg, hydrogen peroxide) can activate host cell signaling pathways that function in repair. We show that mice deficient in their capacity to generate reactive oxygen by the NADPH oxidase 2 holoenzyme, an enzyme complex highly expressed in neutrophils and macrophages, have disrupted capacity to orchestrate signaling events that function in mucosal repair. Similar observations were made for mice after neutrophil depletion, pinpointing this cell type as the source of the reactive oxygen driving oxidation-reduction protein signaling in the epithelium. To simulate epithelial exposure to high levels of reactive oxygen produced by neutrophils and gain new insight into this oxidation-reduction signaling, epithelial cells were treated with hydrogen peroxide, biochemical experiments were conducted, and a proteome-wide screen was performed using isotope-coded affinity tags to detect proteins oxidized after exposure. This analysis implicated signaling pathways regulating focal adhesions, cell junctions, and maintenance of the cytoskeleton. These pathways are also known to act via coordinated phosphorylation events within proteins that constitute the focal adhesion complex, including focal adhesion kinase and Crk-associated substrate. We identified the Rho family small GTP-binding protein Ras-related C3 botulinum toxin substrate 1 and p21 activated kinases 2 as operational in these signaling and localization pathways. These data support the hypothesis that reactive oxygen species from neutrophils can orchestrate epithelial cell-signaling events functioning in intestinal repair.
- Published
- 2019
41. Lactococcus lactis sp. cremoris elicits protection against metabolic changes induced in females by a Western style diet
- Author
-
Rheinallt M. Jones, Bejan Saeedi, Joshua A. Owens, Crystal Naudin, and Trevor Darby
- Subjects
biology ,Lactococcus lactis ,Genetics ,biology.organism_classification ,Molecular Biology ,Biochemistry ,Biotechnology ,Microbiology - Published
- 2019
42. Lactobacilli ‐induced Generation of Reactive Oxygen Species via Formyl Peptide Receptor‐1 (FPR1) Regulates Intestinal Motility in Mice
- Author
-
Rheinallt M. Jones, Malú G. Tansey, Andrew S. Neish, M Ashfaqul Alam, Bindu Chandrasekharan, Shanthi Srinivasan, Bejan Saeedi, and Asma Nusrat
- Subjects
chemistry.chemical_classification ,Reactive oxygen species ,Biochemistry ,Chemistry ,Genetics ,Molecular Biology ,Formyl peptide receptor 1 ,Biotechnology ,Intestinal motility - Published
- 2019
43. Regulation of the Hepatic Antioxidant Response by the Probiotic Lactobacillus rhamnosus GG
- Author
-
Richard U. Eboka, Joshua A. Owens, Bejan Saeedi, Rheinallt M. Jones, Ken Liu, Dean P. Jones, Brian S. Robinson, Andrew S. Neish, Liping Luo, and Trevor Darby
- Subjects
Probiotic ,Lactobacillus rhamnosus ,law ,Genetics ,Antioxidant response element ,Food science ,Biology ,biology.organism_classification ,Molecular Biology ,Biochemistry ,Biotechnology ,law.invention - Published
- 2019
44. Lactobacillus rhamnosus GG‐mediated expression of leptin in the intestine induces cell proliferation
- Author
-
Rheinallt M. Jones, Liping Luo, Trevor Darby, and Crystal Naudin
- Subjects
Lactobacillus rhamnosus ,Cell growth ,Leptin ,Genetics ,Biology ,biology.organism_classification ,Molecular Biology ,Biochemistry ,Molecular biology ,Biotechnology - Published
- 2019
45. Elucidating the cell signaling pathways for epithelial restitution of a novel probiotic
- Author
-
Rheinallt M. Jones, Crystal Naudin, Bejan Saeedi, Joshua A. Owens, and Trevor Darby
- Subjects
Probiotic ,law ,Epithelial restitution ,Genetics ,Biology ,Molecular Biology ,Biochemistry ,Cell signaling pathways ,Biotechnology ,Cell biology ,law.invention - Published
- 2019
46. Anemia Induces Gut Inflammation and Injury in an Animal Model of Preterm Infants
- Author
-
Ashley Bennett, Gretchen A. Cress, Brian S. Robinson, Bejan Saeedi, Martha Sola-Visner, John D. Roback, Michael K. Georgieff, John A. Widness, Demet Nalbant, Rheinallt M. Jones, Andrew S. Neish, Peggy Nopoulos, Cassandra D. Josephson, Nourine A. Kamili, M. Bridget Zimmerman, Connie M. Arthur, Sean R. Stowell, Ravi Mangal Patel, Jason D. Matthews, and Henry A. Feldman
- Subjects
Male ,Anemia ,Immunology ,Population ,Inflammation ,Infant, Premature, Diseases ,030204 cardiovascular system & hematology ,Article ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Intestinal mucosa ,Enterocolitis, Necrotizing ,medicine ,Immunology and Allergy ,Animals ,Humans ,Infant, Very Low Birth Weight ,Intestinal Mucosa ,education ,Barrier function ,Enterocolitis ,education.field_of_study ,business.industry ,Infant, Newborn ,Hematology ,Hypoxia (medical) ,medicine.disease ,Disease Models, Animal ,Necrotizing enterocolitis ,Zonula Occludens-1 Protein ,Female ,medicine.symptom ,business ,Infant, Premature ,030215 immunology - Abstract
Background While very low birth weight (VLBW) infants often require multiple red blood cell transfusions, efforts to minimize transfusion-associated risks have resulted in more restrictive neonatal transfusion practices. However, whether restrictive transfusion strategies limit transfusions without increasing morbidity and mortality in this population remains unclear. Recent epidemiologic studies suggest that severe anemia may be an important risk factor for the development of necrotizing enterocolitis (NEC). However, the mechanism whereby anemia may lead to NEC remains unknown. Study design and methods The potential impact of anemia on neonatal inflammation and intestinal barrier disruption, two well-characterized predisposing features of NEC, was defined by correlation of hemoglobin values to cytokine levels in premature infants and by direct evaluation of intestinal hypoxia, inflammation and gut barrier disruption using a pre-clinical neonatal murine model of phlebotomy-induced anemia (PIA). Results Increasing severity of anemia in the preterm infant correlated with the level of IFN-gamma, a key pro-inflammatory cytokine that may predispose an infant to NEC. Gradual induction of PIA in a pre-clinical model resulted in significant hypoxia throughout the intestinal mucosa, including areas where intestinal macrophages reside. PIA-induced hypoxia significantly increased macrophage pro-inflammatory cytokine levels, while reducing tight junction protein ZO-1 expression and increasing intestinal barrier permeability. Macrophage depletion reversed the impact of anemia on intestinal ZO-1 expression and barrier function. Conclusions Taken together, these results suggest that anemia can increase intestinal inflammation and barrier disruption likely through altered macrophage function, leading to the type of predisposing intestinal injury that may increase the risk for NEC.
- Published
- 2019
47. Vibrio parahaemolyticus VopA Is a Potent Inhibitor of Cell Migration and Apoptosis in the Intestinal Epithelium of Drosophila melanogaster
- Author
-
Brian S. Robinson, Jason D. Matthews, Rheinallt M. Jones, and Liping Luo
- Subjects
0301 basic medicine ,Innate immune system ,Cellular Microbiology: Pathogen-Host Cell Molecular Interactions ,Kinase ,Effector ,Immunology ,Cell migration ,Biology ,Microbiology ,Intestinal epithelium ,Cell biology ,Focal adhesion ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Parasitology ,Signal transduction ,Protein kinase A ,030217 neurology & neurosurgery - Abstract
Animal models have played a key role in providing an understanding of the mechanisms that govern the pathophysiology of intestinal diseases. To expand on the repertoire of organisms available to study enteric diseases, we report on the use of the Drosophila melanogaster model to identify a novel function of an effector protein secreted by Vibrio parahaemolyticus, which is an enteric pathogen found in contaminated seafood. During pathogenesis, V. parahaemolyticus secretes effector proteins that usurp the host’s innate immune signaling pathways, thus allowing the bacterium to evade detection by the innate immune system. One secreted effector protein, VopA, has potent inhibitory effects on mitogen-activated protein kinase (MAPK) signaling pathways via the acetylation of critical residues within the catalytic loops of mitogen-activated protein kinase kinases (MAPKKs). Using the Drosophila model and cultured mammalian cells, we show that VopA also has potent modulating activity on focal adhesion complex (FAC) proteins, where VopA markedly reduced the levels of focal adhesion kinase (FAK) phosphorylation at Ser910, whereas the phosphorylation levels of FAK at Tyr397 and Tyr861 were markedly increased. Cultured cells expressing VopA were also impaired in their ability to migrate and repopulate areas subjected to a scratch wound. Consistently, expression of VopA in Drosophila midgut enterocytes disrupted the normal enterocyte arrangement. Finally, VopA inhibited apoptosis in both Drosophila tissues and mammalian cultured cells. Together, our data show that VopA can alter normal intestinal homeostatic processes to facilitate opportunities for V. parahaemolyticus to prolong infection within the host.
- Published
- 2019
48. Gut Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury
- Author
-
Bejan Saeedi, Ken H. Liu, Brian S. Robinson, Joshua A. Owens, Nusaiba Baker, Dean P. Jones, Richard U. Eboka, Sarah Hunter-Chang, Rheinallt M. Jones, Andrew S. Neish, Trevor Darby, Bindu Chandrasekharan, and Mary C. Camacho
- Subjects
Liver injury ,biology ,Activator (genetics) ,biology.organism_classification ,medicine.disease ,Microbiology ,chemistry.chemical_compound ,Lactobacillus rhamnosus ,chemistry ,Downregulation and upregulation ,Lactobacillus ,medicine ,Microbiome ,Xenobiotic ,Homeostasis - Abstract
The gut microbiome is a dynamic community of microorganisms within the intestinal lumen. Many studies have suggested a role for gut-resident microbes in modulating host health, however the mechanisms by which these communities impact systemic physiology remain largely unknown. In this study, metabolomic and transcriptional profiling of germ-free and conventionalized mouse liver revealed an upregulation of the Nrf2 antioxidant and xenobiotic response in microbiome-replete animals. Using a Drosophila-based screening assay, we identified members of the genus Lactobacillus capable of stimulating Nrf2. Indeed, in both flies and mice, the human commensal Lactobacillus rhamnosus GG (LGG) potently activated Nrf2 in the fat body and liver, respectively, and protected against acetaminophen hepatotoxicity. MS/MS characterization of the portal circulation of LGG-treated mice identified a small molecule activator of Nrf2, 5-methoxyindoleacetic acid (5-MIAA), produced by LGG. Taken together, these data demonstrate a mechanism by which intestinal microbes modulate hepatic homeostasis and susceptibility to oxidative injury.
- Published
- 2019
49. Commensal microbiota induced redox signaling activates proliferative signals in the intestinal stem cell microenvironment
- Author
-
April R. Reedy, Rheinallt M. Jones, Liping Luo, and Andrew S. Neish
- Subjects
medicine.medical_treatment ,Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Stem Cell Niche ,Molecular Biology ,030304 developmental biology ,Progenitor ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,Activator (genetics) ,Stem Cells ,fungi ,food and beverages ,JAK-STAT signaling pathway ,Midgut ,Stem Cells and Regeneration ,Epithelium ,Gastrointestinal Microbiome ,Cell biology ,Drosophila melanogaster ,Enterocytes ,medicine.anatomical_structure ,Cytokine ,chemistry ,Stem cell ,Reactive Oxygen Species ,Oxidation-Reduction ,030217 neurology & neurosurgery ,Lactobacillus plantarum ,Signal Transduction ,Developmental Biology - Abstract
A distinct taxon of the Drosophila microbiota, Lactobacillus plantarum, is capable of stimulating the generation of reactive oxygen species (ROS) within cells, and inducing epithelial cell proliferation. Here we show microbial-induced ROS generation within Drosophila larval stem cell compartments exhibits a distinct spatial distribution. Lactobacilli-induced ROS is strictly excluded from defined midgut compartments that harbor adult midgut progenitor (AMP) cells, forming a functional “ROS sheltered zone” (RSZ). The RSZ is undiscernible in germ-free larvae, but forms following mono-colonization with L. plantarum. L. plantarum is a strong activator of the ROS-sensitive CncC/Nrf2 signaling pathway within enterocytes. Enterocyte-specific activation of CncC stimulated the proliferation of AMPs, demonstrating that pro-proliferative signals are transduced from enterocytes to AMPs. Mechanistically, we show that the cytokine Upd2 is expressed in the gut following L. plantarum colonization in a CncC dependent fashion, and may function in lactobacilli-induced AMP proliferation and intestinal tissue growth and development.
- Published
- 2019
50. Sex steroid deficiency–associated bone loss is microbiota dependent and prevented by probiotics
- Author
-
Benoit Chassaing, Tao Luo, Rheinallt M. Jones, Jennifer G. Mulle, Trevor Darby, M. Neale Weitzmann, Jonathan Adams, Abdul Malik Tyagi, Chiara Vaccaro, Andrew T. Gewirtz, Jau-Yi Li, and Roberto Pacifici
- Subjects
2. Zero hunger ,0301 basic medicine ,medicine.medical_specialty ,biology ,Osteoporosis ,Inflammation ,General Medicine ,Gut flora ,biology.organism_classification ,medicine.disease ,Bone resorption ,3. Good health ,Bone remodeling ,law.invention ,03 medical and health sciences ,Probiotic ,030104 developmental biology ,Endocrinology ,Lactobacillus rhamnosus ,Sex steroid ,law ,Internal medicine ,Immunology ,medicine ,medicine.symptom - Abstract
A eubiotic microbiota influences many physiological processes in the metazoan host, including development and intestinal homeostasis. Here, we have shown that the intestinal microbiota modulates inflammatory responses caused by sex steroid deficiency, leading to trabecular bone loss. In murine models, sex steroid deficiency increased gut permeability, expanded Th17 cells, and upregulated the osteoclastogenic cytokines TNFα (TNF), RANKL, and IL-17 in the small intestine and the BM. In germ-free (GF) mice, sex steroid deficiency failed to increase osteoclastogenic cytokine production, stimulate bone resorption, and cause trabecular bone loss, demonstrating that the gut microbiota is central in sex steroid deficiency-induced trabecular bone loss. Furthermore, we demonstrated that twice-weekly treatment of sex steroid-deficient mice with the probiotics Lactobacillus rhamnosus GG (LGG) or the commercially available probiotic supplement VSL#3 reduces gut permeability, dampens intestinal and BM inflammation, and completely protects against bone loss. In contrast, supplementation with a nonprobiotic strain of E. coli or a mutant LGG was not protective. Together, these data highlight the role that the gut luminal microbiota and increased gut permeability play in triggering inflammatory pathways that are critical for inducing bone loss in sex steroid-deficient mice. Our data further suggest that probiotics that decrease gut permeability have potential as a therapeutic strategy for postmenopausal osteoporosis.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.