Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Food Funct ; 14(8): 3673-3685, 2023 Apr 24.
Article in English | MEDLINE | ID: mdl-36970974

ABSTRACT

The detrimental effects of high concentrations of colonic iron have been linked to intestinal inflammation and microbial dysbiosis. Exploiting chelation against this luminal pool of iron may restore intestinal health and have beneficial impacts on microbial communities. This study aimed to explore whether lignin, a heterogenous polyphenolic dietary component, has iron-binding affinity and can sequester iron within the intestine and thus, potentially modulate the microbiome. Within in vitro cell-culture models, the treatment of RKO and Caco-2 cells with lignin almost abolished intracellular iron import (96% and 99% reduction of iron acquisition respectively) with corresponding changes in iron metabolism proteins (ferritin and transferrin receptor-1) and reductions in the labile-iron pool. In a Fe-59 supplemented murine model, intestinal iron absorption was significantly inhibited by 30% when lignin was co-administered compared to the control group with the residual iron lost in the faeces. The supplementation of lignin into a microbial bioreactor colonic model increased the solubilisation and bio-accessibility of iron present by 4.5-fold despite lignin-iron chelation previously restricting intracellular iron absorption in vitro and in vivo. The supplementation of lignin in the model increased the relative abundance of Bacteroides whilst levels of Proteobacteria decreased which could be attributed to the changes in iron bio-accessibility due to iron chelation. In summary, we demonstrate that lignin is an effective luminal iron chelator. Iron chelation leads to the limitation of intracellular iron import whilst, despite increasing iron solubility, favouring the growth of beneficial bacteria.


Subject(s)
Gastrointestinal Microbiome , Iron , Humans , Animals , Mice , Iron/metabolism , Lignin , Iron Radioisotopes/pharmacology , Caco-2 Cells , Intestines/microbiology , Iron Chelating Agents/pharmacology
2.
EBioMedicine ; 81: 104088, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35660786

ABSTRACT

BACKGROUND: Faecal microbiota transplantation (FMT) has previously been explored as a treatment for ulcerative colitis (UC) however, biomarkers that predict and / or are associated with clinical response are poorly defined. The aim of this systematic review was to identify donor and recipient clinical, microbial and metabolomic predictive biomarkers of response to FMT in UC. METHODS: A systematic search of the relevant literature of studies exploring FMT in UC was conducted. Data on microbial diversity, taxonomic changes, metabolic changes, donor and recipient microbiota relationship and baseline predictors was examined. FINDINGS: 2852 studies were screened, and 25 papers were included in this systematic review. Following FMT, alpha diversity was seen to increase in responders along with increases in the abundance of Clostridiales clusters (order) and Bacteroides genus. Metabolomic analysis revealed short chain fatty acid (SCFA) production as a marker of FMT success. Donors or FMT batches with higher microbial alpha diversity and a greater abundance of taxa belonging to certain Bacteroides and Clostridia clusters were associated with clinical response to FMT. Baseline clinical predictors of response in patients with UC included younger age, less severe disease and possibly shorter disease duration. Baseline recipient microbial predictors at response consisted of higher faecal species richness, greater abundance of Candida and donor microbial profile similarity. INTERPRETATION: Distinct changes in gut microbiota profiles post-FMT indicate that certain baseline characteristics along with specific microbial and metabolomic alterations may predispose patients towards a successful therapeutic outcome. Opportunities towards a biomarker led precision medicine approach with FMT should be explored in future clinical studies. FUNDING: There no specific funding to declare.


Subject(s)
Colitis, Ulcerative , Gastrointestinal Microbiome , Biomarkers , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/etiology , Colitis, Ulcerative/therapy , Fecal Microbiota Transplantation/adverse effects , Feces , Humans , Treatment Outcome
3.
J Nutr Biochem ; 101: 108929, 2022 03.
Article in English | MEDLINE | ID: mdl-34954079

ABSTRACT

The toxic effects of excess dietary iron within the colonic lumen are well documented, particularly in the context of Inflammatory Bowel Disease (IBD) and Colorectal Cancer (CRC). Proposed mechanisms that underpin iron-associated intestinal disease include: (1) the pro-inflammatory and ROS-promoting nature of iron, (2) gene-expression alterations, and (3) intestinal microbial dysbiosis. However, to date no studies have examined the effect of iron on the colonic epigenome. Here we demonstrate that chronic iron exposure of colonocytes leads to significant hypomethylation of the epigenome. Bioinformatic analysis highlights a significant epigenetic effect on NRF2 (nuclear factor erythroid 2-related factor 2) pathway targets (including NAD(P)H Quinone Dehydrogenase 1 [NQO1] and Glutathione peroxidase 2 [GPX2]); this demethylating effect was validated and subsequent gene and protein expression quantified. These epigenetic modifications were not observed upon the diminishment of cellular lipid peroxidation with endogenous glutathione and the subsequent removal of iron. Additionally, the induction of TET1 expression was found post-iron treatment, highlighting the possibility of an oxidative-stress induction of TET1 and subsequent hypomethylation of NRF2 targets. In addition, a strong time dependence on the establishment of iron-orchestrated hypomethylation was found which was concurrent with the increase in the intracellular labile iron pool (LIP) and lipid peroxidation levels. These epigenetic changes were further validated in murine intestinal mucosa in models administered a chronic iron diet, providing evidence for the likelihood of dietary-iron mediated epigenetic alterations in vivo. Furthermore, significant correlations were found between NQO1 and GPX2 demethylation and human intestinal tissue iron-status, thus suggesting that these iron-mediated epigenetic modifications are likely in iron-replete enterocytes. Together, these data describe a novel mechanism by which excess dietary iron is able to alter the intestinal phenotype, which could have implications in iron-mediated intestinal disease and the regulation of ferroptosis.


Subject(s)
Enterocytes/metabolism , Epigenesis, Genetic , Glutathione Peroxidase/genetics , Intestinal Mucosa/metabolism , Iron, Dietary , Iron/metabolism , NAD(P)H Dehydrogenase (Quinone)/genetics , Animals , Caco-2 Cells , Colon/metabolism , DNA Methylation , Epigenome , Ferritins/genetics , Ferritins/metabolism , Ferrous Compounds/pharmacology , Glutathione Peroxidase/metabolism , Humans , Mice , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Oxidoreductases/genetics , Oxidoreductases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34521767

ABSTRACT

Early stages of colorectal cancer (CRC) development are characterized by a complex rewiring of transcriptional networks resulting in changes in the expression of multiple genes. Here, we demonstrate that the deletion of a poorly studied tetraspanin protein Tspan6 in Apcmin/+ mice, a well-established model for premalignant CRC, resulted in increased incidence of adenoma formation and tumor size. We demonstrate that the effect of Tspan6 deletion results in the activation of EGF-dependent signaling pathways through increased production of the transmembrane form of TGF-α (tmTGF-α) associated with extracellular vesicles. This pathway is modulated by an adaptor protein syntenin-1, which physically links Tspan6 and tmTGF-α. In support of this, the expression of Tspan6 is frequently decreased or lost in CRC, and this correlates with poor survival. Furthermore, the analysis of samples from the epidermal growth factor receptor (EGFR)-targeting clinical trial (COIN trial) has shown that the expression of Tspan6 in CRC correlated with better patient responses to EGFR-targeted therapy involving Cetuximab. Importantly, Tspan6-positive patients with tumors in the proximal colon (right-sided) and those with KRAS mutations had a better response to Cetuximab than the patients that expressed low Tspan6 levels. These results identify Tspan6 as a regulator of CRC development and a potential predictive marker for EGFR-targeted therapies in CRC beyond RAS pathway mutations.


Subject(s)
Biomarkers, Tumor/metabolism , Cetuximab/pharmacology , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Tetraspanins/metabolism , Tetraspanins/physiology , Animals , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Prognosis , Survival Rate , Tetraspanins/genetics , Tumor Cells, Cultured
5.
J Crohns Colitis ; 14(7): 935-947, 2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32016358

ABSTRACT

BACKGROUND: Although a majority of patients with PSC have colitis [PSC-IBD; primary sclerosing cholangitis-inflammatory bowel disease], this is phenotypically different from ulcerative colitis [UC]. We sought to define further the pathophysiological differences between PSC-IBD and UC, by applying a comparative and integrative approach to colonic gene expression, gut microbiota and immune infiltration data. METHODS: Colonic biopsies were collected from patients with PSC-IBD [n = 10], UC [n = 10], and healthy controls [HC; n = 10]. Shotgun RNA-sequencing for differentially expressed colonic mucosal genes [DEGs], 16S rRNA analysis for microbial profiling, and immunophenotyping were performed followed by multi-omic integration. RESULTS: The colonic transcriptome differed significantly between groups [p = 0.01]. Colonic transcriptomes from HC were different from both UC [1343 DEGs] and PSC-IBD [4312 DEGs]. Of these genes, only 939 had shared differential gene expression in both UC and PSC-IBD compared with HC. Imputed pathways were predominantly associated with upregulation of immune response and microbial defense in both disease cohorts compared with HC. There were 1692 DEGs between PSC-IBD and UC. Bile acid signalling pathways were upregulated in PSC-IBD compared with UC [p = 0.02]. Microbiota profiles were different between the three groups [p = 0.01]; with inferred function in PSC-IBD also being consistent with dysregulation of bile acid metabolism. Th17 cells and IL17-producing CD4 cells were increased in both PSC-IBD and UC when compared with HC [p < 0.05]. Multi-omic integration revealed networks involved in bile acid homeostasis and cancer regulation in PSC-IBD. CONCLUSIONS: Colonic transcriptomic and microbiota analysis in PSC-IBD point toward dysregulation of colonic bile acid homeostasis compared with UC. This highlights important mechanisms and suggests the possibility of novel approaches in treating PSC-IBD.


Subject(s)
Bile Acids and Salts/metabolism , Cholangitis, Sclerosing/etiology , Colitis, Ulcerative/etiology , Gastrointestinal Microbiome , Transcriptome , Adult , Case-Control Studies , Cholangitis, Sclerosing/pathology , Colitis, Ulcerative/pathology , Colon/pathology , Computational Biology , Female , Homeostasis , Humans , Immunity/genetics , Immunophenotyping , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Middle Aged , RNA, Ribosomal, 16S/analysis , Sequence Analysis, RNA , Signal Transduction/genetics , Th17 Cells/metabolism , Up-Regulation
6.
Nutrients ; 11(3)2019 Mar 21.
Article in English | MEDLINE | ID: mdl-30901846

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.


Subject(s)
Alginates/pharmacology , Dietary Supplements , Gastrointestinal Microbiome/drug effects , Iron Chelating Agents/pharmacology , Prebiotics , Adult , Bacteria/metabolism , Colon/metabolism , Colon/microbiology , Dysbiosis/metabolism , Feasibility Studies , Female , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Healthy Volunteers , Humans , Inflammatory Bowel Diseases/microbiology , Iron/metabolism , Male , Middle Aged
7.
Oncol Rep ; 39(1): 392-400, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115635

ABSTRACT

The obesity epidemic is associated with increases in the incidence of several types of cancer, including colorectal cancer, and is associated with poor outcomes for patients. Adipose tissue is considered biologically active and represents a plausible link between cancer and obesity due to the many factors that it secretes. In the present study, human adipose tissue was cultured in vitro and predifferentiated adipocyte secretome [preadipocyte (PAS)] and differentiated adipocyte secretome (DAS) were collected. Quantification of interleukin-6 (IL-6), leptin and hepcidin in the DAS medium was compared to the PAS medium. Fold change levels of hepcidin, leptin and IL-6 in DAS (2.88±0.28, 12.34±0.95 and 31.29±1.89 fold increases) were significantly higher compared to these in PAS (p=0.05). The SW480 colorectal cancer cells were co-cultured with DAS in the presence or absence of leptin, IL-6 or hepcidin inhibitors and cellular viability and proliferation assays were performed. The culture of SW480 with DAS increased the cell proliferation and viability by 30 and 15% (p=0.02 and p=0.03) respectively, which was reversed in the presence of inhibitors. Challenging the SW480 cells with IL-6 or hepcidin significantly elevated colonocyte­secreted leptin (p=0.05). Challenging the SW480 cells with leptin or hepcidin resulted in elevated levels of colonocyte-secreted IL-6 (p=0.05). Similarly, challenging cells with either IL-6 or leptin markedly elevated the level of secreted hepcidin (p=0.05) and this was associated with an induction in colonocyte iron levels in both cases. Collectively, these data revealed that adipocyte-secreted factors can ultimately modulate colonocyte iron levels and phenotype.


Subject(s)
Adipocytes/cytology , Colorectal Neoplasms/etiology , Hepcidins/metabolism , Interleukin-6/metabolism , Leptin/metabolism , Obesity/complications , Adipocytes/metabolism , Aged , Body Mass Index , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Colorectal Neoplasms/metabolism , Female , Humans , Male , Middle Aged , Obesity/metabolism , Up-Regulation , Wnt Signaling Pathway
8.
J Pathol Clin Res ; 3(3): 155-170, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28770100

ABSTRACT

Endothelial Protein C Receptor (EPCR) is a Major Histocompatibility Complex homologue, with established roles downregulating coagulation and in endothelial protection. Expressed predominantly on endothelium, EPCR affects inflammatory, apoptotic and cell proliferation pathways by binding to activated protein C (APC). However, EPCR can also be expressed on cancer cells, although the underlying reasons are unclear. Moreover, although EPCR has been linked with chemosensitivity in lung cancer, its clinical significance in many tumours is unknown. Here, we explored its significance in colorectal cancer (CRC). Bioinformatic methods revealed EPCR overexpression in many epithelial cancers, which was confirmed on CRC epithelial tumour cells by immunohistochemistry. EPCR upregulation resulted from gene amplification and DNA hypomethylation, and occurred in concert with a cohort of neighbouring genes on chromosome 20q, a region previously implicated in chemoresistance. As in endothelial cells, EPCR reproducibly mediated ERK pathway activation in a model CRC cell line following APC treatment. However, EPCR knockdown studies failed to highlight compelling EPCR-intrinsic impact on CRC cell phenotype, with limited effects on chemosensitivity and no effect on invasion observed, while EPCR appeared to decrease CRC cell migration. Consistent with these observations, differential EPCR expression did not influence response to chemotherapy in a human CRC cohort. Our results provide a compelling explanation for how EPCR is upregulated in diverse epithelial malignancies. They indicate that the clinical significance of EPCR varies across different tumour types. Furthermore, they raise the possibility that the prognostic significance of EPCR in certain tumours relates significantly to co-upregulation of neighbouring genes on chromosome 20q. Therefore, efforts to exploit EPCR as a prognostic marker should be focussed on specific tumours, and in such scenarios EPCR-co-dysregulated genes may represent potential axes for therapeutic intervention.

9.
Eur Respir J ; 49(4)2017 04.
Article in English | MEDLINE | ID: mdl-28404645

ABSTRACT

IREB2 is a gene that produces iron regulatory protein 2 (IRP2), which is critical to intracellular iron homeostasis and which relates to the rate of cellular proliferation. IREB2 lies in a lung cancer susceptibility locus. The aims were to assess 1) the relationship between iron loading, cell proliferation and IRP2 expression in lung cancer; 2) the potential of iron related pathways as therapeutic targets; and 3) the relevance of IRP2 in operated lung cancer patients.Cells of two nonsmall cell cancer (NSCLC) lines and primary bronchial epithelial cells (PBECs) were cultured with and without iron; and proliferation, apoptosis and migration were assessed. Reverse transcriptase PCR and Western blot were used to assess expression of iron homeostasis genes/proteins. Iron chelation and knockdown of IREB2 were used in vitro to explore therapeutics. A cohort of operated NSCLC patients was studied for markers of systemic iron status, tumour IRP2 staining and survival.Iron loading caused cell proliferation in cancer cell lines, which were less able to regulate IREB2 expression than PBECs. Iron chelation resulted in a return of proliferation rates to baseline levels; knockdown of IREB2 had a similar effect. IRP2-positive tumours were larger (p=0.045) and higher percentage staining related to poorer survival (p=0.079).Loss of iron regulation represents a poor prognostic marker in lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Iron Regulatory Protein 2/genetics , Iron/metabolism , Lung Neoplasms/genetics , Aged , Apoptosis , Cell Line , Cell Proliferation , Epithelial Cells/metabolism , Female , Gene Expression , Gene Knockdown Techniques , Humans , Iron Regulatory Protein 2/metabolism , Lung/pathology , Male , Neoplasm Staging , Prognosis , Proportional Hazards Models
10.
Cancer Sci ; 108(6): 1135-1143, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28281325

ABSTRACT

A role for iron in carcinogenesis is supported by evidence that iron metabolism proteins are modulated in cancer progression. To date, however, the expression of iron regulatory protein-2 (IRP2), which is known to regulate several iron metabolism proteins, has not been assessed in colorectal cancer. Expression of IRP2 was assessed by quantitative RT-PCR and immunohistochemistry in human colorectal cancer tissue. By interrogating The Cancer Genome Atlas (TCGA) database, expression of IRP2 and transferrin receptor-1 (TfR1) was assessed relative to common mutations that are known to occur in cancer. The impact of suppressing IRP2 on cellular iron metabolism was also determined by using siRNA and by using the MEK inhibitor trametinib. IRP2 was overexpressed in colorectal cancer compared to normal colonic mucosa and its expression was positively correlated with TfR1 expression. In addition, IRP2 expression was associated with mutations in BRAF. The MEK inhibitor trametinib suppressed IRP2 and this was associated with a suppression in TfR1 and the labile iron pool (LIP). Moreover, epidermal growth factor stimulation resulted in decreased ferritin expression and an increase in the LIP which were independent of IRP2. Results presented here suggest that ablating IRP2 provides a therapeutic platform for intervening in colorectal tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Iron Regulatory Protein 2/genetics , Mutation/genetics , Proto-Oncogene Proteins B-raf/genetics , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Ferritins/metabolism , HCT116 Cells , HT29 Cells , Humans , Iron/metabolism , RNA, Small Interfering/genetics , Receptors, Transferrin/metabolism
11.
Mol Nutr Food Res ; 61(3)2017 03.
Article in English | MEDLINE | ID: mdl-27794191

ABSTRACT

SCOPE: Excess free-iron is detrimental to health through its ability to participate in free radical generation and amplification of oncogenic pathways. The study aims were to identify polyphenols with iron-chelating potential. METHODS AND RESULTS: Of four polyphenols tested quercetin demonstrated potent iron binding with the physiological outcome dictated by the location of interaction. In the presence of extracellular iron and quercetin, ferritin expression and cellular iron concentrations decreased suggesting the resulting quercetin-iron complex is not internalised. However, in the relative absence of extracellular iron, quercetin becomes internalised and complexes with both intracellular iron, and iron which subsequently becomes absorbed as indicated by increased cellular 59 Fe post pre-culture with quercetin. This increased intracellular iron complexed to quercetin does not associate with the labile iron pool and cells behave as though they are iron deficient (increased transferrin receptor-1 and iron regulatory protein-2 expression and low ferritin expression). Additionally, a suppression in reactive oxygen species was observed. CONCLUSION: Quercetin, an exogenous iron chelator, is able to render the cell functionally iron-deficient which not only provides a therapeutic platform for chelating excess free luminal iron but also may be of use in limiting processes such as cancer-cell growth, inflammation and bacterial infections, which all require iron.


Subject(s)
Iron Chelating Agents/pharmacology , Iron/metabolism , Polyphenols/pharmacology , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Anthocyanins/pharmacokinetics , Anthocyanins/pharmacology , Antigens, CD/metabolism , Antioxidants/pharmacology , Biological Transport , Catechin/pharmacokinetics , Catechin/pharmacology , Cell Line, Tumor , Ferritins/metabolism , Glucosides/pharmacokinetics , Glucosides/pharmacology , Glutathione Peroxidase/metabolism , Humans , Iron/pharmacokinetics , Iron Chelating Agents/pharmacokinetics , Iron Regulatory Protein 2/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase , Polyphenols/pharmacokinetics , Quercetin/pharmacokinetics , Receptors, Transferrin/metabolism , Rutin/pharmacokinetics , Rutin/pharmacology
13.
Nanotechnology ; 27(46): 46LT02, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27734804

ABSTRACT

As the major iron storage protein, ferritin stores and releases iron for maintaining the balance of iron in fauna, flora, and bacteria. We present an investigation of the morphology and iron loading of ferritin (from equine spleen) using aberration-corrected high angle annular dark field scanning transmission electron microscopy. Atom counting method, with size selected Au clusters as mass standards, was employed to determine the number of iron atoms in the nanoparticle core of each ferritin protein. Quantitative analysis shows that the nuclearity of iron atoms in the mineral core varies from a few hundred iron atoms to around 5000 atoms. Moreover, a relationship between the iron loading and iron core morphology is established, in which mineral core nucleates from a single nanoparticle, then grows along the protein shell before finally forming either a solid or hollow core structure.

14.
Mol Nutr Food Res ; 60(9): 2098-108, 2016 09.
Article in English | MEDLINE | ID: mdl-27136256

ABSTRACT

SCOPE: Iron is an essential nutrient. However, in animal models, excess unabsorbed dietary iron residing within the colonic lumen has been shown to exacerbate inflammatory bowel disease and intestinal cancer. Therefore, the aims of this study were to screen a panel of alginates to identify a therapeutic that can chelate this pool of iron and thus be beneficial for intestinal health. METHODS AND RESULTS: Using several in vitro intestinal models, it is evident that only one alginate (Manucol LD) of the panel tested was able to inhibit intracellular iron accumulation as assessed by iron-mediated ferritin induction, transferrin receptor expression, intracellular (59) Fe concentrations, and iron flux across a Caco-2 monolayer. Additionally, Manucol LD suppressed iron absorption in mice, which was associated with increased fecal iron levels indicating iron chelation within the gastrointestinal tract. Furthermore, the bioactivity of Manucol LD was found to be highly dependent on both its molecular weight and its unique compositional sequence. CONCLUSION: Manucol LD could be useful for the chelation of this detrimental pool of unabsorbed iron and it could be fortified in foods to enhance intestinal health.


Subject(s)
Alginates/pharmacology , Iron Chelating Agents/pharmacology , Alginates/chemistry , Animals , Caco-2 Cells , Colon/drug effects , Colon/metabolism , Humans , Intestinal Mucosa/metabolism , Intestines/drug effects , Iron/metabolism , Male , Mice, Inbred Strains , Molecular Weight
15.
PLoS One ; 10(9): e0138240, 2015.
Article in English | MEDLINE | ID: mdl-26378798

ABSTRACT

Alginates are a class of biopolymers with known iron binding properties which are routinely used in the fabrication of iron-oxide nanoparticles. In addition, alginates have been implicated in influencing human iron absorption. However, the synthesis of iron oxide nanoparticles employs non-physiological pH conditions and whether nanoparticle formation in vivo is responsible for influencing cellular iron metabolism is unclear. Thus the aims of this study were to determine how alginate and iron interact at gastric-comparable pH conditions and how this influences iron metabolism. Employing a range of spectroscopic techniques under physiological conditions alginate-iron complexation was confirmed and, in conjunction with aberration corrected scanning transmission electron microscopy, nanoparticles were observed. The results infer a nucleation-type model of iron binding whereby alginate is templating the condensation of iron-hydroxide complexes to form iron oxide centred nanoparticles. The interaction of alginate and iron at a cellular level was found to decrease cellular iron acquisition by 37% (p < 0.05) and in combination with confocal microscopy the alginate inhibits cellular iron transport through extracellular iron chelation with the resulting complexes not internalised. These results infer alginate as being useful in the chelation of excess iron, especially in the context of inflammatory bowel disease and colorectal cancer where excess unabsorbed luminal iron is thought to be a driver of disease.


Subject(s)
Alginates/chemistry , Chelating Agents/chemistry , Ion Transport/physiology , Iron Compounds/chemistry , Iron/chemistry , Cell Line, Tumor , Humans , Iron/metabolism , Microscopy, Confocal , Microscopy, Electron, Scanning Transmission , Nanoparticles/chemistry
16.
J Nephrol ; 28(1): 81-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24687402

ABSTRACT

BACKGROUND: Hepcidin-25 is an iron regulator which reduces iron absorption and promotes sequestration in the reticulo-endothelial system. We investigated hepcidin and traditional iron storage marker utility in predicting haemoglobin increment following bolus intravenous iron. METHODS: The cohort included 129 consecutive non-dialysis chronic kidney disease patients that attended for intravenous iron over a 6-month period. Serum hepcidin-25 levels (determined by mass spectrometry) pre iron infusion and 6 weeks post were compared with ferritin and transferrin saturation in multivariate models. RESULTS: Log10 ferritin [coefficient 0.559 (0.435-0.684) p < 0.001] and log10 high-sensitive C-reactive protein [coefficient 0.092 (0.000-0.184) p = 0.049] were significantly associated with baseline log10 hepcidin-25 levels. Log10 estimated glomerular filtration rate was the only independent determinant of pre-infusion haemoglobin [coefficient 1.37 (0.16-2.59) p = 0.027]. Log10 hepcidin-25 was an independent predictor of haemoglobin increment 6 weeks following iron infusion [coefficient -0.84 (-1.38 to -0.31) p = 0.002]. Ferritin, transferrin saturation and hepcidin had similar predictive utility for a 1 g/dl haemoglobin increase (c-statistics: 0.68, 0.70, 0.69). CONCLUSIONS: Hepcidin is an iron sensor marker which predicts the magnitude of haemoglobin increment following protocolised intravenous iron infusion. Although displaying similar predictive performance to ferritin and transferrin saturation, hepcidin may also play a mechanistic role.


Subject(s)
Anemia/blood , Anemia/drug therapy , Hemoglobins/metabolism , Hepcidins/blood , Iron/administration & dosage , Renal Insufficiency, Chronic/blood , Administration, Intravenous , Adult , Aged , Anemia/etiology , C-Reactive Protein/metabolism , Female , Ferritins/blood , Glomerular Filtration Rate , Humans , Iron/adverse effects , Male , Middle Aged , Predictive Value of Tests , Renal Insufficiency, Chronic/complications , Transferrin/metabolism
17.
J Med Chem ; 57(13): 5817-22, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24905419

ABSTRACT

Examples of organometallic compounds as nucleoside analogues are rare within the field of medicinal bioorganometallic chemistry. We report on the synthesis and properties of two chiral ferrocene derivatives containing a nucleobase and a hydroxyalkyl group. These so-called ferronucleosides show promising anticancer activity, with cytostatic studies on five different cancer cell lines indicating that both functional groups are required for optimal activity.


Subject(s)
Ferrous Compounds/chemistry , Nucleosides/chemical synthesis , Organometallic Compounds/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Ferrous Compounds/pharmacokinetics , Humans , Leukemia L1210/drug therapy , Metallocenes , Nucleosides/pharmacology , Organometallic Compounds/chemistry
18.
J Clin Pharmacol ; 53(9): 885-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23740857

ABSTRACT

Iron plays a crucial role in a number of metabolic pathways including oxygen transport, DNA synthesis, and ATP generation. Although insufficient systemic iron can result in physical impairment, excess iron has also been implicated in a number of diseases including ischemic heart disease, diabetes, and cancer. Iron chelators are agents which bind iron and facilitate its excretion. Experimental iron chelators have demonstrated potent anti-neoplastic properties in a number of cancers in vitro. These agents have yet to be translated into clinical practice, however, largely due to the significant side effects encountered in pre-clinical models. A number of licensed chelators, however, are currently in clinical use for the treatment of iron overload associated with certain non-neoplastic diseases. Deferasirox is one such agent and the drug has shown significant anti-tumor effects in a number of in vitro and in vivo studies. Deferasirox is orally administered and has demonstrated a good side effect profile in clinical practice to date. It represents an attractive agent to take forward into clinical trials of iron chelators as anti-cancer agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzoates/therapeutic use , Iron Chelating Agents/therapeutic use , Neoplasms/drug therapy , Triazoles/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Deferasirox , Deferiprone , Deferoxamine/therapeutic use , Humans , Iron Chelating Agents/pharmacology , Pyridones/therapeutic use , Triazoles/pharmacology
19.
Transplantation ; 95(11): 1390-5, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23552742

ABSTRACT

BACKGROUND: Hepcidin-25 is a peptide hormone involved in iron absorption and homeostasis and found at increased serum levels in conditions involving systemic inflammation, renal dysfunction, and increased adiposity. Hepcidin may play a role in the pathogenesis of anemia, but its role in kidney transplantation is undefined. METHODS: This study enrolled 100 stable patients beyond 12 months after transplantation, from a large single United Kingdom center. Serum hepcidin-25 level, and relevant demographic and laboratory data pertinent to posttransplantation anemia, were measured and collected. Independent predictors of serum hepcidin were evaluated, and the relationship between hepcidin and hemoglobin, assessed. RESULTS: Independent associations were seen between higher hepcidin levels and allograft dysfunction (estimated glomerular filtration rate), increased inflammation (high-sensitivity C-reactive peptide), higher transferrin saturation (a marker of iron stores), and the use of marrow-suppressive medication (P<0.05 for all). Higher fat tissue index (whole-body multifrequency bioimpedance measurement) was also associated with higher hepcidin levels, but this relationship did not persist after adjustment for inflammation (high-sensitivity C-reactive peptide). In turn, inflammation was associated with increased fat tissue index (P=0.01) and male gender (P=0.04). A nonlinear association between serum hepcidin level and hemoglobin was seen, with a progressive fall in hemoglobin as hepcidin levels rose to 100 ng/mL, but little effect thereafter (P=0.009). This association was independent of renal dysfunction and female gender, both of which were also independently associated with a lower hemoglobin level. CONCLUSIONS: These results highlight possible mechanisms of hemoglobin reduction in kidney transplantation patients, and the therapeutic opportunities from understanding the role of hepcidin in this context.


Subject(s)
Antimicrobial Cationic Peptides/physiology , Hemoglobins/metabolism , Kidney Transplantation/physiology , Kidney/physiology , Adiposity/physiology , Adult , Antimicrobial Cationic Peptides/blood , C-Reactive Protein/metabolism , Cohort Studies , Female , Glomerular Filtration Rate/physiology , Hepcidins , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies , Sex Factors , United Kingdom
20.
Mol Pharmacol ; 83(1): 179-90, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23074173

ABSTRACT

Deferasirox is an orally effective iron (Fe) chelator currently used for the treatment of iron-overload disease and has been implemented as an alternative to the gold standard chelator, desferrioxamine (DFO). Earlier studies demonstrated that DFO exhibits anticancer activity due to its ability to deplete cancer cells of iron. In this investigation, we examined the in vitro and in vivo activity of deferasirox against cells from human solid tumors. To date, there have been no studies to investigate the effect of deferasirox on these types of tumors in vivo. Deferasirox demonstrated similar activity at inhibiting proliferation of DMS-53 lung carcinoma and SK-N-MC neuroepithelioma cell lines compared with DFO. Furthermore, deferasirox was generally similar or slightly more effective than DFO at mobilizing cellular (59)Fe and inhibiting iron uptake from human transferrin depending on the cell type. However, deferasirox potently inhibited DMS-53 xenograft growth in nude mice when given by oral gavage, with no marked alterations in normal tissue histology. To understand the antitumor activity of deferasirox, we investigated its effect on the expression of molecules that play key roles in metastasis, cell cycle control, and apoptosis. We demonstrated that deferasirox increased expression of the metastasis suppressor protein N-myc downstream-regulated gene 1 and upregulated the cyclin-dependent kinase inhibitor p21(CIP1/WAF1) while decreasing cyclin D1 levels. Moreover, this agent increased the expression of apoptosis markers, including cleaved caspase-3 and cleaved poly(ADP-ribose) polymerase 1. Collectively, we demonstrate that deferasirox is an orally effective antitumor agent against solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Iron Chelating Agents/pharmacology , Lung Neoplasms/drug therapy , Small Cell Lung Carcinoma/drug therapy , Triazoles/pharmacology , Administration, Oral , Animals , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Benzoates/therapeutic use , Cell Cycle/physiology , Cell Line, Tumor , Copper/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Deferasirox , Female , Humans , Iron/metabolism , Iron Chelating Agents/therapeutic use , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Neuroectodermal Tumors, Primitive, Peripheral , Protein Serine-Threonine Kinases/metabolism , Receptors, Transferrin/metabolism , Small Cell Lung Carcinoma/pathology , Transplantation, Heterologous , Triazoles/therapeutic use , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...