151
|
Iheozor-Ejiofor Z, Gordon M, Iqbal T, Allen P, Hoque S, Sinopoulou V, Engineer J, Akobeng AK. Interventions for treating iron deficiency anaemia in inflammatory bowel disease: a network meta-analysis. Hippokratia 2020. [DOI: 10.1002/14651858.cd013529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Zipporah Iheozor-Ejiofor
- Centre for Musculoskeletal Research, Institute of Inflammation and Repair, The University of Manchester; Cochrane Bone Joint and Muscle Trauma Group; Stopford Building, Oxford Road Manchester Oxford Road Manchester UK M13 9PL
| | - Morris Gordon
- University of Central Lancashire; School of Medicine; Preston Lancashire UK
| | - Tariq Iqbal
- Queen Elizabeth Hospital; University Hospitals Birmingham NHS Foundation Trust Birmingham UK
| | - Patrick Allen
- Ulster Hospital; Department of Gastroenterology and Hepatology; Belfast Ireland
| | | | | | | | | |
Collapse
|
152
|
Chen JQ, Chen YY, Tao HJ, Pu ZJ, Shi XQ, Zhang J, Tan YJ, Yue SJ, Zhou GS, Shang EX, Tang YP, Duan JA. An integrated metabolomics strategy to reveal dose-effect relationship and therapeutic mechanisms of different efficacy of rhubarb in constipation rats. J Pharm Biomed Anal 2020; 177:112837. [DOI: 10.1016/j.jpba.2019.112837] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/19/2019] [Accepted: 08/25/2019] [Indexed: 01/23/2023]
|
153
|
Grau Amorós J. Is oral iron always the best option? Rev Clin Esp 2020. [DOI: 10.1016/j.rceng.2019.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
154
|
Grau Amorós J. ¿Es siempre el tratamiento con hierro oral la mejor opción terapéutica? Rev Clin Esp 2020; 220:27-28. [DOI: 10.1016/j.rce.2019.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 11/15/2019] [Indexed: 10/25/2022]
|
155
|
Ikee R, Yano K, Tsuru T. Constipation in chronic kidney disease: it is time to reconsider. RENAL REPLACEMENT THERAPY 2019. [DOI: 10.1186/s41100-019-0246-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractConstipation is highly prevalent in patients with chronic kidney disease (CKD) and is primarily characterized by decreased intestinal motility. This chronic disorder affects the quality of life of patients. However, nephrologist and dialysis clinicians have long had a disproportionately limited understanding of constipation. Accumulating evidence has revealed a relationship between constipation and cardiovascular disease and CKD. The pathogenesis of constipation in CKD patients is multifactorial: decreased physical activity, comorbidities affecting bowel movement, such as diabetes mellitus, cerebrovascular disease, and hyperparathyroidism, a restricted dietary intake of plant-based fiber-rich foods, and multiple medications, including phosphate binders and potassium-binding resins, have all been implicated. CKD is associated with alterations in the composition and function of the gut microbiota, so-called gut dysbiosis. Recent studies showed that CKD-related gut dysbiosis decreased intestinal motility via intestinal inflammation or the increased generation of gut-derived uremic toxins, such as indoxyl sulfate and p-cresyl sulfate. Furthermore, the gastrointestinal secretion of mucin was found to be decreased in CKD animal models, which may delay colonic transit by diminished lubrication in the alimentary tract. Thus, CKD-related gut dysbiosis may play a role in constipation, but limited information is currently available. Since constipation is often intractable, particularly in CKD patients, every available means needs to be employed in its treatment. The effects of probiotics, prebiotics, and synbiotics on the composition of the gut microbiota and gut-derived uremic toxins have been increasingly reported. However, their effects on stool consistency or frequency in CKD patients remain unclear. Some laxatives may be beneficial for improving not only bowel habits but also gut dysbiosis. Further studies are required to elucidate the CKD-specific pathogenesis of constipation and develop novel effective treatment options.
Collapse
|
156
|
Minor EA, Kupec JT, Nickerson AJ, Narayanan K, Rajendran VM. Increased DMT1 and FPN1 expression with enhanced iron absorption in ulcerative colitis human colon. Am J Physiol Cell Physiol 2019; 318:C263-C271. [PMID: 31721611 DOI: 10.1152/ajpcell.00128.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Iron deficiency anemia is a common complication of ulcerative colitis (UC) that can profoundly impact quality of life. Most iron absorption occurs in the duodenum via divalent metal transporter 1 (DMT1)-mediated uptake and ferroportin-1 (FPN1)-mediated export across the apical and basolateral membranes, respectively. However, the colon also contains iron transporters and can participate in iron absorption. Studies have shown increased duodenal DMT1 and FPN1 in patients with UC, but there is conflicting evidence about whether expression is altered in UC colon. We hypothesized that expression of colonic DMT1 and FPN1 will also increase to compensate for iron deficiency. Quantitative RT-PCR and Western blot analyses were performed on duodenal and colonic segmental (right colon, transverse colon, left colon, and rectum) biopsies obtained during colonoscopy. DMT1 mRNA and protein abundances in colonic segments were approximately equal to those in the duodenum, whereas colonic FPN1 mRNA and protein abundances of colonic segments were about one-quarter of those of the duodenum. DMT1 specific mRNA and protein abundances were increased twofold, whereas FPN1 mRNA and protein expressions were increased fivefold in UC distal colon. Immunofluorescence studies revealed enhanced expression of apical membrane- and basolateral membrane-localized DMT1 and FPN1 in UC human colon, respectively. Increased DMT1 expression was associated with enhanced 2-(3-carbamimidoylsulfanylmethyl-benzyl)-isothiourea (CISMBI, DMT1 specific inhibitor)-sensitive 59Fe uptake in UC human colon. We conclude from these results that patients with active UC have increased expression of colonic iron transporters and increased iron absorption, which may be targeted in the treatment of UC-related anemia.
Collapse
Affiliation(s)
- Emily A Minor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia.,Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Justin T Kupec
- Department of Medicine, Digestive Diseases Section, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Andrew J Nickerson
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, West Virginia.,Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Karthikeyan Narayanan
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Vazhaikkurichi M Rajendran
- Department of Biochemistry, West Virginia University School of Medicine, Morgantown, West Virginia.,Department of Medicine, Digestive Diseases Section, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
157
|
Indigo Naturalis Ameliorates Dextran Sulfate Sodium-Induced Colitis in Mice by Modulating the Intestinal Microbiota Community. Molecules 2019; 24:molecules24224086. [PMID: 31726738 PMCID: PMC6891465 DOI: 10.3390/molecules24224086] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/18/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Indigo naturalis (IN) is a traditional Chinese medicine, named Qing-Dai, which is extracted from indigo plants and has been used to treat patients with inflammatory bowel disease (IBD) in China and Japan. Though there are notable effects of IN on colitis, the mechanisms remain elusive. Regarding the significance of alterations of intestinal flora related to IBD and the poor water solubility of the blue IN powder, we predicted that the protective action of IN on colitis may occur through modifying gut microbiota. To investigate the relationships of IN, colitis, and gut microbiomes, a dextran sulfate sodium (DSS)-induced mice colitis model was tested to explore the protective effects of IN on macroscopic colitis symptoms, the histopathological structure, inflammation cytokines, and gut microbiota, and their potential functions. Sulfasalazine (SASP) was used as the positive control. Firstly, because it was a mixture, the main chemical compositions of indigo and indirubin in IN were detected by ultra-performance liquid chromatography (UPLC). The clinical activity score (CAS), hematoxylin and eosin (H&E) staining results, and enzyme-linked immunosorbent assay (ELISA) results in this study showed that IN greatly improved the health conditions of the tested colitis mice, ameliorated the histopathological structure of the colon tissue, down-regulated pro-inflammatory cytokines, and up-regulated anti-inflammatory cytokines. The results of 16S rDNA sequences analysis with the Illumina MiSeq platform showed that IN could modulate the balance of gut microbiota, especially by down-regulating the relative quantity of Turicibacter and up-regulating the relative quantity of Peptococcus. The therapeutic effect of IN may be closely related to the anaerobic gram-positive bacteria of Turicibacter and Peptococcus. The inferred metagenomes from 16S data using PICRUSt demonstrated that decreased metabolic genes, such as through biosynthesis of siderophore group nonribosomal peptides, non-homologous end-joining, and glycosphingolipid biosynthesis of lacto and neolacto series, may maintain microbiota homeostasis during inflammation from IN treatment in DSS-induced colitis.
Collapse
|
158
|
Waters JL, Ley RE. The human gut bacteria Christensenellaceae are widespread, heritable, and associated with health. BMC Biol 2019; 17:83. [PMID: 31660948 PMCID: PMC6819567 DOI: 10.1186/s12915-019-0699-4] [Citation(s) in RCA: 419] [Impact Index Per Article: 69.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
The Christensenellaceae, a recently described family in the phylum Firmicutes, is emerging as an important player in human health. The relative abundance of Christensenellaceae in the human gut is inversely related to host body mass index (BMI) in different populations and multiple studies, making its relationship with BMI the most robust and reproducible link between the microbial ecology of the human gut and metabolic disease reported to date. The family is also related to a healthy status in a number of other different disease contexts, including obesity and inflammatory bowel disease. In addition, Christensenellaceae is highly heritable across multiple populations, although specific human genes underlying its heritability have so far been elusive. Further research into the microbial ecology and metabolism of these bacteria should reveal mechanistic underpinnings of their host-health associations and enable their development as therapeutics.
Collapse
Affiliation(s)
- Jillian L Waters
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076, Tuebingen, Germany
| | - Ruth E Ley
- Department of Microbiome Science, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076, Tuebingen, Germany.
| |
Collapse
|
159
|
Nealon NJ, Parker KD, Lahaie P, Ibrahim H, Maurya AK, Raina K, Ryan EP. Bifidobacterium longum-fermented rice bran and rice bran supplementation affects the gut microbiome and metabolome. Benef Microbes 2019; 10:823-839. [PMID: 31965839 DOI: 10.3920/bm2019.0017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This study investigated gut microbiota composition along with food, host, and microbial derived metabolites in the colon and systemic circulation of healthy mice following dietary rice bran and fermented rice bran intake. Adult male BALB/c mice were fed a control diet or one of two experimental diets containing 10% w/w rice bran fermented by Bifidobacterium longum or 10% w/w non-fermented rice bran for 15 weeks. Metabolomics was performed on the study diets (food), the murine colon and whole blood. These were analysed in concert with 16S rRNA amplicon sequencing of faeces, caecum, and colon microbiomes. Principal components analysis of murine microbiota composition displayed marked separation between control and experimental diets, and between faecal and tissue (caecum and colon) microbiomes. Colon and caecal microbiomes in both experimental diet groups showed enrichment of Roseburia, Lachnospiraceae, and Clostridiales related amplicon sequence variants compared to control. Bacterial composition was largely similar between experimental diets. Metabolite profiling revealed 530 small molecules comprising of 39% amino acids and 21% lipids that had differential abundances across food, colon, and blood matrices, and statistically significant between the control, rice bran, and fermented rice bran groups. The amino acid metabolite, N-delta-acetylornithine, was notably increased by B. longum rice bran fermentation when compared to non-fermented rice bran in food, colon, and blood. These findings support that dietary intake of rice bran fermented with B. longum modulates multiple metabolic pathways important to the gut and overall health.
Collapse
Affiliation(s)
- N J Nealon
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA.,Program in Cellular and Molecular Biology, Colorado State University, Fort Collins, 80521 CO, USA
| | - K D Parker
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA
| | - P Lahaie
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA
| | - H Ibrahim
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA.,Zagazig University, Department of Medical Biochemistry, Faculty of Medicine, 44511 Zagazig, Egypt
| | - A K Maurya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Raina
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO 80045, USA.,Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - E P Ryan
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80521, USA.,Program in Cellular and Molecular Biology, Colorado State University, Fort Collins, 80521 CO, USA.,University of Colorado Cancer Center, Division of Cancer Control and Prevention, Aurora, CO 80045, USA
| |
Collapse
|
160
|
La Carpia F, Wojczyk BS, Annavajhala MK, Rebbaa A, Culp-Hill R, D’Alessandro A, Freedberg DE, Uhlemann AC, Hod EA. Transfusional iron overload and intravenous iron infusions modify the mouse gut microbiota similarly to dietary iron. NPJ Biofilms Microbiomes 2019; 5:26. [PMID: 31583109 PMCID: PMC6760189 DOI: 10.1038/s41522-019-0097-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/12/2019] [Indexed: 01/19/2023] Open
Abstract
Iron is essential for both microorganisms and their hosts. Although effects of dietary iron on gut microbiota have been described, the effect of systemic iron administration has yet to be explored. Here, we show that dietary iron, intravenous iron administration, and chronic transfusion in mice increase the availability of iron in the gut. These iron interventions have consistent and reproducible effects on the murine gut microbiota; specifically, relative abundance of the Parabacteroides and Lactobacillus genera negatively correlate with increased iron stores, whereas members of the Clostridia class positively correlate with iron stores regardless of the route of iron administration. Iron levels also affected microbial metabolites, in general, and indoles, in particular, circulating in host plasma and in stool pellets. Taken together, these results suggest that by shifting the balance of the microbiota, clinical interventions that affect iron status have the potential to alter biologically relevant microbial metabolites in the host.
Collapse
Affiliation(s)
- Francesca La Carpia
- Department of Pathology and Cell biology, Columbia University Irving Medical Center, New York, NY USA
| | - Boguslaw S. Wojczyk
- Department of Pathology and Cell biology, Columbia University Irving Medical Center, New York, NY USA
| | - Medini K. Annavajhala
- Department of Medicine, Columbia University, Irving Medical Center-New York Presbyterian Hospital, New York, NY USA
- Columbia Medicine Microbiome and Pathogen Genomic core, Columbia University Irving Medical Center, New York, NY USA
| | - Abdelhadi Rebbaa
- Department of Pathology and Cell biology, Columbia University Irving Medical Center, New York, NY USA
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado USA
| | - Daniel E. Freedberg
- Department of Medicine, Columbia University, Irving Medical Center-New York Presbyterian Hospital, New York, NY USA
| | - Anne-Catrin Uhlemann
- Department of Medicine, Columbia University, Irving Medical Center-New York Presbyterian Hospital, New York, NY USA
| | - Eldad A. Hod
- Department of Pathology and Cell biology, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
161
|
Thingholm LB, Rühlemann MC, Koch M, Fuqua B, Laucke G, Boehm R, Bang C, Franzosa EA, Hübenthal M, Rahnavard A, Frost F, Lloyd-Price J, Schirmer M, Lusis AJ, Vulpe CD, Lerch MM, Homuth G, Kacprowski T, Schmidt CO, Nöthlings U, Karlsen TH, Lieb W, Laudes M, Franke A, Huttenhower C. Obese Individuals with and without Type 2 Diabetes Show Different Gut Microbial Functional Capacity and Composition. Cell Host Microbe 2019; 26:252-264.e10. [PMID: 31399369 DOI: 10.1016/j.chom.2019.07.004] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/17/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
Obesity and type 2 diabetes (T2D) are metabolic disorders that are linked to microbiome alterations. However, their co-occurrence poses challenges in disentangling microbial features unique to each condition. We analyzed gut microbiomes of lean non-diabetic (n = 633), obese non-diabetic (n = 494), and obese individuals with T2D (n = 153) from German population and metabolic disease cohorts. Microbial taxonomic and functional profiles were analyzed along with medical histories, serum metabolomics, biometrics, and dietary data. Obesity was associated with alterations in microbiome composition, individual taxa, and functions with notable changes in Akkermansia, Faecalibacterium, Oscillibacter, and Alistipes, as well as in serum metabolites that correlated with gut microbial patterns. However, microbiome associations were modest for T2D, with nominal increases in Escherichia/Shigella. Medications, including antihypertensives and antidiabetics, along with dietary supplements including iron, were significantly associated with microbiome variation. These results differentiate microbial components of these interrelated metabolic diseases and identify dietary and medication exposures to consider in future studies.
Collapse
Affiliation(s)
- Louise B Thingholm
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Malte C Rühlemann
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Manja Koch
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Brie Fuqua
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Guido Laucke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Ruwen Boehm
- Institute of Experimental and Clinical Pharmacology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Eric A Franzosa
- Biostatistics Department, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
| | - Matthias Hübenthal
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany; Department of Dermatology, Venereology and Allergy, University Hospital, Schleswig-Holstein, 24105 Kiel, Germany
| | - Ali Rahnavard
- Biostatistics Department, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
| | - Fabian Frost
- Department of Medicine A, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Jason Lloyd-Price
- Biostatistics Department, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
| | - Melanie Schirmer
- Biostatistics Department, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
| | - Aldons J Lusis
- Department of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Chris D Vulpe
- College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Georg Homuth
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Tim Kacprowski
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany; Research Group on Computational Systems Medicine, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Weihenstephan, Technical University of Munich, Freising-Weihenstephan 85354, Germany
| | - Carsten O Schmidt
- Institute for Community Medicine SHIP-KEF, University Medicine Greifswald, Greifswald 17475, Germany
| | - Ute Nöthlings
- Department of Nutrition and Food Sciences, Nutritional Epidemiology, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Tom H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine and Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital, Rikshospitalet, 0372 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0315 Oslo, Norway
| | - Wolfgang Lieb
- Institute of Epidemiology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany
| | - Matthias Laudes
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany.
| | - Curtis Huttenhower
- Biostatistics Department, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; The Broad Institute of MIT and Harvard, Cambridge, MA 02115, USA
| |
Collapse
|
162
|
Stevens AJ, Purcell RV, Darling KA, Eggleston MJF, Kennedy MA, Rucklidge JJ. Human gut microbiome changes during a 10 week Randomised Control Trial for micronutrient supplementation in children with attention deficit hyperactivity disorder. Sci Rep 2019; 9:10128. [PMID: 31300667 PMCID: PMC6625977 DOI: 10.1038/s41598-019-46146-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 06/20/2019] [Indexed: 12/20/2022] Open
Abstract
It has been widely hypothesized that both diet and the microbiome play a role in the regulation of attention-deficit/hyperactivity disorder (ADHD) behaviour. However, there has been very limited scientific investigation into the potential biological connection. We performed a 10-week pilot study investigating the effects of a broad spectrum micronutrient administration on faecal microbiome content, using 16S rRNA gene sequencing. The study consisted of 17 children (seven in the placebo and ten in the treatment group) between the ages of seven and 12 years, who were diagnosed with ADHD. We found that micronutrient treatment did not drive large-scale changes in composition or structure of the microbiome. However, observed OTUs significantly increased in the treatment group, and showed no mean change in the placebo group. The differential abundance and relative frequency of Actinobacteria significantly decreased post- micronutrient treatment, and this was largely attributed to species from the genus Bifidobacterium. This was compensated by an increase in the relative frequency of species from the genus Collinsella. Further research is required to establish the role that Bifidobacterium contribute towards neuropsychiatric disorders; however, these findings suggest that micronutrient administration could be used as a safe, therapeutic method to modulate Bifidobacterium abundance, which could have potential implications for modulating and regulating ADHD behaviour. Our pilot study provides an initial observation into this area of research, and highlights an interesting avenue for further investigation in a larger cohort. Furthermore, these novel results provide a basis for future research on the biological connection between ADHD, diet and the microbiome.
Collapse
Affiliation(s)
- Aaron J Stevens
- Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand.
| | - Rachel V Purcell
- Department of Surgery, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand
| | - Kathryn A Darling
- Department of Psychology, University of Canterbury, Christchurch, New Zealand
| | - Matthew J F Eggleston
- Mental Health Division, Canterbury District Health Board, Private Bag 4733, Christchurch, New Zealand
| | - Martin A Kennedy
- Department of Pathology and Biomedical Science, University of Otago Christchurch, P.O. Box 4345, Christchurch, New Zealand
| | - Julia J Rucklidge
- Department of Psychology, University of Canterbury, Christchurch, New Zealand
| |
Collapse
|
163
|
Verhoog S, Taneri PE, Roa Díaz ZM, Marques-Vidal P, Troup JP, Bally L, Franco OH, Glisic M, Muka T. Dietary Factors and Modulation of Bacteria Strains of Akkermansia muciniphila and Faecalibacterium prausnitzii: A Systematic Review. Nutrients 2019; 11:nu11071565. [PMID: 31336737 PMCID: PMC6683038 DOI: 10.3390/nu11071565] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
Akkermansia muciniphila and Faecalibacterium prausnitzii are highly abundant human gut microbes in healthy individuals, and reduced levels are associated with inflammation and alterations of metabolic processes involved in the development of type 2 diabetes. Dietary factors can influence the abundance of A. muciniphila and F. prausnitzii, but the evidence is not clear. We systematically searched PubMed and Embase to identify clinical trials investigating any dietary intervention in relation to A. muciniphila and F. prausnitzii. Overall, 29 unique trials were included, of which five examined A. muciniphila, 19 examined F. prausnitzii, and six examined both, in a total of 1444 participants. A caloric restriction diet and supplementation with pomegranate extract, resveratrol, polydextrose, yeast fermentate, sodium butyrate, and inulin increased the abundance of A. muciniphila, while a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols decreased the abundance of A. muciniphila. For F. prausnitzii, the main studied intervention was prebiotics (e.g. fructo-oligosaccharides, inulin type fructans, raffinose); seven studies reported an increase after prebiotic intervention, while two studies reported a decrease, and four studies reported no difference. Current evidence suggests that some dietary factors may influence the abundance of A. muciniphila and F. prausnitzii. However, more research is needed to support these microflora strains as targets of microbiome shifts with dietary intervention and their use as medical nutrition therapy in prevention and management of chronic disease.
Collapse
Affiliation(s)
- Sanne Verhoog
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland.
| | - Petek Eylul Taneri
- Corlu Cancer Early Diognosis and Training Center, 59100 Tekirdag, Turkey
| | - Zayne M Roa Díaz
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
| | - John P Troup
- Standard Process Inc Nutrition Innovation Center, Kannapolis, NC 28018, USA
| | - Lia Bally
- Department of Diabetes, Endocrinology, Clinical Nutrition and Metabolism, Bern University Hospital, 3010 Bern, Switzerland
| | - Oscar H Franco
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
| | - Marija Glisic
- Leibniz Institute for Prevention Research and Epidemiology-BIPS, 28359 Bremen, Germany
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, 3012 Bern, Switzerland
| |
Collapse
|
164
|
Markota A, Metzger R, Heiseke AF, Jandl L, Dursun E, Eisenächer K, Reindl W, Haller D, Krug AB. Comparison of iron-reduced and iron-supplemented semisynthetic diets in T cell transfer colitis. PLoS One 2019; 14:e0218332. [PMID: 31276514 PMCID: PMC6611680 DOI: 10.1371/journal.pone.0218332] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/30/2019] [Indexed: 12/18/2022] Open
Abstract
Clinical observations in inflammatory bowel disease patients and experimental studies in rodents suggest that iron in the intestinal lumen derived from iron-rich food or oral iron supplementation could exacerbate inflammation and that iron depletion from the diet could be protective. To test the hypothesis that dietary iron reduction is protective against colitis development, the impact of iron reduction in the diet below 10 mg/kg on the course of CD4+ CD62L+ T cell transfer colitis was investigated in adult C57BL/6 mice. Weight loss as well as clinical and histological signs of inflammation were comparable between mice pretreated with semisynthetic diets with either < 10mg/kg iron content or supplemented with 180 mg/kg iron in the form of ferrous sulfate or hemin. Accumulation and activation of Ly6Chigh monocytes, changes in dendritic cell subset composition and induction of proinflammatory Th1/Th17 cells in the inflamed colon were not affected by the iron content of the diets. Thus, dietary iron reduction did not protect adult mice against severe intestinal inflammation in T cell transfer induced colitis.
Collapse
Affiliation(s)
- Anamarija Markota
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Rebecca Metzger
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Alexander F. Heiseke
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Lisa Jandl
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Ezgi Dursun
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Katharina Eisenächer
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Wolfgang Reindl
- Klinikum Mannheim, II. Medizinische Klinik, Mannheim, Germany
| | - Dirk Haller
- Chair for Nutrition and Immunology, Technical University Munich, Freising, Germany
| | - Anne B. Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilians-University Munich, Martinsried, Germany
- * E-mail:
| |
Collapse
|
165
|
Cheng C, Huang DC, Zhao LY, Cao CJ, Chen GT. Preparation and in vitro absorption studies of a novel polysaccharide‑iron (III) complex from Flammulina velutipes. Int J Biol Macromol 2019; 132:801-810. [DOI: 10.1016/j.ijbiomac.2019.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022]
|
166
|
Ellermann M, Gharaibeh RZ, Maharshak N, Peréz-Chanona E, Jobin C, Carroll IM, Arthur JC, Plevy SE, Fodor AA, Brouwer CR, Sartor RB. Dietary iron variably modulates assembly of the intestinal microbiota in colitis-resistant and colitis-susceptible mice. Gut Microbes 2019; 11:32-50. [PMID: 31179826 PMCID: PMC6973310 DOI: 10.1080/19490976.2019.1599794] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/30/2019] [Accepted: 03/05/2019] [Indexed: 02/03/2023] Open
Abstract
Iron deficiency, a common comorbidity of gastrointestinal inflammatory disorders such as inflammatory bowel diseases (IBD), is often treated with oral iron supplementation. However, the safety of oral iron supplementation remains controversial because of its association with exacerbated disease activity in a subset of IBD patients. Because iron modulates bacterial growth and function, one possible mechanism by which iron may exacerbate inflammation in susceptible hosts is by modulating the intestinal microbiota. We, therefore, investigated the impact of dietary iron on the intestinal microbiota, utilizing the conventionalization of germ-free mice as a model of a microbial community in compositional flux to recapitulate the instability of the IBD-associated intestinal microbiota. Our findings demonstrate that altering intestinal iron availability during community assembly modulated the microbiota in non-inflamed wild type (WT) and colitis-susceptible interleukin-10-deficient (Il10-/-) mice. Depletion of luminal iron availability promoted luminal compositional changes associated with dysbiotic states irrespective of host genotype, including an expansion of Enterobacteriaceae such as Escherichia coli. Mechanistic in vitro growth competitions confirmed that high-affinity iron acquisition systems in E. coli enhance its abundance over other bacteria in iron-restricted conditions, thereby enabling pathobiont iron scavenging during dietary iron restriction. In contrast, distinct luminal community assembly was observed with dietary iron supplementation in WT versus Il10-/- mice, suggesting that the effects of increased iron on the microbiota differ with host inflammation status. Taken together, shifts in dietary iron intake during community assembly modulate the ecological structure of the intestinal microbiota and is dependent on host genotype and inflammation status.
Collapse
Affiliation(s)
- Melissa Ellermann
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Raad Z Gharaibeh
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Kannapolis, NC, USA
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Nitsan Maharshak
- Department of Gastroenterology and Liver Diseases, Tel Aviv Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel, USA
| | - Ernesto Peréz-Chanona
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, FL, USA
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA
| | - Ian M Carroll
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| | - Janelle C Arthur
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Scott E Plevy
- Immunology Research and Development, Janssen Pharmaceuticals, Spring House, PA, USA
| | - Anthony A Fodor
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Cory R Brouwer
- Bioinformatics Services Division, Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Kannapolis, NC, USA
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - R Balfour Sartor
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
167
|
Abstract
Although gene sequencing has elucidated several mutations associated with mild cytopenias in older individuals, iron deficiency remains the most common cause of anemia. Oral iron has remained the frontline standard despite evidence that it is poorly tolerated, often ineffective, and frequently harmful. Studies of different formulations of intravenous iron have shown it effective, with marginal to no toxicity. Serious adverse events have not been described and the failure to address its administration in iron-deficient elderly patients is an unmet clinical need. This article outlines situations in which oral iron should be proscribed and offers an approach to administration of available formulations.
Collapse
Affiliation(s)
- Michael Auerbach
- Auerbach Hematology and Oncology, 5233 King Avenue #308, Baltimore, MD 21237, USA; Georgetown University School of Medicine, Washington, DC, USA.
| | - Jerry Spivak
- Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
168
|
[Chinese expert consensus on the application of intravenous iron (2019)]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 40:358-362. [PMID: 31207697 PMCID: PMC7342231 DOI: 10.3760/cma.j.issn.0253-2727.2019.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Indexed: 12/03/2022]
|
169
|
DeLoughery TG. Safety of Oral and Intravenous Iron. Acta Haematol 2019; 142:8-12. [PMID: 30970354 DOI: 10.1159/000496966] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Abstract
As the adverse effects of iron deficiency are better recognized, the use of oral and intravenous iron has increased dramatically. Oral iron is often poorly tolerated, with up to 70% or more of patients noting gastrointestinal issues; this may affect adherence to therapy. In addition, many patients will not respond to oral iron due to their underlying illness. Intravenous iron is being used more frequently to replete iron stores. True anaphylaxis is very rare, but complement-mediated infusion reactions may be seen in up to 1 in every 200 patients. Previous concerns about intravenous iron increasing the risk of infection or cardiovascular disease are unfounded.
Collapse
Affiliation(s)
- Thomas G DeLoughery
- Division of Hematology/Medical Oncology, Department of Pathology and Pediatrics, Oregon Health and Science University, Portland, Oregon, USA,
| |
Collapse
|
170
|
Jimenez KM, Gasche C. Management of Iron Deficiency Anaemia in Inflammatory Bowel Disease. Acta Haematol 2019; 142:30-36. [PMID: 30970351 DOI: 10.1159/000496728] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 12/21/2022]
Abstract
Inflammatory bowel disease (IBD) is a group of chronic relapsing inflammatory disorders affecting the large and small intestine, with a rising worldwide incidence and prevalence. Anaemia is the most common extraintestinal manifestation of IBD, correlating with disease activity, and tending to relapse even after successful therapy. Iron deficiency is the most common cause; however, it often manifests in combination with anaemia of inflammation. As such, multiple parameters are used for the diagnosis of iron deficiency anaemia in IBD. Timely recognition and selection of appropriate therapy leads to an improvement in the quality of life and prevention of potential sequelae. Oral iron can be effective under specific circumstances; however, as luminal iron changes microbiota and bacterial metabolism, oral administration should be avoided. Intravenous iron is preferred as it bypasses the sites of inflammation. Nevertheless, the optimization of IBD treatment should occur simultaneously, as this improves both patient condition and response to iron therapy. Herein, we discuss the screening, diagnosis, selection of therapy, and follow-up for iron deficiency anaemia in IBD.
Collapse
Affiliation(s)
- Kristine Michelle Jimenez
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Christoph Gasche
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria,
| |
Collapse
|
171
|
Gut Microbiota-Mediated NLRP12 Expression Drives the Attenuation of Dextran Sulphate Sodium-Induced Ulcerative Colitis by Qingchang Wenzhong Decoction. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9839474. [PMID: 31061672 PMCID: PMC6466890 DOI: 10.1155/2019/9839474] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/13/2018] [Accepted: 03/04/2019] [Indexed: 02/06/2023]
Abstract
Qingchang Wenzhong Decoction (QCWZD) is a newly developed, effective traditional Chinese herbal formulation for ulcerative colitis (UC). In earlier studies, we found that QCWZD could relieve the clinical symptoms of UC patients, reduce inflammation, and improve the intestinal barrier function in dextran sulphate sodium (DSS)-induced UC rats. However, the relationship between QCWZD and the gut microbiota in colitis was not clarified. In this study, we established a rat model of DSS-induced UC and then investigated the regulatory effects of QCWZD on the gut microbiota using 16S rRNA analysis. We also determined the expression of NLRP12 after QCWZD administration. Our findings suggested that QCWZD administration could modulate gut microbiota composition and selectively promote the protective strains such as Butyricimonas, Blautia, and Odoribacter, whereas the enteric pathogens including Clostridium and Dorea were significantly reduced after QCWZD treatment. It is noteworthy that QCWZD administration was identified to promote gut microbiota-mediated NLRP12 expression by inhibiting the activity of the TLR4/Blimp-1 axis. In conclusion, our study supports the potential of QCWZD administration as a beneficial therapeutic strategy for UC.
Collapse
|
172
|
Elli L, Norsa L, Zullo A, Carroccio A, Girelli C, Oliva S, Romano C, Leandro G, Bellini M, Marmo R, Soncini M, Monica F, De Francesco V, Paulon E, Cappellini MD, Motta I, Ferretti F, Orlando S, Mansueto P, Buscarini E, Manfredi G, Agostoni C, Tomba C, Cannizzaro R. Diagnosis of chronic anaemia in gastrointestinal disorders: A guideline by the Italian Association of Hospital Gastroenterologists and Endoscopists (AIGO) and the Italian Society of Paediatric Gastroenterology Hepatology and Nutrition (SIGENP). Dig Liver Dis 2019; 51:471-483. [PMID: 30850345 DOI: 10.1016/j.dld.2019.01.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022]
Abstract
Anaemia is a common pathologic condition, present in almost 5% of the adult population. Iron deficiency is the most common cause; other mechanisms can be involved, making anaemia a multi-factorial disorder in most cases. Anaemia being a frequent manifestation in the diseases of the gastrointestinal tract, patients are often referred to gastroenterologists. Furthermore, upper and lower endoscopy and enteroscopy are pivotal to the diagnostic roadmap of anaemia. In spite of its relevance in the daily clinical practice, there is a limited number of gastroenterological guidelines dedicated to the diagnosis of anaemia. For this reason, the Italian Association of Hospital Gastroenterologists and Endoscopists and the Italian Society of Paediatric Gastroenterology, Hepatology and Nutrition commissioned a panel of experts to prepare a specific guideline on anaemia and its diagnostic roadmap in the gastroenterological scenario. The panel also discussed about the potential involvement of gastroenterologists and endoscopists in the management of patients with anaemia, with particular attention to the correct use of investigations. The panel paid particular attention to practical issues with the aim to support gastroenterologists in their clinical practice when dealing with patients with anaemia.
Collapse
Affiliation(s)
- Luca Elli
- Gastroenterology and Endoscopy Division/Center for Prevention and Diagnosis of Coeliac Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano.
| | - Lorenzo Norsa
- Division of Paediatric Gastroenterology, Hepatology and Transplantation, ASST "Pope Giovanni XXIII", Bergamo
| | - Angelo Zullo
- Gastroenterology and Digestive Endoscopy, "Nuovo Regina Margherita" Hospital, Rome
| | - Antonio Carroccio
- Internal Medicine, "Giovanni Paolo II" Hospital, Sciacca; Biomedical Department of Internal and Specialist Medicine - DiBiMIS, University of Palermo, Palermo
| | - Carlo Girelli
- Gastroenterology and Digestive Endoscopy Unit, Hospital of Busto Arsizio, Busto Arsizio
| | - Salvatore Oliva
- Paediatric Gastroenterology and Liver Unit, University "La Sapienza" of Rome, Roma
| | - Claudio Romano
- Paediatric Gastroenterology and Cystic Fibrosis Unit, Department of Human Pathology in Adulthood and Childhood "G. Barresi", University of Messina, Messina
| | - Gioacchino Leandro
- National Institute of Gastroenterology "S. De Bellis" Research Hospital, Castellana Grotte, Italy
| | - Massimo Bellini
- Gastrointestinal Unit, Department of Translational Research and New Technology in Medicine and Surgery, University of Pisa, Pisa
| | | | - Marco Soncini
- Internal Medicine, "Alessandro Manzoni" Hospital, ASST-Lecco, Lecco
| | - Fabio Monica
- Gastroenterology and Digestive Endoscopy Unit, Cattinara Hospital, Trieste
| | | | - Emma Paulon
- Gastroenterology and Digestive Endoscopy Unit, Cattinara Hospital, Trieste
| | - Maria Domenica Cappellini
- Rare Diseases Center, Department of Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano; Department of Clinical Sciences and Community Health, University of Milan, Milano
| | - Irene Motta
- Rare Diseases Center, Department of Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano; Department of Clinical Sciences and Community Health, University of Milan, Milano
| | - Francesca Ferretti
- Gastroenterology and Endoscopy Division/Center for Prevention and Diagnosis of Coeliac Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano
| | - Stefania Orlando
- Gastroenterology and Endoscopy Division/Center for Prevention and Diagnosis of Coeliac Disease, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano
| | - Pasquale Mansueto
- Biomedical Department of Internal and Specialist Medicine - DiBiMIS, University of Palermo, Palermo
| | | | - Guido Manfredi
- Gastroenterology Unit, ASST Ospedale Maggiore di Crema, Crema
| | - Carlo Agostoni
- Paediatric Intermediate Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano; Department of Clinical Sciences and Community Health, University of Milan, Milano
| | | | - Renato Cannizzaro
- Oncological Gastroenterology Division, Centro di Riferimento Oncologico (CRO) IRCCS, Aviano
| |
Collapse
|
173
|
Horniblow RD, Mistry P, Quraishi MN, Beggs AD, Van de Wiele T, Iqbal TH, Tselepis C. The Safety and Tolerability of a Potential Alginate-Based Iron Chelator; Results of A Healthy Participant Study. Nutrients 2019; 11:E674. [PMID: 30901846 PMCID: PMC6471009 DOI: 10.3390/nu11030674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/16/2022] Open
Abstract
Evidence supporting the ferro-toxic nature of iron in the progression of inflammatory bowel disease (IBD) is becoming well established. A microbial dysbiosis is observed in IBD patients, and intra-luminal colonic-iron is able to support a more pathogenic community of bacteria; whether this is attributed to the development of IBD and how iron could be mediating these microbial changes is still unknown. Dietary fibres are commonly used in pre-biotic supplements to beneficially affect the host by improving the viability of bacterial communities within the colon. Alginates are a class of biopolymers considered as prebiotics due to their fibre-like composition and are able to bind metal cations, in particular, iron. Considering that iron excess is able to negatively alter the microbiome, the use of alginate as a food supplement could be useful in colonic-iron chelation. As such, this first-in-man study aimed to assess whether the use of alginate as a dietary iron chelator was both safe and well tolerated. In addition, the impact of alginate on the microbiome and iron levels was assessed by using an intestinal model SHIME (Simulation of the Human Intestinal Microbial Ecosystem). Alginate was supplemented into the diets (3 g/day) of healthy volunteers (n = 17) for 28 days. Results from this study suggest that daily ingestion of 3 g alginate was well tolerated with very minor side effects. There were no detrimental changes in a variety of haematological parameters or the intestinal microbiome. The bacterial communities within the SHIME model were also not influenced by iron and or alginate; it is possible that alginate may be susceptible to bacterial or enzymatic degradation within the gastro-intestinal tract.
Collapse
Affiliation(s)
- Richard D Horniblow
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Pritesh Mistry
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
| | - Mohammed N Quraishi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- The University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham B15 2TT, UK.
| | - Andrew D Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
| | - Tom Van de Wiele
- CMET, Center for Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Gent, Belgium.
| | - Tariq H Iqbal
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
- Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham B15 2TH, UK.
- The University of Birmingham Microbiome Treatment Centre, University of Birmingham, Birmingham B15 2TT, UK.
| | - Chris Tselepis
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
174
|
Álvarez-Mercado AI, Navarro-Oliveros M, Robles-Sánchez C, Plaza-Díaz J, Sáez-Lara MJ, Muñoz-Quezada S, Fontana L, Abadía-Molina F. Microbial Population Changes and Their Relationship with Human Health and Disease. Microorganisms 2019; 7:E68. [PMID: 30832423 PMCID: PMC6463060 DOI: 10.3390/microorganisms7030068] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022] Open
Abstract
Specific microbial profiles and changes in intestinal microbiota have been widely demonstrated to be associated with the pathogenesis of a number of extra-intestinal (obesity and metabolic syndrome) and intestinal (inflammatory bowel disease) diseases as well as other metabolic disorders, such as non-alcoholic fatty liver disease and type 2 diabetes. Thus, maintaining a healthy gut ecosystem could aid in avoiding the early onset and development of these diseases. Furthermore, it is mandatory to evaluate the alterations in the microbiota associated with pathophysiological conditions and how to counteract them to restore intestinal homeostasis. This review highlights and critically discusses recent literature focused on identifying changes in and developing gut microbiota-targeted interventions (probiotics, prebiotics, diet, and fecal microbiota transplantation, among others) for the above-mentioned pathologies. We also discuss future directions and promising approaches to counteract unhealthy alterations in the gut microbiota. Altogether, we conclude that research in this field is currently in its infancy, which may be due to the large number of factors that can elicit such alterations, the variety of related pathologies, and the heterogeneity of the population involved. Further research on the effects of probiotics, prebiotics, or fecal transplantations on the composition of the human gut microbiome is necessary.
Collapse
Affiliation(s)
- Ana Isabel Álvarez-Mercado
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
| | - Miguel Navarro-Oliveros
- Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
| | - Cándido Robles-Sánchez
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
| | - Julio Plaza-Díaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
| | - María José Sáez-Lara
- Department of Biochemistry and Molecular Biology I, School of Sciences, University of Granada, 18071 Granada, Spain.
| | - Sergio Muñoz-Quezada
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 6094411, Chile.
- National Agency for Medicines (ANAMED), Public Health Institute, Santiago 7780050, Chile.
| | - Luis Fontana
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain.
- Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain.
| | - Francisco Abadía-Molina
- Institute of Nutrition and Food Technology "José Mataix," Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. 18016 Armilla, Granada, Spain.
- Department of Cell Biology, School of Sciences, University of Granada, 18071 Granada, Spain.
| |
Collapse
|
175
|
Venturieri MO, Komati JTS, Lopes LHC, Sdepanian VL. Treatment with Noripurum EV ® is effective and safe in pediatric patients with inflammatory bowel disease and iron deficiency anemia. Scand J Gastroenterol 2019; 54:198-204. [PMID: 30782036 DOI: 10.1080/00365521.2019.1570326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES To evaluate the therapeutic response and adverse effects of Noripurum EV® in children and adolescents with inflammatory bowel disease (IBD) and iron deficiency anemia. MATERIALS AND METHODS Cohort study involving patients with Crohn's disease (CD) and ulcerative colitis (UC) who received treatment for iron deficiency anemia with Noripurum EV®. Anemia was defined according to WHO 2011 criteria. Iron deficiency anemia was established when ferritin <30µg/l and transferrin saturation <16%. Iron deficiency anemia and anemia of chronic disease were established when ferritin was between 30 and 100µg/l and transferrin saturation <16%. The total dose of Noripurum EV® was estimated by the Ganzoni formula and divided into weekly administrations. When there was an increase in hemoglobin (Hb) by a minimum of 2g/dl and or when Hb reached the target determined by WHO, treatment was considered a therapeutic success. RESULTS Noripurum EV® was administered to 16 patients (9.3% of total patients with IBD). Ten (65.5%) were male, the mean (SD) age was 11.3(4.6) years old, 75%(12/16) had CD and 25%(4/16) had UC. All patients presented an increase in Hb (p < .001) at a mean (SD) of 2.8(1.3)g/dl, after median and interquartile range(IQR) of 4.5(3.0-6.0) weeks that iron infusions were completed. It was found that the proportion of patients that achieved therapeutic success (68.8%) was statistically higher (p = .031) than those who did not (31.2%). No adverse events were reported. CONCLUSION Noripurum EV® in pediatric patients with IBD and iron deficiency anemia was effective and safe, making it an appropriate option for the clinical management of these patients.
Collapse
Affiliation(s)
- Maissara O Venturieri
- a Pediatric Gastroenterology, Pediatric Department , Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brasil
| | - Juliana T S Komati
- a Pediatric Gastroenterology, Pediatric Department , Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brasil
| | - Letícia H C Lopes
- a Pediatric Gastroenterology, Pediatric Department , Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brasil
| | - Vera L Sdepanian
- a Pediatric Gastroenterology, Pediatric Department , Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brasil
| |
Collapse
|
176
|
Parmanand BA, Kellingray L, Le Gall G, Basit AW, Fairweather-Tait S, Narbad A. A decrease in iron availability to human gut microbiome reduces the growth of potentially pathogenic gut bacteria; an in vitro colonic fermentation study. J Nutr Biochem 2019; 67:20-27. [PMID: 30831460 PMCID: PMC6546957 DOI: 10.1016/j.jnutbio.2019.01.010] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 02/06/2023]
Abstract
Iron supplements are widely consumed; however most of the iron is not absorbed and enters the colon where potentially pathogenic bacteria can utilise it for growth. This study investigated the effect of iron availability on human gut microbial composition and function using an in vitro colonic fermentation model inoculated with faecal microbiota from healthy adult donors, as well as examining the effect of iron on the growth of individual gut bacteria. Batch fermenters were seeded with fresh faecal material and supplemented with the iron chelator, bathophenanthroline disulphonic acid (BPDS). Samples were analysed at regular intervals to assess impact on the gut bacterial communities. The growth of Escherichia coli and Salmonella typhimurium was significantly impaired when cultured independently in iron-deficient media. In contrast, depletion of iron did not affect the growth of the beneficial species, Lactobacillus rhamnosus, when cultured independently. Analysis of the microbiome composition via 16S-based metataxonomics indicated that under conditions of iron chelation, the relative abundance decreased for several taxa, including a 10% decrease in Escherichia and a 15% decrease in Bifidobacterium. Metabolomics analysis using 1 H-NMR indicated that the production of SCFAs was reduced under iron-limited conditions. These results support previous studies demonstrating the essentiality of iron for microbial growth and metabolism, but, in addition, they indicate that iron chelation changes the gut microbiota profile and influences human gut microbial homeostasis through both compositional and functional changes.
Collapse
Affiliation(s)
- Bhavika A Parmanand
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, NR4 7UA, UK; Faculty of Medicine and Health, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Lee Kellingray
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, NR4 7UA, UK
| | - Gwenaelle Le Gall
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, NR4 7UA, UK
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, UK; Intract Pharma, 29-39 Brunswick Square, London, WC1N 1AX, UK
| | | | - Arjan Narbad
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich, NR4 7UA, UK
| |
Collapse
|
177
|
Buret AG, Motta JP, Allain T, Ferraz J, Wallace JL. Pathobiont release from dysbiotic gut microbiota biofilms in intestinal inflammatory diseases: a role for iron? J Biomed Sci 2019; 26:1. [PMID: 30602371 PMCID: PMC6317250 DOI: 10.1186/s12929-018-0495-4] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 12/21/2018] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota interacting with an intact mucosal surface are key to the maintenance of homeostasis and health. This review discusses the current state of knowledge of the biofilm mode of growth of these microbiota communities, and how in turn their disruptions may cause disease. Beyond alterations of relative microbial abundance and diversity, the aim of the review is to focus on the disruptions of the microbiota biofilm structure and function, the dispersion of commensal bacteria, and the mechanisms whereby these dispersed commensals may become pathobionts. Recent findings have linked iron acquisition to the expression of virulence factors in gut commensals that have become pathobionts. Causal studies are emerging, and mechanisms common to enteropathogen-induced disruptions, as well as those reported for Inflammatory Bowel Disease and colo-rectal cancer are used as examples to illustrate the great translational potential of such research. These new observations shed new light on our attempts to develop new therapies that are able to protect and restore gut microbiota homeostasis in the many disease conditions that have been linked to microbiota dysbiosis.
Collapse
Affiliation(s)
- Andre Gerald Buret
- Departments of Biological Sciences, and Pharmacology and Therapeutics, Inflammation Research Network, University of Calgary, 2500 University Dr. N.W, Calgary, T2N 1N4, Canada.
| | - Jean-Paul Motta
- Departments of Biological Sciences, and Pharmacology and Therapeutics, Inflammation Research Network, University of Calgary, 2500 University Dr. N.W, Calgary, T2N 1N4, Canada.,Institute of Digestive Health Research, INSERM UMR1220, Université Toulouse Paul Sabatier, Toulouse, France
| | - Thibault Allain
- Departments of Biological Sciences, and Pharmacology and Therapeutics, Inflammation Research Network, University of Calgary, 2500 University Dr. N.W, Calgary, T2N 1N4, Canada
| | - Jose Ferraz
- Division of Gastroenterology, Cumming School of Medicine, University of Calgary, Calgary, T2N 1N4, Canada
| | - John Lawrence Wallace
- Departments of Biological Sciences, and Pharmacology and Therapeutics, Inflammation Research Network, University of Calgary, 2500 University Dr. N.W, Calgary, T2N 1N4, Canada
| |
Collapse
|
178
|
Abstract
Iron deficiency anemia affects >1.2 billions individuals worldwide, and iron deficiency in the absence of anemia is even more frequent. Total-body (absolute) iron deficiency is caused by physiologically increased iron requirements in children, adolescents, young and pregnant women, by reduced iron intake, or by pathological defective absorption or chronic blood loss. Adaptation to iron deficiency at the tissue level is controlled by iron regulatory proteins to increase iron uptake and retention; at the systemic level, suppression of the iron hormone hepcidin increases iron release to plasma by absorptive enterocytes and recycling macrophages. The diagnosis of absolute iron deficiency is easy unless the condition is masked by inflammatory conditions. All cases of iron deficiency should be assessed for treatment and underlying cause. Special attention is needed in areas endemic for malaria and other infections to avoid worsening of infection by iron treatment. Ongoing efforts aim at optimizing iron salts-based therapy by protocols of administration based on the physiology of hepcidin control and reducing the common adverse effects of oral iron. IV iron, especially last-generation compounds administered at high doses in single infusions, is becoming an effective alternative in an increasing number of conditions because of a more rapid and persistent hematological response and acceptable safety profile. Risks/benefits of the different treatments should be weighed in a personalized therapeutic approach to iron deficiency.
Collapse
Affiliation(s)
- Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
179
|
Carman N, Muir R, Lewindon P. Ferric carboxymaltose in the treatment of iron deficiency in pediatric inflammatory bowel disease. Transl Pediatr 2019; 8:28-34. [PMID: 30881896 PMCID: PMC6382504 DOI: 10.21037/tp.2019.01.01] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Iron deficiency (ID) with or without anemia is a common complication of pediatric inflammatory bowel disease (IBD), causing significant morbidity. Despite this, ID remains prevalent and undertreated, related in part to questions surrounding optimal formulation and route of administration. Ferric carboxymaltose (FCM) is a recent formulation of intravenous iron, allowing higher doses and rapid infusion times. This study aims to demonstrate the efficacy and safety of FCM in paediatric patients with IBD, and explore the differences between patients with active and quiescent disease. METHODS Paediatric patients 6-18 years with IBD with iron deficiency (ID) or iron deficiency anemia (IDA) were treated prospectively with FCM at the Queensland Children's Hospital in Brisbane. Patients received FCM as a single dose of 15 mg/kg up to 1,000 mg over 15-20 min. Biochemical parameters measured prior to and approximately 8 weeks after the infusion were: hemoglobin (Hb), mean corpuscular volume (MCV), ferritin, and transferrin saturation (TS). C-reactive protein (CRP) was measured as a marker of co-existing inflammation. Resolution of anemia or ID was assessed following treatment, with adverse events captured. RESULTS A total of 101 patients received infusions of FCM during the study period and were analysed, median age 14 (IQR 14-16) years. A total of 44% of patients underwent treatment for IDA, while 56% were for ID without anemia. Following FCM infusion, 64% of patients with IDA had resolution of anemia, with 81% showing resolution for ID without anemia. Elevation of CRP throughout the study period had no influence on resolution of IDA with FCM (P=0.68), but in patients with ID, patients with quiescent disease activity were more likely to have resolution of ID [odds ratios (ORs) 5.1; P=0.03]. CONCLUSIONS Rapid, high dose FCM in children aged 6 and over is safe, well tolerated and efficacious for correction of ID. Replenishing iron in IBD is important and FCM improves our ability to meet this need.
Collapse
Affiliation(s)
- Nicholas Carman
- Department of Gastroenterology, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Canada
| | - Richard Muir
- Department of Gastroenterology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Peter Lewindon
- Department of Gastroenterology, Queensland Children's Hospital, South Brisbane, Queensland, Australia.,Queensland Children's Medical Research Institute, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
180
|
Petzer V, Theurl I, Weiss G. Established and Emerging Concepts to Treat Imbalances of Iron Homeostasis in Inflammatory Diseases. Pharmaceuticals (Basel) 2018; 11:E135. [PMID: 30544952 PMCID: PMC6315795 DOI: 10.3390/ph11040135] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/05/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
Inflammation, being a hallmark of many chronic diseases, including cancer, inflammatory bowel disease, rheumatoid arthritis, and chronic kidney disease, negatively affects iron homeostasis, leading to iron retention in macrophages of the mononuclear phagocyte system. Functional iron deficiency is the consequence, leading to anemia of inflammation (AI). Iron deficiency, regardless of anemia, has a detrimental impact on quality of life so that treatment is warranted. Therapeutic strategies include (1) resolution of the underlying disease, (2) iron supplementation, and (3) iron redistribution strategies. Deeper insights into the pathophysiology of AI has led to the development of new therapeutics targeting inflammatory cytokines and the introduction of new iron formulations. Moreover, the discovery that the hormone, hepcidin, plays a key regulatory role in AI has stimulated the development of several therapeutic approaches targeting the function of this peptide. Hence, inflammation-driven hepcidin elevation causes iron retention in cells and tissues. Besides pathophysiological concepts and diagnostic approaches for AI, this review discusses current guidelines for iron replacement therapies with special emphasis on benefits, limitations, and unresolved questions concerning oral versus parenteral iron supplementation in chronic inflammatory diseases. Furthermore, the review explores how therapies aiming at curing the disease underlying AI can also affect anemia and discusses emerging hepcidin antagonizing drugs, which are currently under preclinical or clinical investigation.
Collapse
Affiliation(s)
- Verena Petzer
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Igor Theurl
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University of Innsbruck, 6020 Innsbruck, Austria.
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, Medical University of Innsbruck, 6020 Innsbruck, Austria.
| |
Collapse
|
181
|
Iron Treatment May Be Difficult in Inflammatory Diseases: Inflammatory Bowel Disease as a Paradigm. Nutrients 2018; 10:nu10121959. [PMID: 30544934 PMCID: PMC6316243 DOI: 10.3390/nu10121959] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022] Open
Abstract
Iron plays a key role in many physiological processes; cells need a very exact quantity of iron. In patients with inflammatory bowel disease, anaemia is a unique example of multifactorial origins, frequently being the result of a combination of iron deficiency and anaemia of chronic disease. The main cause of iron deficiency is the activity of the disease. Therefore, the first aim should be to reach complete clinical remission. The iron supplementation route should be determined according to symptoms, severity of anaemia and taking into account comorbidities and individual risks. Oral iron can only be used in patients with mild anaemia, whose disease is inactive and who have not been previously intolerant to oral iron. Intravenous iron should be the first line treatment in patients with moderate-severe anaemia, in patients with active disease, in patients with poor tolerance to oral iron and when erythropoietin agents or a fast response is needed. Erythropoietin is used in a few patients with anaemia to overcome functional iron deficiency, and blood transfusion is being restricted to refractory cases or acute life-threatening situations.
Collapse
|
182
|
Karu N, Deng L, Slae M, Guo AC, Sajed T, Huynh H, Wine E, Wishart DS. A review on human fecal metabolomics: Methods, applications and the human fecal metabolome database. Anal Chim Acta 2018; 1030:1-24. [DOI: 10.1016/j.aca.2018.05.031] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/05/2018] [Accepted: 05/09/2018] [Indexed: 12/19/2022]
|
183
|
Mahalhal A, Williams JM, Johnson S, Ellaby N, Duckworth CA, Burkitt MD, Liu X, Hold GL, Campbell BJ, Pritchard DM, Probert CS. Oral iron exacerbates colitis and influences the intestinal microbiome. PLoS One 2018; 13:e0202460. [PMID: 30308045 PMCID: PMC6181268 DOI: 10.1371/journal.pone.0202460] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/27/2018] [Indexed: 12/21/2022] Open
Abstract
Inflammatory bowel disease (IBD) is associated with anaemia and oral iron replacement to correct this can be problematic, intensifying inflammation and tissue damage. The intestinal microbiota also plays a key role in the pathogenesis of IBD, and iron supplementation likely influences gut bacterial diversity in patients with IBD. Here, we assessed the impact of dietary iron, using chow diets containing either 100, 200 or 400 ppm, fed ad libitum to adult female C57BL/6 mice in the presence or absence of colitis induced using dextran sulfate sodium (DSS), on (i) clinical and histological severity of acute DSS-induced colitis, and (ii) faecal microbial diversity, as assessed by sequencing the V4 region of 16S rRNA. Increasing or decreasing dietary iron concentration from the standard 200 ppm exacerbated both clinical and histological severity of DSS-induced colitis. DSS-treated mice provided only half the standard levels of iron ad libitum (i.e. chow containing 100 ppm iron) lost more body weight than those receiving double the amount of standard iron (i.e. 400 ppm); p<0.01. Faecal calprotectin levels were significantly increased in the presence of colitis in those consuming 100 ppm iron at day 8 (5.94-fold) versus day-10 group (4.14-fold) (p<0.05), and for the 400 ppm day-8 group (8.17-fold) versus day-10 group (4.44-fold) (p<0.001). In the presence of colitis, dietary iron at 400 ppm resulted in a significant reduction in faecal abundance of Firmicutes and Bacteroidetes, and increase of Proteobacteria, changes which were not observed with lower dietary intake of iron at 100 ppm. Overall, altering dietary iron intake exacerbated DSS-induced colitis; increasing the iron content of the diet also led to changes in intestinal bacteria diversity and composition after colitis was induced with DSS.
Collapse
Affiliation(s)
- Awad Mahalhal
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- Department of Anatomy and Histology, Faculty of Medicine, Benghazi University, Benghazi, Libya
- * E-mail:
| | - Jonathan M. Williams
- Pathobiology and Population Sciences, Royal Veterinary College, North Mymms, United Kingdom
| | - Sophie Johnson
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Nicholas Ellaby
- Department of Functional & Comparative Genomics, Institute of Integrative Biology, Liverpool University, Liverpool, United Kingdom
| | - Carrie A. Duckworth
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Michael D. Burkitt
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Xuan Liu
- Department of Functional & Comparative Genomics, Institute of Integrative Biology, Liverpool University, Liverpool, United Kingdom
| | - Georgina L. Hold
- Department of Medicine, St George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Barry J. Campbell
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - D. Mark Pritchard
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Chris S. Probert
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
184
|
Gut Microbiota and Iron: The Crucial Actors in Health and Disease. Pharmaceuticals (Basel) 2018; 11:ph11040098. [PMID: 30301142 PMCID: PMC6315993 DOI: 10.3390/ph11040098] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 09/30/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023] Open
Abstract
Iron (Fe) is a highly ample metal on planet earth (~35% of the Earth’s mass) and is particularly essential for most life forms, including from bacteria to mammals. Nonetheless, iron deficiency is highly prevalent in developing countries, and oral administration of this metal is so far the most effective treatment for human beings. Notably, the excessive amount of unabsorbed iron leave unappreciated side effects at the highly interactive host–microbe interface of the human gastrointestinal tract. Recent advances in elucidating the molecular basis of interactions between iron and gut microbiota shed new light(s) on the health and pathogenesis of intestinal inflammatory diseases. We here aim to present the dynamic modulation of intestinal microbiota by iron availability, and conversely, the influence on dietary iron absorption in the gut. The central part of this review is intended to summarize our current understanding about the effects of luminal iron on host–microbe interactions in the context of human health and disease.
Collapse
|
185
|
Gómez-Ramírez S, Brilli E, Tarantino G, Muñoz M. Sucrosomial ® Iron: A New Generation Iron for Improving Oral Supplementation. Pharmaceuticals (Basel) 2018; 11:E97. [PMID: 30287781 PMCID: PMC6316120 DOI: 10.3390/ph11040097] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 09/28/2018] [Accepted: 10/02/2018] [Indexed: 01/28/2023] Open
Abstract
Iron deficiency (ID) is usually treated with oral iron salts, but up to 50% of patients complain of gastrointestinal side effects, leading to reduced treatment compliance. Intravenous (IV) iron formulations are increasingly safer, but there is still a risk of infusion and hypersensitivity reactions and the need for a venous access and infusion monitoring. Sucrosomial® iron (SI) is an innovative oral iron formulation in which ferric pyrophosphate is protected by a phospholipid bilayer plus a sucrester matrix (sucrosome), which is absorbed through para-cellular and trans-cellular routes (M cells). This confers SI unique structural, physicochemical and pharmacokinetic characteristics, together with high iron bioavailability and excellent gastrointestinal tolerance. The analysis of available evidence supports oral SI iron as a valid option for ID treatment, which is more efficacious and better tolerated than oral iron salts. SI has also demonstrated similar effectiveness, with lower risks, in patients usually receiving IV iron (e.g., chronic kidney disease, cancer, bariatric surgery). Thus, oral SI emerges as a most valuable first option for treating ID, even more for subjects with intolerance to or inefficacy of iron salts. Moreover, SI should be also considered as an alternative to IV iron for initial and/or maintenance treatment in different patient populations.
Collapse
Affiliation(s)
- Susana Gómez-Ramírez
- Department of Internal Medicine, University Hospital Virgen de la Victoria. Campus de Teatinos, 2010 Málaga, Spain.
| | - Elisa Brilli
- Scientific Department, Alesco S.r.l. Via delle Lenze, 216/B, 56122 Pisa, Italy.
| | - Germano Tarantino
- Scientific Department, Pharmanutra S.p.A. Via delle Lenze, 216/B, 56122 Pisa, Italy.
| | - Manuel Muñoz
- Perioperative Transfusion Medicine, Department of Surgical Specialties, Biochemistry and Immunology, School of Medicine, Campus de Teatinos, 29071 Málaga, Spain.
| |
Collapse
|
186
|
Harris KG, Chang EB. The intestinal microbiota in the pathogenesis of inflammatory bowel diseases: new insights into complex disease. Clin Sci (Lond) 2018; 132:2013-2028. [PMID: 30232239 PMCID: PMC6907688 DOI: 10.1042/cs20171110] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic diseases of increasing worldwide prevalence characterized by gastrointestinal (GI) inflammation leading to debilitating symptoms and complications. The contribution of the intestinal microbiota to the pathogenesis and etiology of these diseases is an area of active research interest. Here, we discuss key mechanisms underlying the chronic inflammation seen in IBD as well as evidence implicating the intestinal microbiota in the development and potentiation of that inflammation. We also discuss recently published work in areas of interest within the field of microbial involvement in IBD pathogenesis - the importance of proper microecology within the GI tract, the evidence that the intestinal microbiota transduces environmental and genetic risk factors for IBD, and the mechanisms by which microbial products contribute to communication between microbe and host. There is an extensive body of published research on the evidence for microbial involvement in IBD; the goal of this review is to highlight the growing edges of the field where exciting and innovative research is pushing the boundaries of the conceptual framework of the role of the intestinal microbiota in IBD pathogenesis.
Collapse
Affiliation(s)
| | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, U.S.A.
| |
Collapse
|
187
|
Zhang H, Zhabyeyev P, Wang S, Oudit GY. Role of iron metabolism in heart failure: From iron deficiency to iron overload. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1925-1937. [PMID: 31109456 DOI: 10.1016/j.bbadis.2018.08.030] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/25/2018] [Accepted: 08/22/2018] [Indexed: 12/11/2022]
Abstract
Iron metabolism is a balancing act, and biological systems have evolved exquisite regulatory mechanisms to maintain iron homeostasis. Iron metabolism disorders are widespread health problems on a global scale and range from iron deficiency to iron-overload. Both types of iron disorders are linked to heart failure. Iron play a fundamental role in mitochondrial function and various enzyme functions and iron deficiency has a particular negative impact on mitochondria function. Given the high-energy demand of the heart, iron deficiency has a particularly negative impact on heart function and exacerbates heart failure. Iron-overload can result from excessive gut absorption of iron or frequent use of blood transfusions and is typically seen in patients with congenital anemias, sickle cell anemia and beta-thalassemia major, or in patients with primary hemochromatosis. This review provides an overview of normal iron metabolism, mechanisms underlying development of iron disorders in relation to heart failure, including iron-overload cardiomyopathy, and clinical perspective on the treatment options for iron metabolism disorders.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada
| | - Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada
| | - Shaohua Wang
- Mazankowski Alberta Heart Institute, Canada; Division of Cardiac Surgery, Department of Surgery, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Canada; Mazankowski Alberta Heart Institute, Canada.
| |
Collapse
|
188
|
Motta JP, Allain T, Green-Harrison LE, Groves RA, Feener T, Ramay H, Beck PL, Lewis IA, Wallace JL, Buret AG. Iron Sequestration in Microbiota Biofilms As A Novel Strategy for Treating Inflammatory Bowel Disease. Inflamm Bowel Dis 2018; 24:1493-1502. [PMID: 29788224 PMCID: PMC5995063 DOI: 10.1093/ibd/izy116] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Indexed: 12/12/2022]
Abstract
Significant alterations of intestinal microbiota and anemia are hallmarks of inflammatory bowel disease (IBD). It is widely accepted that iron is a key nutrient for pathogenic bacteria, but little is known about its impact on microbiota associated with IBD. We used a model device to grow human mucosa-associated microbiota in its physiological anaerobic biofilm phenotype. Compared to microbiota from healthy donors, microbiota from IBD patients generate biofilms ex vivo that were larger in size and cell numbers, contained higher intracellular iron concentrations, and exhibited heightened virulence in a model of human intestinal epithelia in vitro and in the nematode Caenorhabditis elegans. We also describe an unexpected iron-scavenging property for an experimental hydrogen sulfide-releasing derivative of mesalamine. The findings demonstrate that this new drug reduces the virulence of IBD microbiota biofilms through a direct reduction of microbial iron intake and without affecting bacteria survival or species composition within the microbiota. Metabolomic analyses indicate that this drug reduces the intake of purine nucleosides (guanosine), increases the secretion of metabolite markers of purine catabolism (urate and hypoxanthine), and reduces the secretion of uracil (a pyrimidine nucleobase) in complex multispecies human biofilms. These findings demonstrate a new pathogenic mechanism for dysbiotic microbiota in IBD and characterize a novel mode of action for a class of mesalamine derivatives. Together, these observations pave the way towards a new therapeutic strategy for treatment of patients with IBD.
Collapse
Affiliation(s)
- Jean-Paul Motta
- Department of Biological Sciences, University of Calgary, University Drive NW, Calgary, Alberta, Canada,Department of Physiology & Pharmacology, University of Calgary, Hospital Drive NW, Calgary, Alberta, Canada
| | - Thibault Allain
- Department of Biological Sciences, University of Calgary, University Drive NW, Calgary, Alberta, Canada
| | - Luke E Green-Harrison
- Department of Biological Sciences, University of Calgary, University Drive NW, Calgary, Alberta, Canada
| | - Ryan A Groves
- Department of Biological Sciences, University of Calgary, University Drive NW, Calgary, Alberta, Canada
| | - Troy Feener
- Department of Physiology & Pharmacology, University of Calgary, Hospital Drive NW, Calgary, Alberta, Canada
| | - Hena Ramay
- International Microbiome Centre, University of Calgary, Hospital Drive NW, Calgary, Alberta, Canada
| | - Paul L Beck
- Department of Medicine, University of Calgary, Hospital Drive NW, Calgary, Alberta, Canada
| | - Ian A Lewis
- Department of Biological Sciences, University of Calgary, University Drive NW, Calgary, Alberta, Canada
| | - John L Wallace
- Department of Physiology & Pharmacology, University of Calgary, Hospital Drive NW, Calgary, Alberta, Canada
| | - Andre G Buret
- Department of Biological Sciences, University of Calgary, University Drive NW, Calgary, Alberta, Canada,Address correspondence to: Andre G. Buret, PhD, Department of Biological Sciences, Faculty of Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 4N1, Canada. E-mail:
| |
Collapse
|
189
|
Camaschella C, Pagani A. Advances in understanding iron metabolism and its crosstalk with erythropoiesis. Br J Haematol 2018; 182:481-494. [PMID: 29938779 DOI: 10.1111/bjh.15403] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recent years have witnessed impressive advances in our understanding of iron metabolism. A number of studies of iron disorders and of their animal models have provided landmark insights into the mechanisms of iron trafficking, distribution and homeostatic regulation, the latter essential to prevent both iron deficiency and iron excess. Our perception of iron metabolism has been completely changed by an improved definition of cellular and systemic iron homeostasis, of the molecular pathogenesis of iron disorders, the fine tuning of the iron hormone hepcidin by activators and inhibitors and the dissection of the components of the hepcidin regulatory pathway. Important for haematology, the crosstalk of erythropoiesis, the most important iron consumer, and the hepcidin pathway has been at least partially clarified. Novel potential biomarkers are available and novel therapeutic targets for iron-related disorders have been tested in murine models. These preclinical studies provided proofs of principle and are laying the ground for clinical trials. Understanding iron control in tissues other than erythropoiesis remains a challenge for the future.
Collapse
Affiliation(s)
- Clara Camaschella
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| | - Alessia Pagani
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Vita Salute University, Milano, Italy
| |
Collapse
|
190
|
Størdal K, McArdle HJ, Hayes H, Tapia G, Viken MK, Lund-Blix NA, Haugen M, Joner G, Skrivarhaug T, Mårild K, Njølstad PR, Eggesbø M, Mandal S, Page CM, London SJ, Lie BA, Stene LC. Prenatal iron exposure and childhood type 1 diabetes. Sci Rep 2018; 8:9067. [PMID: 29899542 PMCID: PMC5998022 DOI: 10.1038/s41598-018-27391-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Iron overload due to environmental or genetic causes have been associated diabetes. We hypothesized that prenatal iron exposure is associated with higher risk of childhood type 1 diabetes. In the Norwegian Mother and Child cohort study (n = 94,209 pregnancies, n = 373 developed type 1 diabetes) the incidence of type 1 diabetes was higher in children exposed to maternal iron supplementation than unexposed (36.8/100,000/year compared to 28.6/100,000/year, adjusted hazard ratio 1.33, 95%CI: 1.06-1.67). Cord plasma biomarkers of high iron status were non-significantly associated with higher risk of type 1 diabetes (ferritin OR = 1.05 [95%CI: 0.99-1.13] per 50 mg/L increase; soluble transferrin receptor: OR = 0.91 [95%CI: 0.81-1.01] per 0.5 mg/L increase). Maternal but not fetal HFE genotypes causing high/intermediate iron stores were associated with offspring diabetes (odds ratio: 1.45, 95%CI: 1.04, 2.02). Maternal anaemia or non-iron dietary supplements did not significantly predict type 1 diabetes. Perinatal iron exposures were not associated with cord blood DNA genome-wide methylation, but fetal HFE genotype was associated with differential fetal methylation near HFE. Maternal cytokines in mid-pregnancy of the pro-inflammatory M1 pathway differed by maternal iron supplements and HFE genotype. Our results suggest that exposure to iron during pregnancy may be a risk factor for type 1 diabetes in the offspring.
Collapse
Affiliation(s)
- Ketil Størdal
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway.
- Pediatric Department, Ostfold Hospital Trust, Fredrikstad, Norway.
| | - Harry J McArdle
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Helen Hayes
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - German Tapia
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Marte K Viken
- Department of Medical Genetics, University of Oslo, Oslo University Hospital, Ullevål, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Nicolai A Lund-Blix
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
- Department of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Margaretha Haugen
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Geir Joner
- Department of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Torild Skrivarhaug
- Department of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Karl Mårild
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål R Njølstad
- Department of Paediatrics and Adolescent Medicine, Haukeland University Hospital, Bergen, Norway
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Merete Eggesbø
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Siddhartha Mandal
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Christian M Page
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Stephanie J London
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, 27709, USA
| | - Benedicte A Lie
- Department of Medical Genetics, University of Oslo, Oslo University Hospital, Ullevål, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Lars C Stene
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
191
|
Iebba V, Guerrieri F, Di Gregorio V, Levrero M, Gagliardi A, Santangelo F, Sobolev AP, Circi S, Giannelli V, Mannina L, Schippa S, Merli M. Combining amplicon sequencing and metabolomics in cirrhotic patients highlights distinctive microbiota features involved in bacterial translocation, systemic inflammation and hepatic encephalopathy. Sci Rep 2018; 8:8210. [PMID: 29844325 PMCID: PMC5974022 DOI: 10.1038/s41598-018-26509-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 05/09/2018] [Indexed: 12/13/2022] Open
Abstract
In liver cirrhosis (LC), impaired intestinal functions lead to dysbiosis and possible bacterial translocation (BT). Bacteria or their byproducts within the bloodstream can thus play a role in systemic inflammation and hepatic encephalopathy (HE). We combined 16S sequencing, NMR metabolomics and network analysis to describe the interrelationships of members of the microbiota in LC biopsies, faeces, peripheral/portal blood and faecal metabolites with clinical parameters. LC faeces and biopsies showed marked dysbiosis with a heightened proportion of Enterobacteriaceae. Our approach showed impaired faecal bacterial metabolism of short-chain fatty acids (SCFAs) and carbon/methane sources in LC, along with an enhanced stress-related response. Sixteen species, mainly belonging to the Proteobacteria phylum, were shared between LC peripheral and portal blood and were functionally linked to iron metabolism. Faecal Enterobacteriaceae and trimethylamine were positively correlated with blood proinflammatory cytokines, while Ruminococcaceae and SCFAs played a protective role. Within the peripheral blood and faeces, certain species (Stenotrophomonas pavanii, Methylobacterium extorquens) and metabolites (methanol, threonine) were positively related to HE. Cirrhotic patients thus harbour a 'functional dysbiosis' in the faeces and peripheral/portal blood, with specific keystone species and metabolites related to clinical markers of systemic inflammation and HE.
Collapse
Affiliation(s)
- Valerio Iebba
- Istituto Pasteur Cenci Bolognetti Foundation, Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Francesca Guerrieri
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Vincenza Di Gregorio
- Gastroenterology, Department of Clinical Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Massimo Levrero
- Center for Life NanoScience@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), Centre Léon Bérard, Lyon, France
| | - Antonella Gagliardi
- Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Floriana Santangelo
- Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Anatoly P Sobolev
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
- Magnetic Resonance Laboratory "Annalaura Segre", Institute of Chemical Methodologies, CNR, via Salaria km 29.300, 00015, Monterotondo, (RM), Italy
| | - Simone Circi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
| | - Valerio Giannelli
- Gastroenterology, Department of Clinical Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy
| | - Luisa Mannina
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Rome, Italy
- Magnetic Resonance Laboratory "Annalaura Segre", Institute of Chemical Methodologies, CNR, via Salaria km 29.300, 00015, Monterotondo, (RM), Italy
| | - Serena Schippa
- Public Health and Infectious Diseases Department, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Manuela Merli
- Gastroenterology, Department of Clinical Medicine, Sapienza University of Rome, Viale dell'Università 37, 00185, Rome, Italy.
| |
Collapse
|
192
|
Iron supplementation has minor effects on gut microbiota composition in overweight and obese women in early pregnancy. Br J Nutr 2018; 120:283-289. [PMID: 29789023 DOI: 10.1017/s0007114518001149] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fe is an essential nutrient for many bacteria, and Fe supplementation has been reported to affect the composition of the gut microbiota in both Fe-deficient and Fe-replete individuals outside pregnancy. This study examined whether the dose of Fe in pregnancy multivitamin supplements affects the overall composition of the gut microbiota in overweight and obese pregnant women in early pregnancy. Women participating in the SPRING study with a faecal sample obtained at 16 weeks' gestation were included in this substudy. For each subject, the brand of multivitamin used was recorded. Faecal microbiome composition was assessed by 16S rRNA sequencing and analysed with the QIIME software suite. Dietary intake of Fe was assessed using a FFQ at 16 weeks' gestation. Women were grouped as receiving low (<60 mg/d, n 94) or high (≥60 mg/d; n 65) Fe supplementation. The median supplementary Fe intake in the low group was 10 (interquartile range (IQR) 5-10) v. 60 (IQR 60-60) mg/d in the high group (P<0·001). Dietary Fe intake did not differ between the groups (10·0 (IQR 7·4-13·3) v. 9·8 (IQR 8·2-13·2) mg/d). Fe supplementation did not significantly affect the composition of the faecal microbiome at any taxonomic level. Network analysis showed that the gut microbiota in the low Fe supplementation group had a higher predominance of SCFA producers. Pregnancy multivitamin Fe content has a minor effect on the overall composition of the gut microbiota of overweight and obese pregnant women at 16 weeks' gestation.
Collapse
|
193
|
Skrypnik K, Suliburska J. Association between the gut microbiota and mineral metabolism. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:2449-2460. [PMID: 28991359 DOI: 10.1002/jsfa.8724] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/23/2017] [Accepted: 09/27/2017] [Indexed: 06/07/2023]
Abstract
The aim of this review is to present the most recent scientific evidence of interactions between the intestinal microbiota and minerals, and the effect of this interaction on the health of the host. The Web of Science database from the years 2013-2017 on this topic was reviewed. Numerous in vitro studies have shown that iron significantly affects the intestinal microbiota. However, Bifidobacteriaceae are capable of binding iron in the large intestine, thereby limiting the formation of free radicals synthesized in the presence of iron, and thus reducing the risk of colorectal cancer. Animal studies have revealed that supplementation with probiotics, prebiotics and synbiotics has a significant effect on bone calcium, phosphate and bone metabolism. The dynamic interaction between microbiota and zinc was shown. Human studies have provided evidence of the influence of probiotic bacteria on parathormone, calcium and phosphate levels and thus on bone resorption. Recent studies have produced new information mainly on the impact of the intestinal bacteria on the metabolism of calcium and iron. From a scientific perspective, the most urgent fields that remain to be investigated are the identification of all human gut microbes and new therapies targeting the interaction between intestinal bacteria and minerals. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Katarzyna Skrypnik
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| | - Joanna Suliburska
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| |
Collapse
|
194
|
McCormack UM, Curião T, Wilkinson T, Metzler-Zebeli BU, Reyer H, Ryan T, Calderon-Diaz JA, Crispie F, Cotter PD, Creevey CJ, Gardiner GE, Lawlor PG. Fecal Microbiota Transplantation in Gestating Sows and Neonatal Offspring Alters Lifetime Intestinal Microbiota and Growth in Offspring. mSystems 2018; 3:e00134-17. [PMID: 29577087 PMCID: PMC5864416 DOI: 10.1128/msystems.00134-17] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/16/2018] [Indexed: 12/29/2022] Open
Abstract
Previous studies suggest a link between intestinal microbiota and porcine feed efficiency (FE). Therefore, we investigated whether fecal microbiota transplantation (FMT) in sows and/or neonatal offspring, using inocula derived from highly feed-efficient pigs, could improve offspring FE. Pregnant sows were assigned to control or FMT treatments and the subsequent offspring to control treatment, FMT once (at birth), or FMT four times (between birth and weaning). FMT altered sow fecal and colostrum microbiota compositions and resulted in lighter offspring body weight at 70 and 155 days of age when administered to sows and/or offspring. This was accompanied by FMT-associated changes within the offspring's intestinal microbiota, mostly in the ileum. These included transiently higher fecal bacterial diversity and load and numerous compositional differences at the phylum and genus levels (e.g., Spirochaetes and Bacteroidetes at high relative abundances and mostly members of Clostridia, respectively), as well as differences in the abundances of predicted bacterial pathways. In addition, intestinal morphology was negatively impacted, duodenal gene expression altered, and serum protein and cholesterol concentrations reduced due to FMT in sows and/or offspring. Taken together, the results suggest poorer absorptive capacity and intestinal health, most likely explaining the reduced body weight. An additive effect of FMT in sows and offspring also occurred for some parameters. Although these findings have negative implications for the practical use of the FMT regime used here for improving FE in pigs, they nonetheless demonstrate the enormous impact of early-life intestinal microbiota on the host phenotype. IMPORTANCE Here, for the first time, we investigate FMT as a novel strategy to modulate the porcine intestinal microbiota in an attempt to improve FE in pigs. However, reprogramming the maternal and/or offspring microbiome by using fecal transplants derived from highly feed-efficient pigs did not recapitulate the highly efficient phenotype in the offspring and, in fact, had detrimental effects on lifetime growth. Although these findings may not be wholly attributable to microbiota transplantation, as antibiotic and purgative were also part of the regime in sows, similar effects were also seen in offspring, in which these interventions were not used. Nonetheless, additional work is needed to unravel the effects of each component of the FMT regime and to provide additional mechanistic insights. This may lead to the development of an FMT procedure with practical applications for the improvement of FE in pigs, which could in turn improve the profitability of pig production.
Collapse
Affiliation(s)
- Ursula M. McCormack
- Teagasc, Pig Development Department, Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, County Cork, Ireland
- Department of Science, Waterford Institute of Technology, Waterford, Ireland
| | - Tânia Curião
- Teagasc, Pig Development Department, Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Toby Wilkinson
- Animal and Microbial Sciences, Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, United Kingdom
| | - Barbara U. Metzler-Zebeli
- Institute of Animal Nutrition and Functional Plant Compounds, University Clinic for Swine, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Henry Reyer
- Leibeniz institute (FBN), Dummerstorf, Germany
| | - Tomas Ryan
- Teagasc, Pig Development Department, Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Julia A. Calderon-Diaz
- Teagasc, Pig Development Department, Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, County Cork, Ireland
- Department of Animal Behaviour and Welfare, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Jastrzębiec, Magdalenka, Poland
| | - Fiona Crispie
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
- APC Microbiome Institute, Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
- APC Microbiome Institute, Cork, Ireland
| | - Christopher J. Creevey
- Animal and Microbial Sciences, Institute of Biological, Environmental and Rural Sciences (IBERS), Aberystwyth University, Aberystwyth, United Kingdom
| | - Gillian E. Gardiner
- Department of Science, Waterford Institute of Technology, Waterford, Ireland
| | - Peadar G. Lawlor
- Teagasc, Pig Development Department, Animal and Grassland Research and Innovation Centre, Moorepark, Fermoy, County Cork, Ireland
| |
Collapse
|
195
|
Niepel D, Klag T, Malek NP, Wehkamp J. Practical guidance for the management of iron deficiency in patients with inflammatory bowel disease. Therap Adv Gastroenterol 2018; 11:1756284818769074. [PMID: 29760784 PMCID: PMC5946590 DOI: 10.1177/1756284818769074] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Accepted: 03/11/2018] [Indexed: 02/04/2023] Open
Abstract
Iron deficiency or iron deficiency anemia (IDA) are some of the most common systemic complications of inflammatory bowel diseases (IBD). Symptoms such as fatigue, reduced ability to concentrate and reduced exercise tolerance can mimic common symptoms of IBD and can therefore easily be overseen. Furthermore, clinicians tend to see mild to moderate anemia as an inevitable accompaniment of IBD that is sufficiently explained by the underlying disease and does not require further workup. But in contrast to these clinical routines, current guidelines recommend that any degree of anemia in patients with IBD should be further evaluated and treated. Multiple studies have shown that anemia is a main factor for decreased quality of life (QoL) in patients with IBD. Correction of anemia, however, can significantly improve the QoL of patients with IBD. It is therefore recommended that every patient with IBD is regularly screened for iron deficiency and anemia. If detected, appropriate workup and treatment should be initiated. Over the last years, a number of new diagnostic tools and treatment options have been developed. Multiple studies have demonstrated the safety of newer formulations of intravenous iron in patients with IBD and have compared oral and intravenous iron in various situations. Treatment recommendations have changed and new evidence-based guidelines were developed. However, to date these guidelines are still not widely implemented in clinical practice. The aim of this review is to draw attention to the need for treatment for every level of anemia in patients with IBD and to provide some practical guidance for screening, diagnostics, treatment and follow up of IDA in patients with IBD following current international guidelines.
Collapse
Affiliation(s)
- Dorothea Niepel
- Department of Internal Medicine I (Gastroenterology, Hepatology, Infectious Diseases), University Hospital Tübingen, Tübingen, Germany
| | - Thomas Klag
- Department of Internal Medicine I (Gastroenterology, Hepatology, Infectious Diseases), University Hospital Tübingen, Tübingen, Germany
| | - Nisar P. Malek
- Department of Internal Medicine I (Gastroenterology, Hepatology, Infectious Diseases), University Hospital Tübingen, Tübingen, Germany
| | | |
Collapse
|
196
|
Chieppa M, Giannelli G. Immune Cells and Microbiota Response to Iron Starvation. Front Med (Lausanne) 2018; 5:109. [PMID: 29721497 PMCID: PMC5915481 DOI: 10.3389/fmed.2018.00109] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/03/2018] [Indexed: 12/21/2022] Open
Abstract
Metal ions are essential for life on Earth, mostly as crucial components of all living organisms; indeed, they are necessary for bioenergetics functions as crucial redox catalysts. Due to the essential role of iron in biological processes, body iron content is finely regulated and is the battlefield of a tug-of-war between the host and the microbiota.
Collapse
Affiliation(s)
- Marcello Chieppa
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology "S. de Bellis", Research Hospital, Castellana Grotte, Italy
| |
Collapse
|
197
|
Abstract
Enterococci are important human commensals and significant opportunistic pathogens. Biofilm-related enterococcal infections, such as endocarditis, urinary tract infections, wound and surgical site infections, and medical device-associated infections, often become chronic upon the formation of biofilm. The biofilm matrix establishes properties that distinguish this state from free-living bacterial cells and increase tolerance to antimicrobial interventions. The metabolic versatility of the enterococci is reflected in the diversity and complexity of environments and communities in which they thrive. Understanding metabolic factors governing colonization and persistence in different host niches can reveal factors influencing the transition to biofilm pathogenicity. Here, we report a form of iron-dependent metabolism for Enterococcus faecalis where, in the absence of heme, extracellular electron transfer (EET) and increased ATP production augment biofilm growth. We observe alterations in biofilm matrix depth and composition during iron-augmented biofilm growth. We show that the ldh gene encoding l-lactate dehydrogenase is required for iron-augmented energy production and biofilm formation and promotes EET. Bacterial metabolic versatility can often influence the outcome of host-pathogen interactions, yet causes of metabolic shifts are difficult to resolve. The bacterial biofilm matrix provides the structural and functional support that distinguishes this state from free-living bacterial cells. Here, we show that the biofilm matrix can immobilize iron, providing access to this growth-promoting resource which is otherwise inaccessible in the planktonic state. Our data show that in the absence of heme, Enterococcus faecalisl-lactate dehydrogenase promotes EET and uses matrix-associated iron to carry out EET. Therefore, the presence of iron within the biofilm matrix leads to enhanced biofilm growth.
Collapse
|
198
|
Miele E, Shamir R, Aloi M, Assa A, Braegger C, Bronsky J, de Ridder L, Escher JC, Hojsak I, Kolaček S, Koletzko S, Levine A, Lionetti P, Martinelli M, Ruemmele F, Russell RK, Boneh RS, van Limbergen J, Veereman G, Staiano A. Nutrition in Pediatric Inflammatory Bowel Disease: A Position Paper on Behalf of the Porto Inflammatory Bowel Disease Group of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr 2018; 66:687-708. [PMID: 29570147 DOI: 10.1097/mpg.0000000000001896] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS A growing body of evidence supports the need for detailed attention to nutrition and diet in children with inflammatory bowel disease (IBD). We aimed to define the steps in instituting dietary or nutritional management in light of the current evidence and to offer a useful and practical guide to physicians and dieticians involved in the care of pediatric IBD patients. METHODS A group of 20 experts in pediatric IBD participated in an iterative consensus process including 2 face-to-face meetings, following an open call to Nutrition Committee of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition Porto, IBD Interest, and Nutrition Committee. A list of 41 predefined questions was addressed by working subgroups based on a systematic review of the literature. RESULTS A total of 53 formal recommendations and 47 practice points were endorsed with a consensus rate of at least 80% on the following topics: nutritional assessment; macronutrients needs; trace elements, minerals, and vitamins; nutrition as a primary therapy of pediatric IBD; probiotics and prebiotics; specific dietary restrictions; and dietary compounds and the risk of IBD. CONCLUSIONS This position paper represents a useful guide to help the clinicians in the management of nutrition issues in children with IBD.
Collapse
Affiliation(s)
- Erasmo Miele
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," Naples, Italy
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marina Aloi
- Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, Rome, Italy
| | - Amit Assa
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Christian Braegger
- Division of Gastroenterology and Nutrition, University Children's Hospital Zurich, Zurich, Switzerland
| | - Jiri Bronsky
- Gastroenterology and Nutrition Unit, Department of Paediatrics, 2nd Faculty of Medicine, Charles, University and Motol University Hospital, Prague, Czech Republic
| | - Lissy de Ridder
- Department of Paediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Johanna C Escher
- Department of Paediatric Gastroenterology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Iva Hojsak
- Department of Gastroenterology, Children Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sanja Kolaček
- Department of Gastroenterology, Children Hospital Zagreb, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Sibylle Koletzko
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Arie Levine
- Wolfson Medical Center, Sackler School of Medicine, Tel-Aviv, Israel
| | - Paolo Lionetti
- Meyer Children Hospital, University of Florence, Florence, Italy
| | - Massimo Martinelli
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," Naples, Italy
| | - Frank Ruemmele
- Université Sorbonne Paris Cité, Université Paris Descartes, and Assistance publique-hôpitaux de Paris, Hôpital Necker-Enfants malades, Service de gastroentérologie pédiatrique, Paris, France
| | - Richard K Russell
- Department of Paediatric Gastroenterology, The Royal Hospital for Children, Glasgow, Scotland
| | | | - Johan van Limbergen
- Department of Pediatrics, Division of Pediatric Gastroenterology & Nutrition, IWK Health Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gigi Veereman
- Department of Paediatric Gastroenterology and Nutrition, University Hospital Brussels, Free University Brussels, Brussels, Belgium
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples "Federico II," Naples, Italy
| |
Collapse
|
199
|
Nielsen OH, Soendergaard C, Vikner ME, Weiss G. Rational Management of Iron-Deficiency Anaemia in Inflammatory Bowel Disease. Nutrients 2018; 10:nu10010082. [PMID: 29342861 PMCID: PMC5793310 DOI: 10.3390/nu10010082] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023] Open
Abstract
Anaemia is the most frequent, though often neglected, comorbidity of inflammatory bowel disease (IBD). Here we want to briefly present (1) the burden of anaemia in IBD, (2) its pathophysiology, which mostly arises from bleeding-associated iron deficiency, followed by (3) diagnostic evaluation of anaemia, (4) a balanced overview of the different modes of iron replacement therapy, (5) evidence for their therapeutic efficacy and subsequently, (6) an updated recommendation for the practical management of anaemia in IBD. Following the introduction of various intravenous iron preparations over the last decade, questions persist about when to use these preparations as opposed to traditional and other novel oral iron therapeutic agents. At present, oral iron therapy is generally preferred for patients with quiescent IBD and mild iron-deficiency anaemia. However, in patients with flaring IBD that hampers intestinal iron absorption and in those with inadequate responses to or side effects with oral preparations, intravenous iron supplementation is the therapy of choice, although information on the efficacy of intravenous iron in patients with active IBD and anaemia is scare. Importantly, anaemia in IBD is often multifactorial and a careful diagnostic workup is mandatory for optimized treatment. Nevertheless, limited information is available on optimal therapeutic start and end points for treatment of anaemia. Of note, neither oral nor intravenous therapies seem to exacerbate the clinical course of IBD. However, additional prospective studies are still warranted to determine the optimal therapy in complex conditions such as IBD.
Collapse
Affiliation(s)
- Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, DK-2730, Denmark.
| | - Christoffer Soendergaard
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, DK-2730, Denmark.
| | - Malene Elbaek Vikner
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Herlev, DK-2730, Denmark.
| | - Günter Weiss
- Department of Internal Medicine II, Medical University Hospital of Innsbruck, Innsbruck, A-6020, Austria.
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research, University of Innsbruck, Innsbruck, A-6020, Austria..
| |
Collapse
|
200
|
Coleman OI, Haller D. Bacterial Signaling at the Intestinal Epithelial Interface in Inflammation and Cancer. Front Immunol 2018; 8:1927. [PMID: 29354132 PMCID: PMC5760496 DOI: 10.3389/fimmu.2017.01927] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 12/15/2017] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract provides a compartmentalized interface with an enormous repertoire of immune and metabolic activities, where the multicellular structure of the mucosa has acquired mechanisms to sense luminal factors, such as nutrients, microbes, and a variety of host-derived and microbial metabolites. The GI tract is colonized by a complex ecosystem of microorganisms, which have developed a highly coevolved relationship with the host’s cellular and immune system. Intestinal epithelial pattern recognition receptors (PRRs) substantially contribute to tissue homeostasis and immune surveillance. The role of bacteria-derived signals in intestinal epithelial homeostasis and repair has been addressed in mouse models deficient in PRRs and signaling adaptors. While critical for host physiology and the fortification of barrier function, the intestinal microbiota poses a considerable health challenge. Accumulating evidence indicates that dysbiosis is associated with the pathogenesis of numerous GI tract diseases, including inflammatory bowel diseases (IBD) and colorectal cancer (CRC). Aberrant signal integration at the epithelial cell level contributes to such diseases. An increased understanding of bacterial-specific structure recognition and signaling mechanisms at the intestinal epithelial interface is of great importance in the translation to future treatment strategies. In this review, we summarize the growing understanding of the regulation and function of the intestinal epithelial barrier, and discuss microbial signaling in the dynamic host–microbe mutualism in both health and disease.
Collapse
Affiliation(s)
| | - Dirk Haller
- Technical University of Munich, Munich, Germany.,ZIEL-Institute for Food & Health, Technical University of Munich, Munich, Germany
| |
Collapse
|