1
|
Vieco-Saiz N, Prévéraud DP, Pinloche E, Morat A, Govindin P, Blottière HM, Matthieu E, Devillard E, Consuegra J. Unraveling the benefits of Bacillus subtilis DSM 29784 poultry probiotic through its secreted metabolites: an in vitro approach. Microbiol Spectr 2024; 12:e0017724. [PMID: 39287453 PMCID: PMC11537077 DOI: 10.1128/spectrum.00177-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 08/04/2024] [Indexed: 09/19/2024] Open
Abstract
The probiotic Bacillus subtilis 29784 (Bs29784) sustains chicken's intestinal health, enhancing animal resilience and performance through the production of the bioactive metabolites hypoxanthine (HPX), niacin (NIA), and pantothenate (PTH). Here, using enterocyte in vitro models, we determine the functional link between these metabolites and the three pillars of intestinal resilience: immune response, intestinal barrier, and microbiota. We evaluated in vitro the capacity of Bs29784 vegetative cells, spores, and metabolites to modulate global immune regulators (using HT-29-NF-κB and HT-29-AP-1 reporter cells), intestinal integrity (HT-29-MUC2 reporter cells and Caco-2 cells), and cytokine production (Caco-2 cells). Finally, we simulated intestinal fermentations using chicken's intestinal contents as inocula to determine the effect of Bs29784 metabolites on the microbiota and their fermentation profile. Bs29784 vegetative cells reduced the inflammatory response more effectively than spores, indicating that their benefit is linked to metabolic activity. To assess this hypothesis, we studied Bs29784 metabolites individually. The results showed that each metabolite had different beneficial effects. PTH and NIA reduced the activation of the pro-inflammatory pathways AP-1 and NF-κB. HPX upregulated mucin production by enhancing MUC2 expression. HPX, NIA, and PTH increased cell proliferation. PTH and HPX increased epithelial resilience to an inflammatory challenge by limiting permeability increase. In cecal fermentations, NIA increased acetate, HPX increased butyrate, whereas PTH increased acetate, butyrate, and propionate. In ileal fermentations, PTH increased butyrate. All molecules modulated microbiota, explaining the different fermentation patterns. Altogether, we show that Bs29784 influences intestinal health by acting on the three lines of resilience via its secreted metabolites. IMPORTANCE Probiotics provide beneficial metabolites to its host. Here, we describe the mode of action of a commonly used probiotic in poultry, Bs29784. By using in vitro cellular techniques and simulated chickens' intestinal model, we show the functional link between Bs29784 metabolites and the three lines of animal resilience. Indeed, both Bs29784 vegetative cells and its metabolites stimulate cellular anti-inflammatory responses, strengthen intestinal barrier, and positively modulate microbiota composition and fermentative profile. Taken together, these results strengthen our understanding of the effect of Bs29784 on its host and explain, at least partly, its positive effects on animal health, resilience, and performance.
Collapse
Affiliation(s)
- Nuria Vieco-Saiz
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| | | | - Eric Pinloche
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| | - Aurélien Morat
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Pauline Govindin
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Hervé M. Blottière
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
- Nantes Université, INRAE, UMR 1280, PhAN, Nantes, France
| | - Elliot Matthieu
- MGP Metagenopolis, INRAE, Université Paris-Saclay, Jouy-en-Josas, France
| | - Estelle Devillard
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| | - Jessika Consuegra
- European Laboratory of Innovation Science & Expertise (ELISE). Adisseo France S.A.S., Saint Fons, France
| |
Collapse
|
2
|
Kaden T, Alonso-Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2024:e2402756. [PMID: 39491534 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH, 07745, Jena, Germany
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
| | - Raquel Alonso-Román
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, 07745, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
| | - Johannes Stallhofer
- Department of Internal Medicine IV, Jena University Hospital, 07747, Jena, Germany
| | - Mark S Gresnigt
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
- Junior Research Group Adaptive Pathogenicity Strategies, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, 07745, Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology - Hans-Knöll-Institute, 07745, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University, 07743, Jena, Germany
| | - Alexander S Mosig
- Institute of Biochemistry II, Center for Sepsis Control and Care, Jena University Hospital, 07747, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, 07745, Jena, Germany
| |
Collapse
|
3
|
Anthonymuthu S, Sabui S, Manzon KI, Sheikh A, Fleckenstein JM, Said HM. Bacterial lipopolysaccharide inhibits free thiamin uptake along the intestinal tract via interference with membrane expression of thiamin transporters 1 and 2. Am J Physiol Cell Physiol 2024; 327:C1163-C1177. [PMID: 39246143 PMCID: PMC11559647 DOI: 10.1152/ajpcell.00570.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
This study examined the effect of exposure of small and large intestinal epithelial cells to the bacterial lipopolysaccharide (LPS) on uptake of free form of vitamin B1, i.e., thiamin. The intestinal tract encounters two sources of thiamin: diet and the gut microbiota. Absorption of thiamin in both the small and large intestine occurs via a carrier-mediated process that involves thiamin transporters 1 and 2 (THTR-1 and -2). Complementary in vitro (human duodenal epithelial HuTu-80 cells and human colonic epithelial NCM460 cells), in vivo (mice), and ex vivo (human primary differentiated enteroid and colonoid monolayers) models were used. The results showed that exposure to LPS causes a significant inhibition in carrier-mediated [3H]-thiamin uptake by small and large intestinal epithelia, with no change in the levels of expression of THTR-1 and -2 mRNAs and their total cellular proteins. However, a significant decrease in the fractions of the THTR-1 and -2 proteins that are expressed at the cell membranes of these epithelial cells was observed. These effects of LPS appeared to involve a protein kinase A (PKA) signaling pathway as activating this pathway caused a reversal in the inhibition of thiamin uptake and level of expression of its transporters at the cell membrane. These findings demonstrate that exposure of gut epithelia to LPS (a situation that occurs under different pathological conditions) leads to inhibition in thiamin uptake due to a decrease in level of expression of its transporters at the cell membrane that is likely mediated via a PKA signaling pathway. NEW & NOTEWORTHY This study shows that the exposure of gut epithelial cells to bacterial LPS negatively impact the uptake process of the free form of vitamin B1 (i.e., thiamin). This appears to be mediated via suppression in the level of thiamin transporters 1 and 2 (THTR-1 and -2) expression at the cell membrane and involves a protein kinase A (PKA) signaling pathway.
Collapse
Affiliation(s)
- Selvaraj Anthonymuthu
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Subrata Sabui
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| | - Kameron Isaiah Manzon
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Medicine Service, Infectious Disease Section, Veterans Affairs Health Care System, St. Louis, Missouri, United States
| | - Hamid M Said
- Departments of Physiology/Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medicine, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| |
Collapse
|
4
|
Shin HY, Kim YS, Shin KS, Han SH, Suh HJ, Yu KW. Effect of rhamnogalacturonan-I-rich polysaccharides isolated from crabapple hydrolysates on IL-1β-induced inflammation in intestinal epithelial cells. Int J Biol Macromol 2024; 277:134240. [PMID: 39094865 DOI: 10.1016/j.ijbiomac.2024.134240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/08/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024]
Abstract
This study aimed to investigate the structural characteristics and intracellular mechanisms of polysaccharides (MP-PE-I) purified from a crabapple (Malus prunifolia) enzymatic hydrolysate (MP-PE). Activity-guided fractionation revealed that MP-PE-I was the active moiety and significantly reduced the production and gene expression of pro-inflammatory factors in interleukin (IL)-1β-treated intestinal epithelial cells (Caco-2). Moreover, MP-PE-I downregulated the phosphorylation and nuclear localization of proteins involved in the mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) pathways, as evidenced by immunoblotting and immunofluorescence analysis. In antagonistic studies with specific inhibitors of the MAPK and NF-κB pathways, IL-6 inhibition was significantly regulated by p38; IL-8 by IκBα, JNK, and p38; and monocyte chemoattractant protein-1 (MCP-1) by JNK, p38, and ERK. Additionally, MP-PE-I significantly decreased the mRNA and protein expression of IL-1 receptor type 1. Chemical and structural characteristic analyses showed that MP-PE-I is a polysaccharide rich in rhamnogalacturonan (RG)-I and plays a crucial role in intestinal immunomodulation. To our knowledge, this is the first study to demonstrate the intestinal immunomodulatory activity, intracellular mechanisms, and structural characteristics of RG-I-rich polysaccharides isolated from crabapples.
Collapse
Affiliation(s)
- Hyun Young Shin
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
| | - Yeon Suk Kim
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea
| | - Kwang-Soon Shin
- Department of Food Science and Biotechnology, Kyonggi University, Suwon 16227, Republic of Korea.
| | - Sung Hee Han
- Institute of Human Behavior & Genetics, Korea University, Seoul 02841, Republic of Korea.
| | - Hyung Joo Suh
- Transdisciplinary Major in Learning Health Systems, Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
| | - Kwang-Won Yu
- Major in Food & Nutrition, Korea National University of Transportation, Chungbuk 27909, Republic of Korea.
| |
Collapse
|
5
|
Amir A, Shahid M, Farooq Khan S, Nisar U, Faizi S, Usman Simjee S. Nicotinic acid modulates microglial TREM-2 gene in Phytohaemagglutinin-Induced in vitro model of Alzheimer's disease like pathology. Brain Res 2024; 1824:148686. [PMID: 38008243 DOI: 10.1016/j.brainres.2023.148686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 11/28/2023]
Abstract
Alzheimer's disease (AD) is a multifactorial,neurodegenerative disorder linked withextracellular amyloid beta (Aβ) plaques deposition and formation of intracellular neurofibrillary tangles (NFTs). Currently, no effective therapies are available to cure AD. Neuroinflammation isa well-known hallmark in the onset and advancement of AD and triggering receptor expressed on myeloid cells-2 (TREM-2), a microglial gene, is responsible for regulating inflammatory responses and clearance of cellular debris. Loss of TREM-2functionincreases neuroinflammation associated expression of pro-inflammatory markersthus resultingin reduced clearance of Aβ that further aid in disease progression.Therefore, targeting neuroinflammation is a good therapeutic approach for AD. This study aimed to determine the neuroprotective effect of nicotinic acid (NA) in vitro model of AD-like pathology induced in F-98 cell line using Phytohemagglutinin (PHA). MTT assay was employed for checking the cell viability as well as the proliferation of the cells following treatment with NA. PHA at the concentration of 10 μg/mL produces maximum plaques. The neuroprotective effect of NA was next evaluated against PHA-induced plaques and it was observed that NA reverses the damages induced by PHA i.e., by inhibiting the clustering of the cells and replacing the damaged cells with the new ones. Further, NA also increased the expression of TREM-2/DAP-12 with parallel decreased in the expression of IL-1β, TNF-α and iNOS. It also successfully altered disease associated ADAM-10 and BACE-1 compared to PHA control. These findings suggest that NA might be considered as a good therapeutic candidate for the treatment of neurodegenerative disorders like AD.
Collapse
Affiliation(s)
- Aiman Amir
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Maha Shahid
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sarosh Farooq Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Uzair Nisar
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shaheen Faizi
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Shabana Usman Simjee
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
6
|
Crescenzi MA, Serreli G, Deiana M, Tuberoso CIG, Montoro P, Piacente S. Metabolite Profiling, through LC-ESI/LTQOrbitrap/MS Analysis, of Antioxidant Extracts from Physalis alkekengi L. Antioxidants (Basel) 2023; 12:2101. [PMID: 38136220 PMCID: PMC10741110 DOI: 10.3390/antiox12122101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/05/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023] Open
Abstract
Due to the increasing use of Physalis alkekengi L. as a food supplement and starting material for tea preparation, a comprehensive analysis of green extracts was performed. Two different extraction methods were applied to yellow Physalis alkekengi L. fruit and calyx and compared: hydroalcoholic extraction and decoction. Characterization of the metabolome of the calyx and fruit of yellow Physalis alkekengi L. was performed by LC-ESI/LTQOrbitrap/MS followed by LC-ESI/LTQOrbitrap/MS/MS to identify 58 phytocompounds using the two different extraction techniques. Subsequently, through preliminary spectrophotometric assays followed by cell studies, the antioxidant activity of the different Physalis alkekengi L. extracts were evaluated. It was found that Physalis alkekengi L. extracts are a good source of metabolites such as flavonoids, organic acids, phenylpropanoids, physalins and carotenoids, with various biological activities, in particular, antioxidant activity capable of reducing the production of free radicals in intestinal Caco-2 cells. For the first time, an integrated approach (metabolomics approach and antioxidant evaluation) was applied to the study of Physalis alkekengi green extracts and decoctions, the green extraction method mostly used in herbal preparations. An interesting finding was the high antioxidant activity shown by these extracts.
Collapse
Affiliation(s)
- Maria Assunta Crescenzi
- Department of Pharmacy, University of the Study of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (M.A.C.); (S.P.)
| | - Gabriele Serreli
- Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.D.)
| | - Monica Deiana
- Unit of Experimental Pathology, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.D.)
| | - Carlo I. G. Tuberoso
- Department of Life and Environmental Sciences, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy;
| | - Paola Montoro
- Department of Pharmacy, University of the Study of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (M.A.C.); (S.P.)
| | - Sonia Piacente
- Department of Pharmacy, University of the Study of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy; (M.A.C.); (S.P.)
| |
Collapse
|
7
|
Anthonymuthu S, Sabui S, Lee K, Sheikh A, Fleckenstein JM, Said HM. Bacterial lipopolysaccharide inhibits colonic carrier-mediated uptake of thiamin pyrophosphate: roles for TLR4 receptor and NF-κB/P38/JNK signaling pathway. Am J Physiol Cell Physiol 2023; 325:C758-C769. [PMID: 37519229 PMCID: PMC10635650 DOI: 10.1152/ajpcell.00272.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 08/01/2023]
Abstract
This study investigated the effect of the bacterial endotoxin lipopolysaccharide (LPS) on colonic uptake of thiamin pyrophosphate (TPP), the biologically active form of vitamin B1 that is generated by gut microbiota. We used three complementary models in our study: in vitro (human-derived colonic epithelial NCM460), ex vivo (human differentiated colonoid monolayers), and in vivo (mouse colonic tissue). The results showed that exposure of NCM460 cells to LPS leads to a significant inhibition of carrier-mediated TPP uptake as well as in decreased expression of the colonic TPP transporter (cTPPT) protein, mRNA, and heterologous nuclear RNA (hnRNA) compared with untreated controls. Similarly, exposure of human differentiated colonoid monolayers and mice to LPS caused significant inhibition in colonic carrier-mediated TPP uptake and in cTPPT protein, mRNA, and hnRNA expression. The effect of LPS on colonic TPP uptake and cTTPT expression was also found to be associated with a significant reduction in activity of the SLC44A4 promoter as well as in decreased expression of the nuclear factor Elf-3 (E74-like ETS transcription factor 3), which is needed for promoter activity. Finally, we found that knocking down the Toll-like receptor 4 (TLR4) and blocking the nuclear factor kappa B (NF-κB), JNK, and p38 signaling pathways with the use of pharmacological inhibitors lead to significant abrogation in the degree of LPS-mediated inhibition in TPP uptake and cTPPT expression. These results demonstrated that exposure of colonic epithelia to LPS inhibits colonic TPP uptake via transcriptional mechanism(s) and that the effect is mediated via TLR4 receptor and NF-κB/p38/JNK signaling pathways.NEW & NOTEWORTHY This study examined the effect of the bacterial lipopolysaccharide (LPS) on the colonic uptake of thiamin pyrophosphate (TPP), the biologically active form of vitamin B1. Three complementary models were used: in vitro (human NCM460 cells), ex vivo (human colonoids), and in vivo (mice). The results showed LPS to significantly suppress TPP uptake and the expression of its transporter, and that these effects are mediated via the membrane TLR4 receptor, and involve the NF-κB/p38/JNK signaling pathways.
Collapse
Affiliation(s)
- Selvaraj Anthonymuthu
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Subrata Sabui
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| | - Katherine Lee
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States
- Veterans Affairs Medical Center, St. Louis, Missouri, United States
| | - Hamid M Said
- Department of Physiology & Biophysics, School of Medicine, University of California, Irvine, California, United States
- Department of Medicine, School of Medicine, University of California, Irvine, California, United States
- Department of Medical Research, Tibor Rubin VA Medical Center, Long Beach, California, United States
| |
Collapse
|
8
|
Niño-Narvión J, Rojo-López MI, Martinez-Santos P, Rossell J, Ruiz-Alcaraz AJ, Alonso N, Ramos-Molina B, Mauricio D, Julve J. NAD+ Precursors and Intestinal Inflammation: Therapeutic Insights Involving Gut Microbiota. Nutrients 2023; 15:2992. [PMID: 37447318 DOI: 10.3390/nu15132992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
The oxidized form of nicotinamide adenine dinucleotide (NAD+) is a critical metabolite for living cells. NAD+ may act either as a cofactor for many cellular reactions as well as a coenzyme for different NAD+-consuming enzymes involved in the physiological homeostasis of different organs and systems. In mammals, NAD+ is synthesized from either tryptophan or other vitamin B3 intermediates that act as NAD+ precursors. Recent research suggests that NAD+ precursors play a crucial role in maintaining the integrity of the gut barrier. Indeed, its deficiency has been associated with enhanced gut inflammation and leakage, and dysbiosis. Conversely, NAD+-increasing therapies may confer protection against intestinal inflammation in experimental conditions and human patients, with accumulating evidence indicating that such favorable effects could be, at least in part, mediated by concomitant changes in the composition of intestinal microbiota. However, the mechanisms by which NAD+-based treatments affect the microbiota are still poorly understood. In this context, we have focused specifically on the impact of NAD+ deficiency on intestinal inflammation and dysbiosis in animal and human models. We have further explored the relationship between NAD+ and improved host intestinal metabolism and immunity and the composition of microbiota in vivo. Overall, this comprehensive review aims to provide a new perspective on the effect of NAD+-increasing strategies on host intestinal physiology.
Collapse
Affiliation(s)
- Julia Niño-Narvión
- Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- Grupo de Obesidad y Metabolismo, Instituto Murciano de Investigación Biosanitaria (IMIB), 30120 Murcia, Spain
- Departamento de Bioquímica y Biología Molecular B e Inmunología, Facultad de Medicina, Universidad de Murcia (UMU), 30120 Murcia, Spain
| | | | | | - Joana Rossell
- Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 08041 Barcelona, Spain
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
| | - Antonio J Ruiz-Alcaraz
- Departamento de Bioquímica y Biología Molecular B e Inmunología, Facultad de Medicina, Universidad de Murcia (UMU), 30120 Murcia, Spain
| | - Núria Alonso
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias I Pujol, 08916 Badalona, Spain
| | - Bruno Ramos-Molina
- Grupo de Obesidad y Metabolismo, Instituto Murciano de Investigación Biosanitaria (IMIB), 30120 Murcia, Spain
| | - Didac Mauricio
- Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 08041 Barcelona, Spain
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Faculty of Medicine, University of Vic/Central University of Catalonia (UVIC/UCC), 08500 Vic, Spain
| | - Josep Julve
- Institut d'Investigació Biomèdica Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, 08041 Barcelona, Spain
| |
Collapse
|
9
|
Chu W, Li YL, Li JJ, Lin J, Li M, Wang J, He JZ, Zhang YM, Yao J, Jin XJ, Cai H, Liu YQ. Guiqi Baizhu prescription ameliorates cytarabine-induced intestinal mucositis by targeting JAK2 to inhibit M1 macrophage polarization. Biomed Pharmacother 2023; 164:114902. [PMID: 37209628 DOI: 10.1016/j.biopha.2023.114902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Intestinal mucositis (IM) is characterized by damage to the intestinal mucosa resulting from inhibition of epithelial cell division and loss of renewal capacity following anticancer chemotherapy and radiotherapy. Cytarabine (Ara-C), the main chemotherapy drug for the treatment of leukemia and lymphoma, is a frequent cause of IM. Guiqi Baizhu prescription (GQBZP) is a traditional Chinese medicine with anti-cancer and anti-inflammatory effects. PURPOSE To determine if GQBZP can ameliorate Ara-C induced IM and identify and characterize the pharmacologic and pharmacodynamic mechanisms. STUDY DESIGN AND METHODS IM was induced in mice with Ara-C and concurrently treated with orally administered GQBZP. Body weight and food intake was monitored, with HE staining to calculate ileal histomorphometric scoring and villus length/crypt depth. Immunoblotting was used to detect intestinal tissue inflammatory factors. M1 macrophages (M1) were labeled with CD86 by flow cytometry and iNOS + F4/80 by immunofluorescence. Virtual screening was used to find potentially active compounds in GQBZP that targeted JAK2. In vitro, RAW264.7 cells were skewed to M1 macrophage polarization by lipopolysaccharide (LPS) and interferon-γ (INF-γ) and treated orally with GQBZP or potential active compounds. M1 was labeled with CD86 by flow cytometry and iNOS by immunofluorescence. ELISA was used to detect inflammatory factor expression. Active compounds against JAK2, p-JAK2, STAT1 and p-STAT1 were identified by western blotting and HCS fluorescence. Molecular dynamics simulations and pharmacokinetic predictions were carried out on representative active compounds. RESULTS Experimental results with mice in vivo suggest that GQBZP significantly attenuated Ara-C-induced ileal damage and release of pro-inflammatory factors by inhibiting macrophage polarization to M1. Molecular docking was used to identify potentially active compounds in GQBZP that targeted JAK2, a key factor in macrophage polarization to M1. By examining the main components of each herb and applying Lipinski's rules, ten potentially active compounds were identified. In vitro experimental results suggested that all 10 compounds of GQBZP targeted JAK2 and could inhibit M1 polarization in RAW264.7 cells treated with LPS and INF-γ. Among them, acridine and senkyunolide A down-regulated the expression of JAK2 and STAT1. MD simulations revealed that acridine and senkyunolide A were stable in the active site of JAK2 and exhibited good interactions with the surrounding amino acids. CONCLUSIONS GQBZP can ameliorate Ara-C-induced IM by reducing macrophage polarization to M1, and acridine and senkyunolide A are representative active compounds in GQBZP that target JAK2 to inhibit M1 polarization. Targeting JAK2 to regulate M1 polarization may be a valuable therapeutic strategy for IM.
Collapse
Affiliation(s)
- Wei Chu
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Ya-Ling Li
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China; Key Laboratory of Dun Huang Medical and Transformation, Ministry of Education of The People's Republic of China, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jun-Jie Li
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jia Lin
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Mi Li
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jiao Wang
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jian-Zheng He
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Yue-Mei Zhang
- Ophthalmology Department, First Hospital of Lanzhou University, 730000 Lanzhou, China
| | - Juan Yao
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Xiao-Jie Jin
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China; College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China.
| | - Hui Cai
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 730000 Lanzhou, China; NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, 730000 Lanzhou, China.
| | - Yong-Qi Liu
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China; Key Laboratory of Dun Huang Medical and Transformation, Ministry of Education of The People's Republic of China, Gansu University of Chinese Medicine, 730000 Lanzhou, China.
| |
Collapse
|
10
|
Glutamine Starvation Affects Cell Cycle, Oxidative Homeostasis and Metabolism in Colorectal Cancer Cells. Antioxidants (Basel) 2023; 12:antiox12030683. [PMID: 36978930 PMCID: PMC10045305 DOI: 10.3390/antiox12030683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Cancer cells adjust their metabolism to meet energy demands. In particular, glutamine addiction represents a distinctive feature of several types of tumors, including colorectal cancer. In this study, four colorectal cancer cell lines (Caco-2, HCT116, HT29 and SW480) were cultured with or without glutamine. The growth and proliferation rate, colony-forming capacity, apoptosis, cell cycle, redox homeostasis and metabolomic analysis were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test (MTT), flow cytometry, high-performance liquid chromatography and gas chromatography/mass spectrometry techniques. The results show that glutamine represents an important metabolite for cell growth and that its deprivation reduces the proliferation of colorectal cancer cells. Glutamine depletion induces cell death and cell cycle arrest in the GO/G1 phase by modulating energy metabolism, the amino acid content and antioxidant defenses. Moreover, the combined glutamine starvation with the glycolysis inhibitor 2-deoxy-D-glucose exerted a stronger cytotoxic effect. This study offers a strong rationale for targeting glutamine metabolism alone or in combination with glucose metabolism to achieve a therapeutic benefit in the treatment of colon cancer.
Collapse
|
11
|
Casula E, Pisano MB, Serreli G, Zodio S, Melis MP, Corona G, Costabile A, Cosentino S, Deiana M. Probiotic lactobacilli attenuate oxysterols-induced alteration of intestinal epithelial cell monolayer permeability: Focus on tight junction modulation. Food Chem Toxicol 2023; 172:113558. [PMID: 36528245 DOI: 10.1016/j.fct.2022.113558] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/16/2022]
Abstract
Oxidative stress and inflammation lead by dietary oxidised lipids, as oxysterols, have been linked to the loss of intestinal barrier integrity, a crucial event in the initiation and progression of intestinal disorders. In the last decade, probiotic lactobacilli have emerged as an interesting tool to improve intestinal health, thanks to their antioxidant and anti-inflammatory properties. The aim of the present study was to evaluate the ability of two commercial probiotic strains of lactobacilli (Lactiplantibacillus plantarum 299v® (DMS 9843) and Lacticaseibacillus casei DG® (CNCMI-1572)), both as live bacteria and intracellular content, to attenuate the oxysterols-induced alteration of intestinal epithelial Caco-2 cell monolayer permeability. Our investigation was focused on the modulation of tight junctions (TJs) proteins, occludin, ZO-1 and JAM-A, in relation to redox-sensitive MAPK p38 activation. Obtained results provided evidence on the ability of the two probiotics to counteract the alteration of monolayer permeability and loss of TJs proteins, at least in part, through the modulation of p38 pathway. The protective action was exerted by live bacteria, whose adhesion to Caco-2 cells was not altered by oxysterols, and bacterial intracellular components equally able to interact with the signaling pathway.
Collapse
Affiliation(s)
- Emanuela Casula
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Maria Barbara Pisano
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Gabriele Serreli
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy.
| | - Sonia Zodio
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Maria Paola Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| | - Giulia Corona
- School of Life and Health Sciences, University of Roehampton, London, SW15 4JD, UK
| | - Adele Costabile
- School of Life and Health Sciences, University of Roehampton, London, SW15 4JD, UK
| | - Sofia Cosentino
- Department of Medical Sciences and Public Health, University of Cagliari, Monserrato, 09042, Cagliari, Italy.
| | - Monica Deiana
- Department of Biomedical Sciences, University of Cagliari, Monserrato, 09042, Cagliari, Italy
| |
Collapse
|
12
|
Qiu P, Song Z. Determination of Nicotinic Acid in Food and Pharmaceuticals by a Simple and Rapid Fluorescence Polarization Immunoassay (FPIA). ANAL LETT 2023. [DOI: 10.1080/00032719.2023.2173220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Peng Qiu
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| | - Zhaorui Song
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, China
| |
Collapse
|
13
|
B Vitamins and Their Roles in Gut Health. Microorganisms 2022; 10:microorganisms10061168. [PMID: 35744686 PMCID: PMC9227236 DOI: 10.3390/microorganisms10061168] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
B vitamins act as coenzymes in a myriad of cellular reactions. These include energy production, methyl donor generation, neurotransmitter synthesis, and immune functions. Due to the ubiquitous roles of these vitamins, their deficiencies significantly affect the host’s metabolism. Recently, novel roles of B vitamins in the homeostasis of gut microbial ecology and intestinal health continue to be unravelled. This review focuses on the functional roles and biosynthesis of B vitamins and how these vitamins influence the growth and proliferation of the gut microbiota. We have identified the gut bacteria that can produce vitamins, and their biosynthetic mechanisms are presented. The effects of B vitamin deficiencies on intestinal morphology, inflammation, and its effects on intestinal disorders are also discussed.
Collapse
|
14
|
Gut Microbial Metabolite-Mediated Regulation of the Intestinal Barrier in the Pathogenesis of Inflammatory Bowel Disease. Nutrients 2021; 13:nu13124259. [PMID: 34959809 PMCID: PMC8704337 DOI: 10.3390/nu13124259] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease. The disease has a multifactorial aetiology, involving genetic, microbial as well as environmental factors. The disease pathogenesis operates at the host-microbe interface in the gut. The intestinal epithelium plays a central role in IBD disease pathogenesis. Apart from being a physical barrier, the epithelium acts as a node that integrates environmental, dietary, and microbial cues to calibrate host immune response and maintain homeostasis in the gut. IBD patients display microbial dysbiosis in the gut, combined with an increased barrier permeability that contributes to disease pathogenesis. Metabolites produced by microbes in the gut are dynamic indicators of diet, host, and microbial interplay in the gut. Microbial metabolites are actively absorbed or diffused across the intestinal lining to affect the host response in the intestine as well as at systemic sites via the engagement of cognate receptors. In this review, we summarize insights from metabolomics studies, uncovering the dynamic changes in gut metabolite profiles in IBD and their importance as potential diagnostic and prognostic biomarkers of disease. We focus on gut microbial metabolites as key regulators of the intestinal barrier and their role in the pathogenesis of IBD.
Collapse
|
15
|
Serreli G, Naitza MR, Zodio S, Leoni VP, Spada M, Melis MP, Boronat A, Deiana M. Ferulic Acid Metabolites Attenuate LPS-Induced Inflammatory Response in Enterocyte-like Cells. Nutrients 2021; 13:nu13093152. [PMID: 34579029 PMCID: PMC8471535 DOI: 10.3390/nu13093152] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Ferulic acid (FA) is a polyphenol pertaining to the class of hydroxycinnamic acids present in numerous foods of a plant origin. Its dietary consumption leads to the formation of several phase I and II metabolites in vivo, which represent the largest amount of ferulates in the circulation and in the intestine in comparison with FA itself. In this work, we evaluated their efficacy against the proinflammatory effects induced by lipopolysaccharide (LPS) in intestinal Caco-2 cell monolayers, as well as the mechanisms underlying their protective action. LPS-induced overexpression of proinflammatory enzymes such as inducible nitric oxide synthase (iNOS) and the consequent hyperproduction of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) were limited by physiological relevant concentrations (1 µM) of FA, its derivatives isoferulic acid (IFA) and dihydroferulic acid (DHFA), and their glucuronidated and sulfated metabolites, which acted upstream by limiting the activation of MAPK p38 and ERK and of Akt kinase, thus decreasing the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) translocation into the nucleus. Furthermore, the compounds were found to promote the expression of Nrf2, which may have contributed to the downregulation of NF-ĸB activity. The overall data show that phase I/II metabolites retain the efficacy of their dietary free form in contrasting inflammatory response.
Collapse
Affiliation(s)
- Gabriele Serreli
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
| | - Micaela Rita Naitza
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
| | - Sonia Zodio
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
| | - Vera Piera Leoni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
| | - Martina Spada
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
| | - Maria Paola Melis
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
| | - Anna Boronat
- Integrative Pharmacology and Systems Neurosciences Research Group, Hospital del Mar Medical Research Institute, 08003 Barcelona, Spain;
| | - Monica Deiana
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria SS 554, 09042 Monserrato, Italy; (G.S.); (M.R.N.); (S.Z.); (V.P.L.); (M.S.); (M.P.M.)
- Correspondence: ; Tel.: +39-070-675-4126
| |
Collapse
|
16
|
Santoru ML, Piras C, Murgia F, Leoni VP, Spada M, Murgia A, Liggi S, Lai MA, Usai P, Caboni P, Manzin A, Atzori L. Metabolic Alteration in Plasma and Biopsies From Patients With IBD. Inflamm Bowel Dis 2021; 27:1335-1345. [PMID: 33512485 DOI: 10.1093/ibd/izab012] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory bowel diseases (IBD) are chronic inflammatory disorders of the gastrointestinal tract, with periods of latency alternating with phases of exacerbation, and include 2 forms: Crohn disease (CD) and ulcerative colitis (UC). Although the etiology of IBD is still unclear, the identification and understanding of pathophysiological mechanisms underlying IBD could reveal newly targeted intestinal alterations and determine therapeutic approaches. METHODS In this study, by using gas chromatography-mass spectrometry, we characterized plasma and biopsies from the metabolomics profiles of patients with IBD compared with those of a control group. RESULTS The results showed a different metabolomics profile between patients with CD (n = 50) and patients with UC (n = 82) compared with the control group (n = 51). Multivariate statistical analysis of the identified metabolites in CD and UC showed changes in energetic metabolism, and lactic acid and ornithine in particular were altered in both plasma and colon biopsies. Moreover, metabolic changes were evidenced between the normal ileum and colon tissues. These differences disappeared when we compared the inflamed ileum and colon tissues, suggesting a common metabolism. CONCLUSIONS This study showed how the metabolomics profile could be a potential tool to identify intestinal alterations associated with IBD and may have application in precision medicine and for better defining the pathogenesis of the disease.
Collapse
Affiliation(s)
| | - Cristina Piras
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| | - Federica Murgia
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| | - Vera Piera Leoni
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| | - Martina Spada
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| | - Antonio Murgia
- Department of Life and Environmental Sciences, University of Cagliari, Sardinia, Italy
| | - Sonia Liggi
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| | - Maria Antonia Lai
- Gastroenterology Unit, University Hospital of Cagliari, Sardinia, Italy
| | - Paolo Usai
- Department of Medical Sciences and Public Health, University of Cagliari, Sardinia, Italy
| | - Pierluigi Caboni
- Department of Life and Environmental Sciences, University of Cagliari, Sardinia, Italy
| | - Aldo Manzin
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| | - Luigi Atzori
- Department of Biomedical Sciences, University of Cagliari, Sardinia, Italy
| |
Collapse
|
17
|
Choi P, Rhayat L, Pinloche E, Devillard E, De Paepe E, Vanhaecke L, Haesebrouck F, Ducatelle R, Van Immerseel F, Goossens E. Bacillus Subtilis 29784 as a Feed Additive for Broilers Shifts the Intestinal Microbial Composition and Supports the Production of Hypoxanthine and Nicotinic Acid. Animals (Basel) 2021; 11:1335. [PMID: 34066686 PMCID: PMC8150382 DOI: 10.3390/ani11051335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/21/2021] [Accepted: 04/24/2021] [Indexed: 12/30/2022] Open
Abstract
The probiotic Bacillus subtilis strain 29784 (Bs29784) has been shown to improve performance in broilers. In this study, we used a metabolomic and 16S rRNA gene sequencing approach to evaluate effects of Bs29874 in the broiler intestine. Nicotinic acid and hypoxanthine were key metabolites that were produced by the strain in vitro and were also found in vivo to be increased in small intestinal content of broilers fed Bs29784 as dietary additive. Both metabolites have well-described anti-inflammatory effects in the intestine. Furthermore, Bs29784 supplementation to the feed significantly altered the ileal microbiome of 13-day-old broilers, thereby increasing the abundance of genus Bacillus, while decreasing genera and OTUs belonging to the Lactobacillaceae and Enterobacteriacae families. Moreover, Bs29784 did not change the cecal microbial community structure, but specifically enriched members of the family Clostridiales VadinBB60, as well as the butyrate-producing families Ruminococcaceae and Lachnospiraceae. The abundance of various OTUs and genera belonging to these families was significantly associated with nicotinic acid levels in the cecum, suggesting a possible cross-feeding between B. subtilis strain 29784 and these beneficial microbes. Taken together, the data indicate that Bs29784 exerts its described probiotic effects through a combined action of its metabolites on both the host and its microbiome.
Collapse
Affiliation(s)
- Pearl Choi
- Livestock Gut Health Team, Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (P.C.); (R.D.)
| | - Lamya Rhayat
- Adisseo France SAS, Center of Expertise and Research in Nutrition (CERN), 6 Route Noire, 03600 Commentry, France; (L.R.); (E.P.); (E.D.)
| | - Eric Pinloche
- Adisseo France SAS, Center of Expertise and Research in Nutrition (CERN), 6 Route Noire, 03600 Commentry, France; (L.R.); (E.P.); (E.D.)
| | - Estelle Devillard
- Adisseo France SAS, Center of Expertise and Research in Nutrition (CERN), 6 Route Noire, 03600 Commentry, France; (L.R.); (E.P.); (E.D.)
| | - Ellen De Paepe
- Laboratory of Chemical Analysis, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (E.D.P.); (L.V.)
| | - Lynn Vanhaecke
- Laboratory of Chemical Analysis, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (E.D.P.); (L.V.)
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium;
| | - Richard Ducatelle
- Livestock Gut Health Team, Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (P.C.); (R.D.)
| | - Filip Van Immerseel
- Livestock Gut Health Team, Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (P.C.); (R.D.)
| | - Evy Goossens
- Livestock Gut Health Team, Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium; (P.C.); (R.D.)
| |
Collapse
|