1
|
Grant ET, Parrish A, Boudaud M, Hunewald O, Hirayama A, Ollert M, Fukuda S, Desai MS. Dietary fibers boost gut microbiota-produced B vitamin pool and alter host immune landscape. MICROBIOME 2024; 12:179. [PMID: 39307855 PMCID: PMC11418204 DOI: 10.1186/s40168-024-01898-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/31/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Dietary fibers can alter microbial metabolic output in support of healthy immune function; however, the impact of distinct fiber sources and immunomodulatory effects beyond short-chain fatty acid production are underexplored. In an effort to discern the effects of diverse fibers on host immunity, we employed five distinct rodent diets with varying fiber content and source in specific-pathogen-free, gnotobiotic (containing a 14-member synthetic human gut microbiota), and germ-free mice. RESULTS Broad-scale metabolomics analysis of cecal contents revealed that fiber deprivation consistently reduced the concentrations of microbiota-produced B vitamins. This phenomenon was not always explained by reduced biosynthesis, rather, metatranscriptomic analyses pointed toward increased microbial usage of certain B vitamins under fiber-free conditions, ultimately resulting in a net reduction of host-available B vitamins. Broad immunophenotyping indicated that the local gut effector immune populations and activated T cells accumulate in a microbiota-dependent manner. Supplementation with the prebiotic inulin recovered the availability of microbially produced B vitamins and restored immune homeostasis. CONCLUSIONS Our findings highlight the potential to use defined fiber polysaccharides to boost microbiota-derived B vitamin availability in an animal model and to regulate local innate and adaptive immune populations of the host. Video abstract.
Collapse
Affiliation(s)
- Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 4365, Esch-Sur-Alzette, Luxembourg
| | - Amy Parrish
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 4365, Esch-Sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354, Esch-Sur-Alzette, Luxembourg
| | - Oliver Hunewald
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354, Esch-Sur-Alzette, Luxembourg
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Yamagata, 997-0052, Japan
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354, Esch-Sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000, Odense, Denmark
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Yamagata, 997-0052, Japan
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, 305-8575, Japan
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kanagawa, 210-0821, Japan
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354, Esch-Sur-Alzette, Luxembourg.
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000, Odense, Denmark.
| |
Collapse
|
2
|
Chang WCL, Ghosh J, Cooper HS, Vanderveer L, Schultz B, Zhou Y, Harvey KN, Kaunga E, Devarajan K, Li Y, Jelinek J, Fragoso MF, Sapienza C, Clapper ML. Folic Acid Supplementation Promotes Hypomethylation in Both the Inflamed Colonic Mucosa and Colitis-Associated Dysplasia. Cancers (Basel) 2023; 15:2949. [PMID: 37296911 PMCID: PMC10252136 DOI: 10.3390/cancers15112949] [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: 05/05/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
PURPOSE The purpose of this study was to assess the effect of folic acid (FA) supplementation on colitis-associated colorectal cancer (CRC) using the azoxymethane/dextran sulfate sodium (AOM/DSS) model. METHODS Mice were fed a chow containing 2 mg/kg FA at baseline and randomized after the first DSS treatment to receive 0, 2, or 8 mg/kg FA chow for 16 weeks. Colon tissue was collected for histopathological evaluation, genome-wide methylation analyses (Digital Restriction Enzyme Assay of Methylation), and gene expression profiling (RNA-Seq). RESULTS A dose-dependent increase in the multiplicity of colonic dysplasias was observed, with the multiplicity of total and polypoid dysplasias higher (64% and 225%, respectively) in the 8 mg FA vs. the 0 mg FA group (p < 0.001). Polypoid dysplasias were hypomethylated, as compared to the non-neoplastic colonic mucosa (p < 0.05), irrespective of FA treatment. The colonic mucosa of the 8 mg FA group was markedly hypomethylated as compared to the 0 mg FA group. Differential methylation of genes involved in Wnt/β-catenin and MAPK signaling resulted in corresponding alterations in gene expression within the colonic mucosa. CONCLUSIONS High-dose FA created an altered epigenetic field effect within the non-neoplastic colonic mucosa. The observed decrease in site-specific DNA methylation altered oncogenic pathways and promoted colitis-associated CRC.
Collapse
Affiliation(s)
- Wen-Chi L. Chang
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
| | - Jayashri Ghosh
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (J.G.); (B.S.)
| | - Harry S. Cooper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Lisa Vanderveer
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
| | - Bryant Schultz
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (J.G.); (B.S.)
| | - Yan Zhou
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Kristen N. Harvey
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
| | - Esther Kaunga
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
| | - Karthik Devarajan
- Biostatistics and Bioinformatics Facility, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Yuesheng Li
- DNA Sequencing and Genomic Core Facility, National Heart, Lung, and Blood Institute, NIH, 10 Center Drive, Bethesda, MD 20892, USA
| | - Jaroslav Jelinek
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (J.G.); (B.S.)
| | - Mariana F. Fragoso
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
| | - Carmen Sapienza
- Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (J.G.); (B.S.)
| | - Margie L. Clapper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA (L.V.); (E.K.)
| |
Collapse
|
3
|
Mass spectrometry-based metabolomics approach and in vitro assays revealed promising role of 2,3-dihydroquinazolin-4(1H)-one derivatives against colorectal cancer cell lines. Eur J Pharm Sci 2023; 182:106378. [PMID: 36638899 DOI: 10.1016/j.ejps.2023.106378] [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: 09/26/2022] [Revised: 12/24/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Colorectal cancer (CRC) is the most frequent form of gastrointestinal cancer and one of the major causes of human mortality worldwide. Many of the current CRC therapies have limitations due to multidrug resistance and/or severe side effects. Quinazoline derivatives are promising lead compounds with a wide range of pharmacological actions. In this study, the effect of seven synthesized 2,3-dihydroquinazolin-4(1H)-one analogues as potential anticancer agents against two CRC cell lines (HCT116 and SW480) was investigated using cell viability proliferation, migration, adhesion and invasion assays. A liquid chromatography-mass spectrometry (LC-MS/MS) metabolomics approach was used to identify the underlying biochemical pathways disturbed in treated-HCT116 cells. Cell viability proliferation assay revealed that four compounds (C2, C3, C5, and C7) had IC50 < 10 µM with C5 displaying the most potent cytotoxic effect (IC50 1.4 and 0.3 µM against HCT116 and SW480, respectively). Additionally, the compounds showed suppression of wound closure after 72 h, and both C2 and C5 significantly decreased the number of adherent cells and suppressed HCT116 cells invasion. Metabolomics study revealed that C5 induced significant perturbations in the level of several metabolites including spermine, polyamines, glutamine, creatine and carnitine, and altered biochemical processes essential for cell proliferation and progression such as amino acids biosynthesis and metabolism, redox homeostasis, energy related processes (e.g., fatty acid oxidation, second Warburg like effect) and one-carbon metabolism. Our findings indicate that 2,3-dihydroquinazolin-4(1H)-one analogues, particularly C5, have promising anticancer properties, and shed light on the role of metabolomics in identifying new therapeutic targets and providing better understanding of the pathways altered in treated cancer cells.
Collapse
|
4
|
Gold SL, Manning L, Kohler D, Ungaro R, Sands B, Raman M. Micronutrients and Their Role in Inflammatory Bowel Disease: Function, Assessment, Supplementation, and Impact on Clinical Outcomes Including Muscle Health. Inflamm Bowel Dis 2023; 29:487-501. [PMID: 36287025 DOI: 10.1093/ibd/izac223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Indexed: 12/09/2022]
Affiliation(s)
- Stephanie L Gold
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Manning
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Kohler
- Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Ryan Ungaro
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce Sands
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maitreyi Raman
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
5
|
Guetterman HM, Huey SL, Knight R, Fox AM, Mehta S, Finkelstein JL. Vitamin B-12 and the Gastrointestinal Microbiome: A Systematic Review. Adv Nutr 2021; 13:S2161-8313(22)00075-8. [PMID: 34612492 PMCID: PMC8970816 DOI: 10.1093/advances/nmab123] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Vitamin B-12 deficiency is a major public health problem affecting individuals across the lifespan, with known hematological, neurological, and obstetric consequences. Emerging evidence suggests that vitamin B-12 may have an important role in other aspects of human health, including the composition and function of the gastrointestinal (gut) microbiome. Vitamin B-12 is synthesized and utilized by bacteria in the human gut microbiome and is required for over a dozen enzymes in bacteria, compared to only two in humans. However, the impact of vitamin B-12 on the gut microbiome has not been established. This systematic review was conducted to examine the evidence that links vitamin B-12 and the gut microbiome. A structured search strategy was used to identify in vitro, animal, and human studies that assessed vitamin B-12 status, dietary intake, or supplementation, and the gut microbiome using culture-independent techniques. A total of 22 studies (3 in vitro, 8 animal, 11 human observational studies) were included. Nineteen studies reported vitamin B-12 intake, status, or supplementation was associated with gut microbiome outcomes, including beta-diversity, alpha-diversity, relative abundance of bacteria, functional capacity, or short chain fatty acid production. Evidence suggests vitamin B-12 may be associated with changes in bacterial abundance. While results from in vitro studies suggest vitamin B-12 may increase alpha-diversity and shift gut microbiome composition (beta-diversity), findings from animal studies and observational human studies were heterogeneous. Based on evidence from in vitro and animal studies, microbiome outcomes may differ by cobalamin form and co-intervention. To date, few prospective observational studies and no randomized trials have been conducted to examine the effects of vitamin B-12 on the human gut microbiome. The impact of vitamin B-12 on the gut microbiome needs to be elucidated to inform screening and public health interventions. Statement of significance: Vitamin B-12 is synthesized and utilized by bacteria in the human gut microbiome and is required by over a dozen enzymes in bacteria. However, to date, no systematic reviews have been conducted to evaluate the impact of vitamin B-12 on the gut microbiome, or its implications for human health.
Collapse
Affiliation(s)
| | - Samantha L Huey
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA,Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA,Department of Bioengineering, University of California San Diego, La Jolla, CA, USA,Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | - Allison M Fox
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Saurabh Mehta
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA,Division of Epidemiology, Department of Population Health Sciences, Weill Cornell Medical College, New York, NY, USA,Institute for Nutritional Sciences, Global Health, and Technology, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
6
|
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: 37] [Impact Index Per Article: 9.3] [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
|
7
|
Ratajczak AE, Szymczak-Tomczak A, Rychter AM, Zawada A, Dobrowolska A, Krela-Kaźmierczak I. Does Folic Acid Protect Patients with Inflammatory Bowel Disease from Complications? Nutrients 2021; 13:nu13114036. [PMID: 34836291 PMCID: PMC8618862 DOI: 10.3390/nu13114036] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/09/2021] [Indexed: 01/12/2023] Open
Abstract
Folic acid, referred to as vitamin B9, is a water-soluble substance, which participates in the synthesis of nucleic acids, amino acids, and proteins. Similarly to B12 and B6, vitamin B9 is involved in the metabolism of homocysteine, which is associated with the MTHFR gene. The human body is not able to synthesize folic acid; thus, it must be supplemented with diet. The most common consequence of folic acid deficiency is anemia; however, some studies have also demonstrated the correlation between low bone mineral density, hyperhomocysteinemia, and folic acid deficiency. Patients with inflammatory bowel disease (IBD) frequently suffer from malabsorption and avoid certain products, such as fresh fruits and vegetables, which constitute the main sources of vitamin B9. Additionally, the use of sulfasalazine by patients may result in folic acid deficiency. Therefore, IBD patients present a higher risk of folic acid deficiency and require particular supervision with regard to anemia and osteoporosis prevention, which are common consequences of IBD.
Collapse
Affiliation(s)
- Alicja Ewa Ratajczak
- Correspondence: (A.E.R.); (I.K.-K.); Tel.: +48-667-385-996 (A.E.R.); +48-8691-343 (I.K.-K.); Fax: +48-8691-686 (A.E.R.)
| | | | | | | | | | - Iwona Krela-Kaźmierczak
- Correspondence: (A.E.R.); (I.K.-K.); Tel.: +48-667-385-996 (A.E.R.); +48-8691-343 (I.K.-K.); Fax: +48-8691-686 (A.E.R.)
| |
Collapse
|
8
|
Zhang J, Cai D, Yang M, Hao Y, Zhu Y, Chen Z, Aziz T, Sarwar A, Yang Z. Screening of folate-producing lactic acid bacteria and modulatory effects of folate-biofortified yogurt on gut dysbacteriosis of folate-deficient rats. Food Funct 2021; 11:6308-6318. [PMID: 32602881 DOI: 10.1039/d0fo00480d] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Folate deficiency is accompanied by gut dysbacteriosis. To understand dietary intervention in folate deficiency, a folate-deficient rat model was used to evaluate the modulatory effects of folate-producing lactic acid bacteria (LAB) and biofortified yogurt on gut dysbacteriosis. The high folate-producing strain was screened from 12 LABs, and its variant, namely Lactobacillus plantarum GSLP-7 V, with folate productivity in yogurt at 3.72 μg mL-1, was obtained by stressing with 5.0 mg L-1 methotrexate and 100.00 mg L-1 Ca2+. To our knowledge, this is the highest folate productivity in yogurt by LAB strains ever reported. To further examine the folate supplement effect in vivo, a folate-deficient rat model was established and fed a folate-free diet for 8 weeks. Also, the effects of L. plantrum GSLP-7 V, yogurt fermented with L. plantrum GSLP-7 V, plain yogurt, and chemical folic acid on folate deficiency and gut dysbacteriosis were examined. Analysis of the change in gut microbiota showed that the gut dysbacteriosis was significantly correlated with folate deficiency. Administration of L. plantrum GSLP-7 V and its fermented yogurt for 10 days restored the disrupted gut microbiota and recovered the serum folate and homocysteine to normal levels, while chemical folic acid worsened the gut dysbacteriosis. Chemical folic acid only enriched Akkermansia, while L. plantrum GSLP-7 V and its fermented yogurt modulated the gut microbiota comprehensively through 7 and 10 key genera, respectively. This study confirmed the effectiveness of dietary intervention with folate-biofortified yogurt through modulating gut microbiota, suggesting the potential of the folate-producing LAB as an agent for the treatment of folate-deficiency related diseases.
Collapse
Affiliation(s)
- Jian Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Dongyan Cai
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Ming Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Yijiang Hao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Yuanhua Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Zexuan Chen
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Tariq Aziz
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Abid Sarwar
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| | - Zhennai Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China.
| |
Collapse
|
9
|
Diaz G S, LeBlanc DP, Gagné R, Behan NA, Wong A, Marchetti F, MacFarlane AJ. Folate Intake Alters Mutation Frequency and Profiles in a Tissue- and Dose-Specific Manner in MutaMouse Male Mice. J Nutr 2021; 151:800-809. [PMID: 33693772 DOI: 10.1093/jn/nxaa402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/09/2020] [Accepted: 11/20/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND While cancer is common, its incidence varies widely by tissue. These differences are attributable to variable risk factors, such as environmental exposure, genetic inheritance, and lifetime number of stem cell divisions in a tissue. Folate deficiency is generally associated with increased risk for colorectal cancer (CRC) and acute lymphocytic leukemia (ALL). Conversely, high folic acid (FA) intake has also been associated with higher CRC risk. OBJECTIVE Our objective was to compare the effect of folate intake on mutant frequency (MF) and types of mutations in the colon and bone marrow of mice. METHODS Five-week-old MutaMouse male mice were fed a deficient (0 mg FA/kg), control (2 mg FA/kg), or supplemented (8 mg FA/kg) diet for 20 wk. Tissue MF was assessed using the lacZ mutant assay and comparisons made by 2-factor ANOVA. LacZ mutant plaques were sequenced using next-generation sequencing, and diet-specific mutation profiles within each tissue were compared by Fisher's exact test. RESULTS In the colon, the MF was 1.5-fold and 1.3-fold higher in mice fed the supplemented diet compared with mice fed the control (P = 0.001) and deficient (P = 0.008) diets, respectively. This contrasted with the bone marrow MF in the same mice where the MF was 1.7-fold and 1.6-fold higher in mice fed the deficient diet compared with mice fed the control (P = 0.02) and supplemented (P = 0.03) diets, respectively. Mutation profiles and signatures (mutation context) were tissue-specific. CONCLUSIONS Our data indicate that dietary folate intake affects mutagenesis in a tissue- and dose-specific manner in mice. Mutation profiles were generally tissue- but not dose-specific, suggesting that altered cellular folate status appears to interact with endogenous mutagenic mechanisms in each tissue to create a permissive context in which specific mutation types accumulate. These data illuminate potential mechanisms underpinning differences in observed associations between folate intake/status and cancer.
Collapse
Affiliation(s)
- Stephanie Diaz G
- Nutrition Research Division, Health Canada, Ottawa, Canada.,Department of Biology, Carleton University, Ottawa, Canada
| | | | - Remi Gagné
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | | | - Alex Wong
- Department of Biology, Carleton University, Ottawa, Canada
| | - Francesco Marchetti
- Department of Biology, Carleton University, Ottawa, Canada.,Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Amanda J MacFarlane
- Nutrition Research Division, Health Canada, Ottawa, Canada.,Department of Biology, Carleton University, Ottawa, Canada
| |
Collapse
|
10
|
Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy. Microorganisms 2019; 7:microorganisms7040097. [PMID: 30987157 PMCID: PMC6518429 DOI: 10.3390/microorganisms7040097] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/21/2019] [Accepted: 03/28/2019] [Indexed: 12/12/2022] Open
Abstract
Pelvic radiotherapy has been frequently reported to cause acute and late onset gastrointestinal (GI) toxicities associated with significant morbidity and mortality. Although the underlying mechanisms of pelvic radiation-induced GI toxicity are poorly understood, they are known to involve a complex interplay between all cell types comprising the intestinal wall. Furthermore, increasing evidence states that the human gut microbiome plays a role in the development of radiation-induced health damaging effects. Gut microbial dysbiosis leads to diarrhea and fatigue in half of the patients. As a result, reinforcement of the microbiome has become a hot topic in various medical disciplines. To counteract GI radiotoxicities, apart from traditional pharmacological compounds, adjuvant therapies are being developed including food supplements like vitamins, prebiotics, and probiotics. Despite the easy, cheap, safe, and feasible approach to protect patients against acute radiation-induced toxicity, clinical trials have yielded contradictory results. In this review, a detailed overview is given of the various clinical, intestinal manifestations after pelvic irradiation as well as the role of the gut microbiome herein. Furthermore, whilst discussing possible strategies to prevent these symptoms, food supplements are presented as auspicious, prophylactic, and therapeutic options to mitigate acute pelvic radiation-induced GI injury by exploring their molecular mechanisms of action.
Collapse
|
11
|
Smith AD, Panickar KS, Urban JF, Dawson HD. Impact of Micronutrients on the Immune Response of Animals. Annu Rev Anim Biosci 2019; 6:227-254. [PMID: 29447473 DOI: 10.1146/annurev-animal-022516-022914] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Vitamins and minerals (micronutrients) play an important role in regulating and shaping an immune response. Deficiencies generally result in inadequate or dysregulated cellular activity and cytokine expression, thereby affecting the immune response. Decreased levels of natural killer, granulocyte, and phagocytic cell activity and T and B cell proliferation and trafficking are associated with inadequate levels of micronutrients, as well as increased susceptibility to various adverse health conditions, including inflammatory disorders, infection, and altered vaccine efficacy. In addition, most studies of micronutrient modulation of immune responses have been done in rodents and humans, thus limiting application to the health and well-being of livestock and companion animals. This exploratory review elucidates the role of vitamins and minerals on immune function and inflammatory responses in animals (pigs, dogs, cats, horses, goats, sheep, and cattle), with reference to rodents and humans.
Collapse
Affiliation(s)
- Allen D Smith
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, Maryland 20705-2350, USA;
| | - Kiran S Panickar
- Science & Technology Center, Hills Pet Nutrition Center, Topeka, Kansas 66617, USA
| | - Joseph F Urban
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, Maryland 20705-2350, USA;
| | - Harry D Dawson
- United States Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, Maryland 20705-2350, USA;
| |
Collapse
|
12
|
Alves da Silva AV, de Castro Oliveira SB, Di Rienzi SC, Brown-Steinke K, Dehan LM, Rood JK, Carreira VS, Le H, Maier EA, Betz KJ, Aihara E, Ley RE, Preidis GA, Shen L, Moore SR. Murine Methyl Donor Deficiency Impairs Early Growth in Association with Dysmorphic Small Intestinal Crypts and Reduced Gut Microbial Community Diversity. Curr Dev Nutr 2018. [PMCID: PMC6324351 DOI: 10.1093/cdn/nzy070] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Folate and choline are essential methyl donor nutrients throughout the life span; however, the adverse effects of combined deficiency on early growth, intestinal epithelial morphology, and the gut microbiome remain only partially understood. Objectives We investigated the effects of dietary folate and choline deficiency on early growth, small intestinal (SI) epithelial architecture, and the gut microbiota of mice. To explore potential mechanisms for adverse effects on gut epithelial morphology, we also evaluated gene expression and DNA methylation in mouse intestinal epithelial organoids (enteroids) maintained in methyl donor–deficient (MDD) conditions. Methods Pregnant dams were administered 1 of 4 diets: 1) control diet (CD−), 2) an isocaloric MDD− diet, or 3) CD+ and 4) MDD+ formulations containing 1% succinylsulfathiazole to inhibit folate-producing gut bacteria. We weaned pups to their dams’ diet at 3 wk of age and monitored body weight and tail length pre- and postweaning. We measured serum folate, SI crypt morphology, and microbiota composition at 7 wk of age. Results Both MDD+ and MDD− diets impaired early ponderal and linear growth, lowered serum folate concentrations, and produced patchy areas of increased crypt depth throughout the SI. Succinylsulfathiazole increased crypt depth independently of diet. MDD or succinylsulfathiazole, alone or in combination, altered the gut microbiome, with decreased Bacteroidales and Clostridiales, increased Lactobacillales and Erysipelotrichaceae taxa, and decreased α-diversity indexes. Enteroids maintained in MDD media displayed dysmorphic crypt domains, altered expression of stem cell and secretory differentiation genes, and decreased DNA methylation of the glycosylation genes Beta-1,4-N-Acetyl-Galactosaminyltransferase-1 (B4galnt1) and Phosphoethanolamine/Phosphocholine-Phosphatase (Phospho1). Conclusion MDD impairs ponderal and linear growth in mice in association with dysmorphic SI crypts and reduced gut microbial diversity. In vitro methyl donor deficiency similarly induced dysmorphic crypts in mouse enteroids in conjunction with altered gene expression and DNA methylation.
Collapse
Affiliation(s)
- Antonio V Alves da Silva
- Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, Brazil
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Stephanie B de Castro Oliveira
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sara C Di Rienzi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | | | - Lauren M Dehan
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jill K Rood
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Hung Le
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Elizabeth A Maier
- Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Kristina J Betz
- Instituto de Biomedicina, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Eitaro Aihara
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Geoffrey A Preidis
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Baylor College of Medicine Houston, TX
| | - Lanlan Shen
- USDA Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| | - Sean R Moore
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
13
|
LeBlanc DP, Behan NA, O'Brien JM, Marchetti F, MacFarlane AJ. Folate deficiency increases chromosomal damage and mutations in hematopoietic cells in the transgenic mutamouse model. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2018; 59:366-374. [PMID: 29668043 DOI: 10.1002/em.22190] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 06/08/2023]
Abstract
Folate deficiency causes megaloblastic anemia and neural tube defects, and is also associated with some cancers. In vitro, folate deficiency increases mutation frequency and genome instability, as well as exacerbates the mutagenic potential of known environmental mutagens. Conversely, it remains unclear whether or not elevated folic acid (FA) intakes are beneficial or detrimental to the induction of DNA mutations and by proxy human health. We used the MutaMouse transgenic model to examine the in vivo effects of FA deficient, control, and supplemented diets on somatic DNA mutant frequency (MF) and genome instability in hematopoietic cells. We also examined the interaction between FA intake and exposure to the known mutagen N-ethyl-N-nitrosourea (ENU) on MF. Male mice were fed the experimental diets for 20 weeks from weaning. Half of the mice from each diet group were gavaged with 50 mg/kg body weight ENU after 10 weeks on diet and remained on their respective diet for an additional 10 weeks. Mice fed a FA-deficient diet had a 1.3-fold increase in normochromatic erythrocyte micronucleus (MN) frequency (P = 0.034), and a doubling of bone marrow lacZ MF (P = 0.035), compared to control-fed mice. Mice exposed to ENU showed significantly higher bone marrow lacZ and Pig-a MF, but there was no effect of FA intake on ENU-induced MF. These data indicate that FA deficiency increases mutations and MN formation in highly proliferative somatic cells, but that FA intake does not mitigate ENU-induced mutations. Also, FA intake above adequacy had no beneficial or detrimental effect on mutations or MN formation. Environ. Mol. Mutagen. 59:366-374, 2018. © 2018 Her Majesty the Queen in Right of Canada 2018.
Collapse
Affiliation(s)
- Danielle P LeBlanc
- Nutrition Research Division, Health Canada, Ottawa, Canada
- Department of Biology, Carleton University, Ottawa, Canada
| | - Nathalie A Behan
- Nutrition Research Division, Health Canada, Ottawa, Canada
- Department of Biology, Carleton University, Ottawa, Canada
| | - Jason M O'Brien
- Department of Biology, Carleton University, Ottawa, Canada
- Environment and Climate Change Canada, National Wildlife Research Centre, Ottawa, Canada
| | - Francesco Marchetti
- Department of Biology, Carleton University, Ottawa, Canada
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Amanda J MacFarlane
- Nutrition Research Division, Health Canada, Ottawa, Canada
- Department of Biology, Carleton University, Ottawa, Canada
| |
Collapse
|
14
|
Abstract
Indiscriminate use of multivitamin/mineral supplements in the general population may be misguided, but patients with chronic Inflammatory Bowel Diseases (IBD) should be monitored and compensated for nutritional deficiencies. Mechanistic links between vitamin/mineral deficiencies and IBD pathology has been found for some micronutrients and normalizing their levels is clinically beneficial. Others, like vitamin A, although instinctively desirable, produced disappointing results. Restoring normal levels of the selected micronutrients requires elevated doses to compensate for defects in absorptive or signaling mechanisms. This article describes some aspects of vitamin and mineral deficiencies in IBD, and summarizes pros and cons of supplementation.
Collapse
Affiliation(s)
- Fayez K Ghishan
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA
| | - Pawel R Kiela
- Department of Pediatrics, University of Arizona, Tucson, AZ, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
15
|
[A two-faced vitamin : Folic acid - prevention or promotion of colon cancer?]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz 2017; 60:332-340. [PMID: 28050621 DOI: 10.1007/s00103-016-2505-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the late 1930s, it was discovered that liver and yeast extracts can be used to correct certain cases of megaloblastic anemia in pregnancy. The factor responsible for this was isolated from spinach leaves in the 1940s, and referred to as folate, a term derived from the Latin word folium for leaf. Folate is considered an essential nutrient for human beings. Folic acid, the synthetic form of the vitamin, is used in dietary supplements, medicines and fortified foods. Since the 1980s, it has been recommended that women who plan to become pregnant and pregnant women during the first trimester of pregnancy take folic acid supplements. This recommendation was based on studies that revealed that periconceptional folic acid supplementation can reduce the risk for neural tube defects (NTDs). Many countries later implemented folic acid fortification programs. The resulting population-wide increase of folic acid intakes led to significant reductions in NTD rates. However, a temporarily increased colorectal cancer incidence has been reported to coincide with the fortification programs in the USA and Canada. On the basis of currently available data from experimental and human studies it can be concluded that the association between folate/folic acid and cancer is rather complex: Folate intake in the range of the dietary reference intake (DRI) is associated with a reduced risk for cancer in healthy populations, whereas high intakes of folic acid might result in an increased risk for cancer incidence or progression in persons with precancerous lesions and under certain conditions. Since no adverse effects have been observed in association with the intake of dietary folate, research activities that aim at investigating cause and effect relationships focus on folic acid.
Collapse
|
16
|
Tang M, Frank DN, Hendricks AE, Ir D, Esamai F, Liechty E, Hambidge KM, Krebs NF. Iron in Micronutrient Powder Promotes an Unfavorable Gut Microbiota in Kenyan Infants. Nutrients 2017; 9:E776. [PMID: 28753958 PMCID: PMC5537890 DOI: 10.3390/nu9070776] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 07/14/2017] [Accepted: 07/16/2017] [Indexed: 12/25/2022] Open
Abstract
Iron supplementation may have adverse health effects in infants, probably through manipulation of the gut microbiome. Previous research in low-resource settings have focused primarily on anemic infants. This was a double blind, randomized, controlled trial of home fortification comparing multiple micronutrient powder (MNP) with and without iron. Six-month-old, non- or mildly anemic, predominantly-breastfed Kenyan infants in a rural malaria-endemic area were randomized to consume: (1) MNP containing 12.5 mg iron (MNP+Fe, n = 13); (2) MNP containing no iron (MNP-Fe, n = 13); or (3) Placebo (CONTROL, n = 7), from 6-9 months of age. Fecal microbiota were profiled by high-throughput bacterial 16S rRNA gene sequencing. Markers of inflammation in serum and stool samples were also measured. At baseline, the most abundant phylum was Proteobacteria (37.6% of rRNA sequences). The proteobacterial genus Escherichia was the most abundant genus across all phyla (30.1% of sequences). At the end of the intervention, the relative abundance of Escherichia significantly decreased in MNP-Fe (-16.05 ± 6.9%, p = 0.05) and CONTROL (-19.75 ± 4.5%, p = 0.01), but not in the MNP+Fe group (-6.23 ± 9%, p = 0.41). The second most abundant genus at baseline was Bifidobacterium (17.3%), the relative abundance of which significantly decreased in MNP+Fe (-6.38 ± 2.5%, p = 0.02) and CONTROL (-8.05 ± 1.46%, p = 0.01), but not in MNP-Fe (-4.27 ± 5%, p = 0.4445). Clostridium increased in MNP-Fe only (1.9 ± 0.5%, p = 0.02). No significant differences were observed in inflammation markers, except for IL-8, which decreased in CONTROL. MNP fortification over three months in non- or mildly anemic Kenyan infants can potentially alter the gut microbiome. Consistent with previous research, addition of iron to the MNP may adversely affect the colonization of potential beneficial microbes and attenuate the decrease of potential pathogens.
Collapse
Affiliation(s)
- Minghua Tang
- Section of Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Daniel N Frank
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Audrey E Hendricks
- Department of Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO 80204, USA.
- Department of Biostatistics and Bioinformatics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO 80045, USA.
| | - Diana Ir
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Fabian Esamai
- School of Medicine, Moi University, P.O. Box 4606, Eldoret 30100, Kenya.
| | - Edward Liechty
- School of Medicine, Indiana University, 705 Riley Hospital Drive, Room 5900, Indianapolis, IN 46202, USA.
| | - K Michael Hambidge
- Section of Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Nancy F Krebs
- Section of Nutrition, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
17
|
Rapozo DCM, Bernardazzi C, de Souza HSP. Diet and microbiota in inflammatory bowel disease: The gut in disharmony. World J Gastroenterol 2017; 23:2124-2140. [PMID: 28405140 PMCID: PMC5374124 DOI: 10.3748/wjg.v23.i12.2124] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/03/2017] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
Bacterial colonization of the gut shapes both the local and the systemic immune response and is implicated in the modulation of immunity in both healthy and disease states. Recently, quantitative and qualitative changes in the composition of the gut microbiota have been detected in Crohn's disease and ulcerative colitis, reinforcing the hypothesis of dysbiosis as a relevant mechanism underlying inflammatory bowel disease (IBD) pathogenesis. Humans and microbes have co-existed and co-evolved for a long time in a mutually beneficial symbiotic association essential for maintaining homeostasis. However, the microbiome is dynamic, changing with age and in response to environmental modifications. Among such environmental factors, food and alimentary habits, progressively altered in modern societies, appear to be critical modulators of the microbiota, contributing to or co-participating in dysbiosis. In addition, food constituents such as micronutrients are important regulators of mucosal immunity, with direct or indirect effects on the gut microbiota. Moreover, food constituents have recently been shown to modulate epigenetic mechanisms, which can result in increased risk for the development and progression of IBD. Therefore, it is likely that a better understanding of the role of different food components in intestinal homeostasis and the resident microbiota will be essential for unravelling the complex molecular basis of the epigenetic, genetic and environment interactions underlying IBD pathogenesis as well as for offering dietary interventions with minimal side effects.
Collapse
|
18
|
Lévesque N, Christensen KE, Van Der Kraak L, Best AF, Deng L, Caldwell D, MacFarlane AJ, Beauchemin N, Rozen R. Murine MTHFD1-synthetase deficiency, a model for the human MTHFD1 R653Q polymorphism, decreases growth of colorectal tumors. Mol Carcinog 2016; 56:1030-1040. [DOI: 10.1002/mc.22568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 08/18/2016] [Accepted: 09/04/2016] [Indexed: 01/11/2023]
Affiliation(s)
- Nancy Lévesque
- Departments of Human Genetics and Pediatrics; The Research Institute of the McGill University Health Centre; McGill University; Montreal Quebec Canada
| | - Karen E. Christensen
- Departments of Human Genetics and Pediatrics; The Research Institute of the McGill University Health Centre; McGill University; Montreal Quebec Canada
| | - Lauren Van Der Kraak
- Departments of Biochemistry; Goodman Cancer Research Centre, Medicine and Oncology; McGill University; Montreal Quebec Canada
| | - Ana F. Best
- Department of Mathematics and Statistics; McGill University; Montreal Quebec Canada
| | - Liyuan Deng
- Departments of Human Genetics and Pediatrics; The Research Institute of the McGill University Health Centre; McGill University; Montreal Quebec Canada
| | - Don Caldwell
- Nutrition Research Division; Health Canada; Ottawa Ontario Canada
| | | | - Nicole Beauchemin
- Departments of Biochemistry; Goodman Cancer Research Centre, Medicine and Oncology; McGill University; Montreal Quebec Canada
| | - Rima Rozen
- Departments of Human Genetics and Pediatrics; The Research Institute of the McGill University Health Centre; McGill University; Montreal Quebec Canada
| |
Collapse
|
19
|
Wędrychowicz A, Zając A, Tomasik P. Advances in nutritional therapy in inflammatory bowel diseases: Review. World J Gastroenterol 2016; 22:1045-1066. [PMID: 26811646 PMCID: PMC4716019 DOI: 10.3748/wjg.v22.i3.1045] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 07/22/2015] [Accepted: 09/13/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease are chronic, life-long, and relapsing diseases of the gastrointestinal tract. Currently, there are no complete cure possibilities, but combined pharmacological and nutritional therapy may induce remission of the disease. Malnutrition and specific nutritional deficiencies are frequent among IBD patients, so the majority of them need nutritional treatment, which not only improves the state of nutrition of the patients but has strong anti-inflammatory activity as well. Moreover, some nutrients, from early stages of life are suspected as triggering factors in the etiopathogenesis of IBD. Both parenteral and enteral nutrition is used in IBD therapy, but their practical utility in different populations and in different countries is not clearly established, and there are sometimes conflicting theories concerning the role of nutrition in IBD. This review presents the actual data from research studies on the influence of nutrition on the etiopathogenesis of IBD and the latest findings regarding its mechanisms of action. The use of both parenteral and enteral nutrition as therapeutic methods in induction and maintenance therapy in IBD treatment is also extensively discussed. Comparison of the latest research data, scientific theories concerning the role of nutrition in IBD, and different opinions about them are also presented and discussed. Additionally, some potential future perspectives for nutritional therapy are highlighted.
Collapse
|
20
|
Abstract
For many years folic acid has been evaluated for its utility as a chemopreventive agent due to its position at the center of the one-carbon metabolic network. This network is responsible for generating precursors to nucleotide synthesis as well as the one-carbon moieties used in DNA methylation reactions, two mechanisms which are frequently disrupted during carcinogenesis. While the use of folic acid for the chemoprevention of colorectal cancer is still controversial, there is evidence that folic acid intake has significant influence on these fundamental cellular mechanisms. Folic acid has a dual role with regards to nucleotide synthesis and colorectal cancer (CRC) prevention; in a healthy colon, adequate folate status is important for nucleotide metabolism homeostasis and the maintenance of DNA integrity, however in a colon harboring premalignant lesions lowered folate status may help to eliminate transformed cells. In addition, folic acid is important for the generation of the one-carbon groups used in DNA methylation reactions, and modulation of folic acid metabolism may be useful in combating the aberrant DNA methylation during carcinogenesis. Interestingly, it has been revealed that decreased folic acid intake can dampen the inflammatory response, which has recently been a popular strategy for colorectal cancer chemoprevention. In this review we discuss the molecular mechanisms influenced by folic acid intake and how they might be relevant to cancer chemoprevention in greater detail.
Collapse
|
21
|
Christensen KE, Mikael LG, Leung KY, Lévesque N, Deng L, Wu Q, Malysheva OV, Best A, Caudill MA, Greene NDE, Rozen R. High folic acid consumption leads to pseudo-MTHFR deficiency, altered lipid metabolism, and liver injury in mice. Am J Clin Nutr 2015; 101:646-58. [PMID: 25733650 PMCID: PMC4340065 DOI: 10.3945/ajcn.114.086603] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 12/05/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Increased consumption of folic acid is prevalent, leading to concerns about negative consequences. The effects of folic acid on the liver, the primary organ for folate metabolism, are largely unknown. Methylenetetrahydrofolate reductase (MTHFR) provides methyl donors for S-adenosylmethionine (SAM) synthesis and methylation reactions. OBJECTIVE Our goal was to investigate the impact of high folic acid intake on liver disease and methyl metabolism. DESIGN Folic acid-supplemented diet (FASD, 10-fold higher than recommended) and control diet were fed to male Mthfr(+/+) and Mthfr(+/-) mice for 6 mo to assess gene-nutrient interactions. Liver pathology, folate and choline metabolites, and gene expression in folate and lipid pathways were examined. RESULTS Liver and spleen weights were higher and hematologic profiles were altered in FASD-fed mice. Liver histology revealed unusually large, degenerating cells in FASD Mthfr(+/-) mice, consistent with nonalcoholic fatty liver disease. High folic acid inhibited MTHFR activity in vitro, and MTHFR protein was reduced in FASD-fed mice. 5-Methyltetrahydrofolate, SAM, and SAM/S-adenosylhomocysteine ratios were lower in FASD and Mthfr(+/-) livers. Choline metabolites, including phosphatidylcholine, were reduced due to genotype and/or diet in an attempt to restore methylation capacity through choline/betaine-dependent SAM synthesis. Expression changes in genes of one-carbon and lipid metabolism were particularly significant in FASD Mthfr(+/-) mice. The latter changes, which included higher nuclear sterol regulatory element-binding protein 1, higher Srepb2 messenger RNA (mRNA), lower farnesoid X receptor (Nr1h4) mRNA, and lower Cyp7a1 mRNA, would lead to greater lipogenesis and reduced cholesterol catabolism into bile. CONCLUSIONS We suggest that high folic acid consumption reduces MTHFR protein and activity levels, creating a pseudo-MTHFR deficiency. This deficiency results in hepatocyte degeneration, suggesting a 2-hit mechanism whereby mutant hepatocytes cannot accommodate the lipid disturbances and altered membrane integrity arising from changes in phospholipid/lipid metabolism. These preliminary findings may have clinical implications for individuals consuming high-dose folic acid supplements, particularly those who are MTHFR deficient.
Collapse
Affiliation(s)
- Karen E Christensen
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Leonie G Mikael
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Kit-Yi Leung
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Nancy Lévesque
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Liyuan Deng
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Qing Wu
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Olga V Malysheva
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Ana Best
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Marie A Caudill
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Nicholas D E Greene
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| | - Rima Rozen
- From the Departments of Human Genetics and Pediatrics, McGill University, and the Montreal Children's Hospital site of the McGill University Health Centre Research Institute, Montreal, Quebec, Canada (KEC, LGM, NL, LD, QW, and RR); Developmental Biology and Cancer Programme, Institute of Child Health, University College London, London, United Kingdom (K-YL and NDEG); the Division of Nutritional Sciences and Genomics, Cornell University, Ithaca, NY (OVM and MAC); and the Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada (AB)
| |
Collapse
|
22
|
Abstract
Most of what is known about the pathogenesis of inflammatory bowel disease (IBD) pertains to complex interplay between host genetics, immunity, and environmental factors. Epigenetic modifications play pivotal roles in intestinal immunity and mucosal homeostasis as well as mediating gene-environment interactions. In this article, we provide a historical account of epigenetic research either directly related or pertinent to the pathogenesis and management of IBD. We further collate emerging evidence supporting roles for epigenetic mechanisms in relevant aspects of IBD biology, including deregulated immunity, host-pathogen recognition and mucosal integrity. Finally, we highlight key epigenetic mechanisms that link chronic inflammation to specific IBD comorbidities, including colitis-associated cancer and discuss their potential utility as novel biomarkers or pharmacologic targets in IBD therapy.
Collapse
|
23
|
Leddin D, Tamim H, Levy AR. Is folate involved in the pathogenesis of inflammatory bowel disease? Med Hypotheses 2013; 81:940-1. [PMID: 24045091 DOI: 10.1016/j.mehy.2013.08.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 08/16/2013] [Accepted: 08/23/2013] [Indexed: 02/08/2023]
Abstract
The inflammatory bowel diseases, Crohn's and ulcerative colitis, are common and a significant cause of morbidity. They were rare before the 1930's but the incidence has been increasing in both developed and developing countries. We have recently reported that the incidence in Nova Scotia, the area with one of the highest reported burden globally, is decreasing since 1997. We postulate that this decrease may be due to the addition of folate to cereals. This was mandated in 1998 but the process of fortification began in 1997. There is circumstantial evidence from epidemiology studies that a diet deficient in folate may have contributed to the global rise in these diseases. This hypothesis, if proven to be correct, has important implications for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Desmond Leddin
- Department of Medicine, Dalhousie University, Halifax, Canada.
| | | | | |
Collapse
|
24
|
Low D, Mizoguchi A, Mizoguchi E. DNA methylation in inflammatory bowel disease and beyond. World J Gastroenterol 2013; 19:5238-5249. [PMID: 23983426 PMCID: PMC3752557 DOI: 10.3748/wjg.v19.i32.5238] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/13/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a consequence of the complex, dysregulated interplay between genetic predisposition, environmental factors, and microbial composition in the intestine. Despite a great advancement in identifying host-susceptibility genes using genome-wide association studies (GWAS), the majority of IBD cases are still underrepresented. The immediate challenge in post-GWAS era is to identify other causative genetic factors of IBD. DNA methylation has received increasing attention for its mechanistical role in IBD pathogenesis. This stable, yet dynamic DNA modification, can directly affect gene expression that have important implications in IBD development. The alterations in DNA methylation associated with IBD are likely to outset as early as embryogenesis all the way until old-age. In this review, we will discuss the recent advancement in understanding how DNA methylation alterations can contribute to the development of IBD.
Collapse
|