Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Adv Nutr ; 15(6): 100238, 2024 06.
Article in English | MEDLINE | ID: mdl-38729263

ABSTRACT

Vaccines can prevent infectious diseases, but their efficacy varies, and factors impacting vaccine effectiveness remain unclear. Iron deficiency is the most common nutrient deficiency, affecting >2 billion individuals. It is particularly common in areas with high infectious disease burden and in groups that are routinely vaccinated, such as infants, pregnant women, and the elderly. Recent evidence suggests that iron deficiency and low serum iron (hypoferremia) not only cause anemia but also may impair adaptive immunity and vaccine efficacy. A report of human immunodeficiency caused by defective iron transport underscored the necessity of iron for adaptive immune responses and spurred research in this area. Sufficient iron is essential for optimal production of plasmablasts and IgG responses by human B-cells in vitro and in vivo. The increased metabolism of activated lymphocytes depends on the high-iron acquisition, and hypoferremia, especially when occurring during lymphocyte expansion, adversely affects multiple facets of adaptive immunity, and may lead to prolonged inhibition of T-cell memory. In mice, hypoferremia suppresses the adaptive immune response to influenza infection, resulting in more severe pulmonary disease. In African infants, anemia and/or iron deficiency at the time of vaccination predict decreased response to diphtheria, pertussis, and pneumococcal vaccines, and response to measles vaccine may be increased by iron supplementation. In this review, we examine the emerging evidence that iron deficiency may limit adaptive immunity and vaccine responses. We discuss the molecular mechanisms and evidence from animal and human studies, highlight important unknowns, and propose a framework of key research questions to better understand iron-vaccine interactions.


Subject(s)
Adaptive Immunity , Iron Deficiencies , Iron , Vaccine Efficacy , Humans , Animals , Anemia, Iron-Deficiency/prevention & control , Anemia, Iron-Deficiency/immunology , Female , Nutritional Status , Mice , Pregnancy , Vaccination , Vaccines/immunology , Infant
2.
Int Arch Allergy Immunol ; 185(5): 449-455, 2024.
Article in English | MEDLINE | ID: mdl-38272014

ABSTRACT

INTRODUCTION: Iron deficiency is the most common cause of anemia in both sexes, although it is more common in women. Intravenous (IV) iron replacement is preferred in patients who cannot tolerate oral treatment or when iron stores need to be replenished rapidly. In this study, we wanted to share the ferric carboxymaltose (FCM) desensitization protocol that we self-created and successfully applied. METHODS: This retrospective cross-sectional study included patients with a history of hypersensitivity reactions (HSRs) to IV or oral iron replacement and patients who were planned to receive IV iron replacement but were referred to the allergy clinic because of have risk factors (atopic diseases, history of HSR to other drugs, high serum tryptase levels, etc.) for HSRs. Before desensitization, some of the patients underwent skin tests (skin prick test and intradermal test) with FCM, and the results were recorded. Skin tests were not performed in patients with a history of drug use (antihistamine, systemic steroid, omalizumab, etc.) that affected the results of skin tests. All patients underwent a one-bag 8-step desensitization protocol with 500 mg FCM and were observed for 2 h after desensitization. RESULTS: A total of 15 patients (14 females and 1 male) with a mean age of 41.13 ± 11.18 years were included in the study. When the patients were evaluated in terms of the risk of allergic reactions according to their clinical history, 8 patients had a history of anaphylaxis with iron preparations (FCM, n = 4; ferric hydroxide sucrose, n = 2; iron [II] glycine sulfate, n = 1; and iron [III] hydroxide polymaltose, n = 1), and 7 patients had a history of HSR other than anaphylaxis with iron preparations (urticaria, n = 6 [FCM, n = 2; iron (II) glycine sulfate, n = 2; and iron (III) hydroxide polymaltose, n = 2] and urticaria + angioedema [ferric hydroxide sucrose, n = 1]). Desensitization was successfully completed in all patients. No HSR was observed during or after the procedure in any of the patients. CONCLUSION: IV iron replacement is a very effective method, especially in cases where iron stores need to be replenished more rapidly. In patients with a history of iron HSR or at risk of developing HSR, replacement can be safely performed without an allergic reaction with successful desensitization protocols.


Subject(s)
Desensitization, Immunologic , Drug Hypersensitivity , Ferric Compounds , Maltose , Maltose/analogs & derivatives , Humans , Maltose/adverse effects , Maltose/administration & dosage , Desensitization, Immunologic/methods , Desensitization, Immunologic/adverse effects , Female , Male , Ferric Compounds/adverse effects , Ferric Compounds/administration & dosage , Drug Hypersensitivity/immunology , Drug Hypersensitivity/etiology , Drug Hypersensitivity/therapy , Adult , Middle Aged , Retrospective Studies , Cross-Sectional Studies , Skin Tests , Iron , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/immunology , Anemia, Iron-Deficiency/etiology
3.
Allergol. immunopatol ; 49(6): 42-47, nov. 2021. tab, graf
Article in English | IBECS | ID: ibc-215069

ABSTRACT

Background Iron deficiency anemia (IDA) is the most prevalent micronutrient deficiency in preschool children in developing countries including India. IDA is associated with immune perturbation, which is reflected in greater frequency of infections in these children. Recent research has shown three distinct monocyte subsets with distinct functions linked to infectious, inflammatory, and autoimmune diseases. These subsets have not been studied in children with IDA. Objective The aim of the study was to assess the percentage of monocyte population and the three subset populations in children with IDA and to compare the data with age-matched healthy controls. Methods Venous blood samples (5 mL) from 40 IDA children and 20 controls were collected after taking informed consent. Monocyte subpopulations were compared across the two groups. The outcome variables were calculated using Students Independent t-test or Mann–Whitney U test. P value of <0.05 was taken as significant. Results No significant difference was found in the absolute numbers as well as percentages of total monocytes between the control and case (study) group. Children in the IDA group showed a significant (p = 0.03) decrease in the nonclassical subset population when compared to the control group. Conclusion This is the first study done on monocyte subsets in iron-deficient children. Decrease in nonclassical monocytes observed may be associated with a pro-inflammatory state and increased risk of inflammatory and auto immune diseases. Follow-up studies are needed to confirm these findings (AU)


Subject(s)
Humans , Infant , Child, Preschool , Child , Anemia, Iron-Deficiency/immunology , Monocytes/immunology , Case-Control Studies , Cross-Sectional Studies , Flow Cytometry
4.
Allergol Immunopathol (Madr) ; 49(6): 42-47, 2021.
Article in English | MEDLINE | ID: mdl-34761655

ABSTRACT

BACKGROUND: Iron deficiency anemia (IDA) is the most prevalent micronutrient deficiency in preschool children in developing countries including India. IDA is associated with immune perturbation, which is reflected in greater frequency of infections in these children. Recent research has shown three distinct monocyte subsets with distinct functions linked to infectious, inflammatory, and autoimmune diseases. These subsets have not been studied in children with IDA. OBJECTIVE: The aim of the study was to assess the percentage of monocyte population and the three subset populations in children with IDA and to compare the data with age-matched healthy controls. METHODS: Venous blood samples (5 mL) from 40 IDA children and 20 controls were collected after taking informed consent. Monocyte subpopulations were compared across the two groups. The outcome variables were calculated using Students Independent t-test or Mann-Whitney U test. P value of <0.05 was taken as significant. RESULTS: No significant difference was found in the absolute numbers as well as percentages of total monocytes between the control and case (study) group. Children in the IDA group showed a significant (p = 0.03) decrease in the nonclassical subset population when compared to the control group. CONCLUSION: This is the first study done on monocyte subsets in iron-deficient children. Decrease in nonclassical monocytes observed may be associated with a pro-inflammatory state and increased risk of inflammatory and auto immune diseases. Follow-up studies are needed to confirm these findings.


Subject(s)
Anemia, Iron-Deficiency , Iron Deficiencies , Monocytes , Anemia, Iron-Deficiency/epidemiology , Anemia, Iron-Deficiency/immunology , Case-Control Studies , Child, Preschool , Humans , India/epidemiology , Iron , Iron Deficiencies/epidemiology , Iron Deficiencies/immunology , Monocytes/cytology
5.
Front Immunol ; 12: 635899, 2021.
Article in English | MEDLINE | ID: mdl-33777027

ABSTRACT

Iron deficiency, with or without anemia, is the most frequent hematological manifestation in individuals with cancer, and is especially common in patients with colorectal cancer. Iron is a vital micronutrient that plays an essential role in many biological functions, in the context of which it has been found to be intimately linked to cancer biology. To date, however, whereas a large number of studies have comprehensively investigated and reviewed the effects of excess iron on cancer initiation and progression, potential interrelations of iron deficiency with cancer have been largely neglected and are not well-defined. Emerging evidence indicates that reduced iron intake and low systemic iron levels are associated with the pathogenesis of colorectal cancer, suggesting that optimal iron intake must be carefully balanced to avoid both iron deficiency and iron excess. Since iron is vital in the maintenance of immunological functions, insufficient iron availability may enhance oncogenicity by impairing immunosurveillance for neoplastic changes and potentially altering the tumor immune microenvironment. Data from clinical studies support these concepts, showing that iron deficiency is associated with inferior outcomes and reduced response to therapy in patients with colorectal cancer. Here, we elucidate cancer-related effects of iron deficiency, examine preclinical and clinical evidence of its role in tumorigenesis, cancer progression and treatment response. and highlight the importance of adequate iron supplementation to limit these outcomes.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Cell Transformation, Neoplastic/metabolism , Colorectal Neoplasms/metabolism , Iron/metabolism , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/immunology , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/prevention & control , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Iron/immunology , Iron Compounds/therapeutic use , Oxidative Stress , Tumor Escape , Tumor Microenvironment
6.
Front Immunol ; 11: 571489, 2020.
Article in English | MEDLINE | ID: mdl-33362760

ABSTRACT

Superparamagnetic iron oxide nanoparticles (SPION) are employed as diagnostics and therapeutics following intravenous delivery for the treatment of iron deficiency anemia (IDA) in adult patients with chronic kidney failure. Neutrophils are the first defense against blood borne foreign insult and recruit to vascular sites of inflammation via a sequential process that is characterized by adhesive capture, rolling, and shear resistant arrest. A primary chemotactic agonist presented on the glycocalyx of inflamed endothelium is IL-8, which upon binding to its cognate membrane receptor (CXCR1/2) activates a suite of responses in neutrophils. An early response is degranulation with accompanying upregulation of ß2-integrin (CD11/CD18) and shedding of L-selectin (CD62L) receptors, which exert differential effects on the efficiency of endothelial recruitment. Feraheme is an FDA approved SPION treatment for IDA, but its effect on the innate immune response of neutrophils during inflammation has not been reported. Here, we studied the immunomodulatory effects of Feraheme on neutrophils freshly isolated from healthy human subjects and stimulated in suspension or on inflammatory mimetic substrates with IL-8. Cells treated with Feraheme exhibited reduced sensitivity to stimulation with IL-8, indicated by reduced upregulation of membrane CD11b/CD18 receptors, high affinity (HA) CD18, and shedding of CD62L. Feraheme also inhibited N-formyl-Met-Leu-Phe (fMLP) induced reactive oxygen species production. Neutrophil rolling, arrest, and migration was assessed in vascular mimetic microfluidic channels coated with E-selectin and ICAM-1 to simulate inflamed endothelium. Neutrophils exposed to Feraheme rolled faster on E-selectin and arrested less frequently on ICAM-1, in a manner dependent upon SPION concentration. Subsequent neutrophil shape change, and migration were also significantly inhibited in the presence of Feraheme. Lastly, Feraheme accelerated clearance of cytosolic calcium flux following IL-8 stimulation. We conclude that uptake of Feraheme by neutrophils inhibits chemotactic activation and downregulates normal rolling to arrest under shear flow. The mechanism involves increased calcium clearance following chemotactic activation, which may diminish the efficiency of recruitment from the circulation at vascular sites of inflammation.


Subject(s)
Anemia, Iron-Deficiency/immunology , Ferrosoferric Oxide/therapeutic use , Inflammation/therapy , Kidney Failure, Chronic/immunology , Magnetic Iron Oxide Nanoparticles/administration & dosage , Neutrophils/immunology , Anemia, Iron-Deficiency/therapy , CD18 Antigens/metabolism , Calcium Signaling , Cell Degranulation , Cells, Cultured , Humans , Interleukin-8/metabolism , Kidney Failure, Chronic/therapy , L-Selectin/metabolism , Neutrophil Activation , Receptors, Interleukin-8A/metabolism
7.
Int J Med Sci ; 17(14): 2232-2239, 2020.
Article in English | MEDLINE | ID: mdl-32922186

ABSTRACT

Background: Current evidence suggests an increased prevalence of iron deficiency (ID) and anemia in chronic obstructive pulmonary disease (COPD). ID and subsequent anemia can be due to iron losses via bleeding resulting in absolute ID or inflammation-driven retention of iron within macrophages resulting in functional ID and anemia of inflammation. Methods: This is a retrospective analysis of 204 non-exacerbated COPD patients in outpatient care. Current definitions of absolute and functional ID were applied to determine the prevalence of ID and to analyze associations to disease severity in terms of lung function parameters and clinical symptoms. Results: The studied cohort of COPD patients demonstrated a high prevalence of ID, ranging from 30 to 40% during the observation time. At the initial presentation, absolute or functional ID was found in 9.3% to 12.3% of COPD individuals, whereas combined forms of absolute and functional ID were most prevalent (25.9% of all individuals). The prevalence of ID increased during longitudinal follow-up (37 ± 15 months), and especially combined forms of ID were significantly related to anemia. Anemia prevalence ranged between 14.2% and 20.8% during the observation period and anemia was associated with lower FEV1, DLCOc, and CRP elevation. Accordingly, ID was associated with decreased FEV1, DLCOc, and an elevation in CRP. Conclusion: ID is common in COPD patients, but a uniform definition for accurate diagnosis does not exist. Prevalence of functional ID and anemia increased during follow-up. The associations of ID and anemia with reduced functional lung capacity and elevated inflammation may reflect a more severe COPD phenotype.


Subject(s)
Anemia, Iron-Deficiency/epidemiology , Iron Deficiencies , Pulmonary Disease, Chronic Obstructive/complications , Aged , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/immunology , C-Reactive Protein/analysis , Female , Follow-Up Studies , Forced Expiratory Volume , Humans , Inflammation/blood , Inflammation/diagnosis , Inflammation/immunology , Iron/blood , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Prevalence , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/immunology , Retrospective Studies , Severity of Illness Index
8.
Front Immunol ; 11: 1313, 2020.
Article in English | MEDLINE | ID: mdl-32754150

ABSTRACT

Background: Iron deficiency may impair adaptive immunity and is common among African infants at time of vaccination. Whether iron deficiency impairs vaccine response and whether iron supplementation improves humoral vaccine response is uncertain. Methods: We performed two studies in southern coastal Kenya. In a birth cohort study, we followed infants to age 18 mo and assessed whether anemia or iron deficiency at time of vaccination predicted vaccine response to three-valent oral polio, diphtheria-tetanus-whole cell pertussis-Haemophilus influenzae type b vaccine, ten-valent pneumococcal-conjugate vaccine and measles vaccine. Primary outcomes were anti-vaccine-IgG and seroconversion at age 24 wk and 18 mo. In a randomized trial cohort follow-up, children received a micronutrient powder (MNP) with 5 mg iron daily or a MNP without iron for 4 mo starting at age 7.5 mo and received measles vaccine at 9 and 18 mo; primary outcomes were anti-measles IgG, seroconversion and avidity at age 11.5 mo and 4.5 y. Findings: In the birth cohort study, 573 infants were enrolled and 303 completed the study. Controlling for sex, birthweight, anthropometric indices and maternal antibodies, hemoglobin at time of vaccination was the strongest positive predictor of: (A) anti-diphtheria and anti-pertussis-IgG at 24 wk (p = 0.0071, p = 0.0339) and 18 mo (p = 0.0182, p = 0.0360); (B) anti-pertussis filamentous hemagglutinin-IgG at 24 wk (p = 0.0423); and (C) anti-pneumococcus 19 IgG at 18 mo (p = 0.0129). Anemia and serum transferrin receptor at time of vaccination were the strongest predictors of seroconversion against diphtheria (p = 0.0484, p = 0.0439) and pneumococcus 19 at 18 mo (p = 0.0199, p = 0.0327). In the randomized trial, 155 infants were recruited, 127 and 88 were assessed at age 11.5 mo and 4.5 y. Compared to infants that did not receive iron, those who received iron at time of vaccination had higher anti-measles-IgG (p = 0.0415), seroconversion (p = 0.0531) and IgG avidity (p = 0.0425) at 11.5 mo. Interpretation: In Kenyan infants, anemia and iron deficiency at time of vaccination predict decreased response to diphtheria, pertussis and pneumococcal vaccines. Primary response to measles vaccine may be increased by iron supplementation at time of vaccination. These findings argue that correction of iron deficiency during early infancy may improve vaccine response.


Subject(s)
Anemia, Iron-Deficiency/immunology , Dietary Supplements , Iron/administration & dosage , Vaccines/administration & dosage , Antibodies, Bacterial/blood , Antibodies, Viral/blood , Child, Preschool , Cohort Studies , Double-Blind Method , Female , Follow-Up Studies , Humans , Immunity, Humoral , Immunoglobulin G/blood , Infant , Infant, Newborn , Male , Vaccination
9.
J Immunotoxicol ; 17(1): 144-152, 2020 12.
Article in English | MEDLINE | ID: mdl-32574507

ABSTRACT

The prevalence of iron (Fe) deficiency and subclinical lead (Pb) toxicity is high in developing countries like India, and information on their potential additive effects on immune responses is scant. The current study assessed immune parameters in dual Pb-exposed\Fe-deficient weanling SD rats. Rats were fed a control (CD) or Fe-deficient (ID) diet for 4 weeks and then evaluated for hemoglobin (Hb) and serum Fe status. Then, half the rats in each group began to receive daily oral Pb exposure (25 mg/4 ml/kg BW; gavage) or vehicle for a further 4 weeks (while maintained on original respective diets). After the 4-weeks of dosing, rats were assessed for Hb and serum Fe, and for blood lead level (BLL) and δ-aminolevulinic acid dehydratase (ALAD) activity. At this point, half the rats in each group (now n = 8) were then vaccinated with tetanus toxoid (TT), and then two boosters at 2-week intervals. All the time, rats stayed on their original respective diets along with exposure to Pb on alternate days. At 2 weeks after the final booster, rats were euthanized and blood collected to assess total/specific IgG and IgM levels; mucosal (intestinal) IgA levels were also determined. Spleens were taken to assess CD4+ and CD8+ cell levels and for ex vivo measures of splenocyte proliferation/TH1 and TH2 cytokine formation. The results indicated significant lowering of Hb and serum Fe levels in ID rats and increased blood Pb and decreased ALAD activity in all Pb-exposed rats. Fe-deficiency alone induced significant increases in ALAD activity, but only in an absence of Pb. While there was no impact of any regimen on total or TT-specific IgG, significant decreases in mucosal IgA and TT-specific IgM were seen in ID-fed Pb-exposed rats. CD4+ cell levels were not impacted by treatment; CD8+ levels were increased in all ID/Pb-exposed rats. Ex-vivo splenocyte proliferation was significantly higher among vaccinated rats, as well as ID-fed Pb-exposed unvaccinated rats. Cytokine formation in all cases was highly variable. The results suggest that Fe deficiency compromised cell-mediated, mucosal, and/or humoral immune response-related endpoints and that Pb exposure during the deficiency further impacted these outcomes.


Subject(s)
Anemia, Iron-Deficiency/immunology , Lead/toxicity , T-Lymphocytes/immunology , Tetanus Toxoid/immunology , Administration, Oral , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/diagnosis , Animals , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Hemoglobins/analysis , Humans , Immunity, Humoral/drug effects , Immunity, Mucosal/drug effects , Immunization, Secondary , Immunogenicity, Vaccine , Iron/blood , Iron Deficiencies , Lead/administration & dosage , Lymphocyte Activation/drug effects , Male , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Spleen/cytology , Spleen/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tetanus Toxoid/administration & dosage , Weaning
10.
Pak J Pharm Sci ; 33(1(Supplementary)): 253-261, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32122856

ABSTRACT

Micronutrient deficiencies (MNDs) are common worldwide, in both developing as well as developed countries. MNDs such as Iron Deficiency not only compromise the nutritional status of individuals but can also put them at an increased risk of developing various other diseases by negatively affecting their immunity. The objective of the current research was to determine the effects of prebiotics and iron fortificants on various immunoglobulins among iron deficient women belonging to childbearing age. To serve the purpose, a total of seventy five iron deficient women were selected and randomly divided into one control and four treatment groups. Accordingly, different types of fortified wheat flour were prepared, based on varying dosage of prebiotics and iron fortificants, to be fed to anemic women on daily basis for three months. Two iron salts (FeSO4 and NaFeEDTA) and two prebiotics (Galacto oligosaccharides and Inulin) were used to fortify wheat flour during the trials. Overnight fasted women were asked to give blood samples on monthly basis, up to three months. Four types of Immunoglobulins including IgA, IgE, IgG and IgM were determined at baseline, 30th, 60th and 90th day of trials using their respective protocols. The results of the study indicated that a statistically significant declining trend for IgA, IgE, IgG and IgM was present among the treatment groups (P-value < 0.05), compared to the control group. The study concluded that provision of iron and prebiotic fortified flour improved the immune function of iron deficient women.


Subject(s)
Anemia, Iron-Deficiency/diet therapy , Anemia, Iron-Deficiency/immunology , Flour , Food, Fortified , Iron/administration & dosage , Prebiotics/administration & dosage , Adolescent , Adult , Anemia, Iron-Deficiency/blood , Female , Humans , Immunomodulation/drug effects , Immunomodulation/physiology , Nutritional Status/drug effects , Nutritional Status/physiology , Young Adult
11.
Biol Trace Elem Res ; 195(1): 117-124, 2020 May.
Article in English | MEDLINE | ID: mdl-31377936

ABSTRACT

The present study was to evaluate the consequences of iron status across oral and parenteral iron administrations in prevention of iron deficiency anemia. A total of 24 one-day-old male neonatal piglets were allocated into three groups given non-iron supplementation (NON), intramuscular iron dextran injection (FeDex), and oral administration of ferrous glycine chelate (FeGly), respectively. At day 8, no significant differences in final body weight, average weight gain, and tissue coefficients were observed among three groups (P > 0.05). Both oral FeGly and FeDex injection significantly increased serum iron, ferritin, hemoglobin, and tissue iron deposition (P < 0.05). However, FeDex-injected supplementation resulted in rapidly rising hepcidin levels and hepatic iron deposition (P < 0.05). In addition, compared to parenteral iron supplementation, greater serum IgA level, SOD, and GSH-Px activities, lower expressions of IL-1ß and TNF-α in the liver, and lower expressions of IL-6 and TNF-α in the spleen were found in oral iron piglets (P < 0.05). According to our results, oral administration of ferrous glycine chelate improved iron homeostasis, and oxidative and immune status in anemic neonatal pigs.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Homeostasis/drug effects , Iron Chelating Agents/pharmacology , Iron-Dextran Complex/pharmacology , Iron/immunology , Administration, Oral , Anemia, Iron-Deficiency/immunology , Animals , Homeostasis/immunology , Infusions, Parenteral , Iron Chelating Agents/administration & dosage , Iron-Dextran Complex/administration & dosage , Male , Oxidative Stress/drug effects , Oxidative Stress/immunology , Swine , Weight Gain/drug effects
12.
J Crohns Colitis ; 14(3): 316-322, 2020 Mar 13.
Article in English | MEDLINE | ID: mdl-31665264

ABSTRACT

BACKGROUND AND AIMS: Anaemia is common in patients with inflammatory bowel disease [IBD], its two main aetiologies being iron deficiency anaemia [IDA] and anaemia of chronic inflammation [ACI]. Impaired intestinal iron absorption due to inflammatory cytokines is thought to play a role in ACI. We undertook for the first time a controlled prospective study investigating effects of differing underlying diseases, disease locations, and types of iron deficiency or anaemia on oral iron absorption in adult IBD patients with and without inflammation. METHODS: This study was a comparative, single-centred open clinical trial in adults with IBD [n = 73] and healthy controls [n = 22]. Baseline parameters included blood count, iron status [ferritin, transferrin, transferrin saturation, soluble transferrin receptor, hepcidin, serum iron], high-sensitivity C-reactive protein [hsCRP] and interleukin-6. Iron absorption was tested using one oral, enteric-coated capsule containing 567.7 mg iron[II]-glycine-sulphate complex. Serum iron was determined 60/90/120/180/240 min after ingestion. RESULTS: Iron absorption capacity was shown to be influenced by inflammation and anaemia or iron deficiency [ID] type but not by underlying disease type or localisation. The ACI group showed a significantly lower iron absorption capacity than all others. Whereas hsCRP levels [-0.387, p < 0.001], IL-6 [-0.331, p = 0.006], ferritin [-0.531, p < 0.001], and serum hepcidin [-0.353, p = 0.003] correlated negatively with serum iron change at 2 h, transferrin showed a positive correlation at the same time point [0.379, p < 0.001]. CONCLUSIONS: Underlying disease type and localisation appear to have little effect on iron absorption capacity, whereas lack of response to oral iron correlates well with serum markers of inflammation. Iron absorption capacity is thus significantly reduced in the presence of inflammation.


Subject(s)
Anemia, Iron-Deficiency , Ferritins/blood , Inflammation/blood , Inflammatory Bowel Diseases , Interleukin-6/blood , Iron , Oral Mucosal Absorption/immunology , Transferrin/analysis , Adult , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/immunology , Biomarkers/blood , C-Reactive Protein/analysis , Correlation of Data , Female , Germany/epidemiology , Hepcidins/blood , Humans , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/epidemiology , Intestinal Absorption/immunology , Iron/blood , Iron Deficiencies , Male , Middle Aged
13.
Int J Biol Macromol ; 132: 213-221, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-30926500

ABSTRACT

As iron supplement, the antioxidant activities of APS-iron (III) complex were comprehensively evaluated by 5-axe cobweb charts, which indicated the APS-iron (III) complex had a certain antioxidant activity and been weaker than that of APS. The results of immunological activity experiments indicated the stimulation index increased with APS-iron (III) complex concentration increase. When the concentration of the APS-iron (III) complex was 50 µg/mL, the lymphocytes proliferation increased by 35.7% compared with APS. APS-iron (III) complex also had better complement fixing activity than APS, 0.589 mg/mL of which achieved 50% complement fixing activities. Through the iron supplement experiments on iron-deficiency anemia mouse model, we found the APS-iron (III) complex faster increased hemoglobin concentration, SOD, CAT and faster decreased MDA to the normal level than Niferex and ferrous sulfate. Histological results revealed that the tissue sections were clear without obvious pathological changes and bone marrow had most hematopoietic cells from APS-iron (III) complex rat group, which also proved the APS-iron (III) complex had no significant side effects. Therefore, APS-iron (III) complex may be developed as a multifunctional iron supplement for clinical application.


Subject(s)
Anemia, Iron-Deficiency/immunology , Anemia, Iron-Deficiency/metabolism , Astragalus propinquus/chemistry , Coordination Complexes/pharmacology , Iron/chemistry , Oxidative Stress/drug effects , Polysaccharides/chemistry , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Lipid Peroxidation/drug effects , Rats , Rats, Wistar
14.
BMJ Case Rep ; 12(12)2019 Dec 29.
Article in English | MEDLINE | ID: mdl-31888907

ABSTRACT

Coeliac disease (CD) is a small bowel disorder known for its intestinal manifestations like diarrhoea and weight loss. Less known are the extraintestinal manifestations of CD like haematological abnormalities but also altered female reproduction and pregnancy outcomes. Especially, undiagnosed CD may lead to adverse reproductive outcomes such as intrauterine growth restriction, stillbirth and preterm birth. In diagnosed and treated CD, adverse pregnancy outcomes might be prevented.


Subject(s)
Celiac Disease/complications , Duodenum/pathology , Fetal Growth Retardation/etiology , Adult , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/etiology , Anemia, Iron-Deficiency/immunology , Biopsy , Celiac Disease/diagnosis , Celiac Disease/diet therapy , Diagnosis, Differential , Diet, Gluten-Free/methods , Female , Fetal Growth Retardation/prevention & control , Humans , Pregnancy , Pregnancy Outcome/epidemiology , Premature Birth/prevention & control , Stillbirth/epidemiology , Thrombocythemia, Essential/diagnosis , Treatment Outcome
15.
Nutrients ; 10(6)2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29874829

ABSTRACT

Early nutrition is key to promoting gut growth and education of the immune system. Although iron deficiency anemia has long been recognized as a serious iron disorder, the effects of iron supplementation on gut development are less clear. Therefore, using suckling piglets as the model for iron deficiency, we assessed the impacts of iron supplementation on hematological status, gut development, and immunity improvement. Piglets were parenterally supplied with iron dextran (FeDex, 60 mg Fe/kg) by intramuscular administration on the third day after birth and slaughtered at the age of two days, five days, 10 days, and 20 days. It was expected that iron supplementation with FeDex improved the iron status with higher levels of serum iron, ferritin, transferrin, and iron loading in the liver by regulating the interaction of hepcidin and ferroportin (FPN). FeDex supplementation increased villus length and crypt depth, attenuated the pathological status of the duodenum, and was beneficial to intestinal mucosa. FeDex also influenced the intestinal immune development by stimulating the cytokines' production of the intestine and enhancing the phagocytotic capacity of monocytes. Overall, the present study suggested that iron supplementation helped promote the development of the intestine by improving its morphology, which maintains its mucosal integrity and enhances the expression of immuno-associated factors.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Duodenum/drug effects , Intestinal Mucosa/drug effects , Iron-Dextran Complex/administration & dosage , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/immunology , Anemia, Iron-Deficiency/physiopathology , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Cation Transport Proteins/metabolism , Cytokines/immunology , Dietary Supplements , Disease Models, Animal , Duodenum/growth & development , Duodenum/immunology , Duodenum/pathology , Ferritins/blood , Hepcidins/metabolism , Injections, Intramuscular , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Liver/drug effects , Liver/metabolism , Monocytes/drug effects , Monocytes/immunology , Nutritional Status , Phagocytosis/drug effects , Sus scrofa , Time Factors , Transferrin/metabolism
16.
Nutr Cancer ; 70(4): 546-556, 2018.
Article in English | MEDLINE | ID: mdl-29697284

ABSTRACT

Iron Deficiency Anemia (IDA) is a universal health problem and a risk factor for the development of cancer. IDA changes the microenvironment of the human body by affecting both the biological and immunological systems. It increases DNA damage and genomic instability by different mechanisms. IDA is one of the leading causes of the imbalance between different antioxidant enzymes as well as enzymes involved in DNA damage and DNA repair systems of the body. It can affect the biogenesis/expression of microRNAs. IDA interrupts the oxidative phosphorylation energy metabolism and intestinal Cytochrome-P450 systems. It also disturbs multicellular signaling pathways involved in cell survival and helps in tumor angiogenesis. Moreover, IDA is also responsible for the functional deterioration of innate and adaptive immune systems that lead to immunological dysfunctions against invading pathogens. Genomic instability and immunological dysfunctions are the hallmarks of cancer development. In this review, we will review the evidence linking IDA to increased cancer risk.


Subject(s)
Anemia, Iron-Deficiency/immunology , Neoplasms/etiology , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/genetics , Apoptosis , Cytochrome P-450 Enzyme System/metabolism , Genomic Instability , Heme Oxygenase-1/metabolism , Humans , Killer Cells, Natural/immunology , MicroRNAs/genetics , Mitochondria/metabolism , Mitochondria/pathology , NF-kappa B/immunology , Neoplasms/blood supply , Neoplasms/epidemiology , Neoplasms/pathology , Neovascularization, Pathologic , Oxidative Stress
17.
BMC Pediatr ; 18(1): 49, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29433459

ABSTRACT

BACKGROUND: Iron plays an important role in body defense and essential for normal immune system development where its deficiency may result in an inadequate immune response. We aimed to assess the lymphocyte subsets in childhood iron deficiency anemia (IDA) with their laboratory correlations. METHODS: Fifty IDA (< 18 years) and 25 age and sex-matched healthy children were enrolled and a complete history was obtained and clinical examination was performed. Complete blood count, serum iron, total iron binding capacity and serum ferritin, were performed. Flow cytometric determination of peripheral blood CD3+, CD4+, CD8+ T-lymphocytes and CD19+ B-lymphocytes and CD4/CD8 ratio were done. RESULTS: Patients had significantly lower hemoglobin, Serum iron, ferritin levels and higher lymphocytic count in patients compared with controls (p = 0.001, 0.03, 0.001, 0.001 respectively). CD3 count and percentage were significantly lower in IDA patients compared to controls (p = 0.007 and 0.005 respectively). There was a Significant reduction in the CD4 count, percentage and CD4/CD8 ratio in patients compared with controls (p = 0.001, 0.001 and 0.005 respectively) while there was no significant difference regarding CD8 count and percentage. No significant difference between the two studied groups regarding either CD19 count or percentage (p = 0.28 and 0.18 respectively) were found. CONCLUSIONS: IDA is associated with impaired cell-mediated immune response specifically T-cell mediated immunity.


Subject(s)
Anemia, Iron-Deficiency/immunology , Lymphocyte Subsets , Adolescent , Anemia, Iron-Deficiency/blood , CD3 Complex , CD4 Lymphocyte Count , CD4-CD8 Ratio , Case-Control Studies , Child , Child, Preschool , Female , Ferritins/blood , Flow Cytometry , Hemoglobinometry , Humans , Iron/blood , Male
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(5): 1314-1320, 2017 Oct.
Article in Chinese | MEDLINE | ID: mdl-29070101

ABSTRACT

OBJECTIVE: To analyze the promoter methylation status, mRNA expression and clinical significance of DKK-3 and WIF-1 genes in patients with acute myeloid leukemia(AML). METHODS: Methylation specific polymerase chain reaction (MS-PCR) mothod was carried out to detect DKK-3 and WIF-1 gene promoter methylation status in bone marrow specimen from 56 patients with AML and 20 patients with iron deficiency anaemia(IDA) as control; then the real-time quantitative reverse transcription polymerase chain reaction (RT-PCR) was used to detect mRNA expression of DKK-3, WIF-1 gene and ß -catenin in the above-mentioned specinens, and their relationship with the clinical features and survival time was analyzed. RESULTS: The promoter methylation rate of DKK-3 and WIF-1 gene in AML patients were significantly higher than that in control group(χ2=15.330,P<0.001; χ2=17.371,P<0.001). There was no relationship between DKK-3 and WIF-1 gene promoter methylation rate and AML patient's sex, age, clinical typing. The relative expression of DKK-3 and WIF-1 gene mRNA in AML group were 0.840±0.320 and 0.792±0.313, which were lower than those in control group (1.134±0.392 and 1.047±0.334) respectively, the difference was statistically significant (t=3.415,P=0.000; t=3.070, P=0.003). The relative expression of ß-catenin mRNA in AML bone marrow specimens in AML group was 0.756±0.304, which was higher than that in control group(0.342±0.105), the difference was statistically significant (t=5.943, P=0.001). The expression of DKK-3 and WIF-1 gene mRNA negatively correlated with ß-catenin mRNA(r=-0.543; r=-0.562). Kaplan Meier survival curve analysis showed that overall survival time in AML patients with DKK-3 gene methylation was shorter than that in the AML patients with DKK-3 gene unmethylation(χ2=3.957, P=0.042). Futhermore, the orerall survival time in AML patients with WIF-1 gene methylation was also shorter than that in AML patients with WIF-1 gene unmethylation (χ2=4.520, P=0.029). CONCLUSION: Wnt/ß-catenin signaling pathway is abnormally activated in AML patients, the DKK-3 and WIF-1 gene promoter methylation may be involved in Wnt pathways activation and the pathogenesis of AML.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Promoter Regions, Genetic , Repressor Proteins/metabolism , Anemia, Iron-Deficiency/genetics , Anemia, Iron-Deficiency/immunology , Anemia, Iron-Deficiency/metabolism , Chemokines , DNA Methylation , Humans , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/metabolism , RNA, Messenger
19.
Mini Rev Med Chem ; 17(17): 1677-1683, 2017.
Article in English | MEDLINE | ID: mdl-28440190

ABSTRACT

OBJECTIVE: Dioscorea opposita Thunb is the famous food and traditional medicine in China and it was rich in polysaccharides. Polysaccharides of Dioscorea Opposita Thunb possess immunoregulatory activity, free radical scavenging activity and anti-diabetic activity. A novel polysaccharide- iron(III) complex (CYPIC) was synthesized by using crude polysaccharide extracted from Dioscorea opposita Thunb. The component, structure, morphology and molecular weights of CYPIC were analysed, and the anti-anemia, acute toxicity and nonspecific immune regulating activities of CYPIC were assayed. The results showed that CYPIC could increase red blood cell count (RBC), hemoglobin (Hb), hematocrit (HCT), thymus and spleen index of mice with iron deficiency anemia (IDA). Although the structure and deeper mechanisms of CYPIC should be further studied, CYPIC has the potential to be used as an iron supplement for the treatment of iron deficiency anemia. CONCLUSION: The large scale industrial production was suggested due to the simple preparation processing of CYPIC.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Dioscorea/chemistry , Ferric Compounds/pharmacology , Polysaccharides/pharmacology , Anemia, Iron-Deficiency/immunology , Animals , Female , Ferric Compounds/chemistry , Ferric Compounds/immunology , Male , Mice , Mice, Inbred Strains , Polysaccharides/chemistry , Polysaccharides/immunology , Rats
20.
J Formos Med Assoc ; 116(3): 145-152, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27825814

ABSTRACT

BACKGROUND/PURPOSE: Serum anti-gastric parietal cell (GPCA), anti-thyroglobulin (TGA), and anti-thyroid microsomal antibodies (TMA) can be found in some recurrent aphthous stomatitis (RAS) patients. This study mainly assessed whether serum GPCA, TGA, TMA and RAS itself played significant roles in causing anemia and hematinic deficiencies in TGA/TMA-positive RAS patients with GPCA positivity (GPCA+/TGA/TMA/RAS patients) or negativity (GPCA-/TGA/TMA/RAS patients). METHODS: The mean corpuscular volume (MCV) and mean blood hemoglobin (Hb), iron, vitamin B12, and folic acid levels were measured and compared between any two of the four groups of 15 GPCA+/TGA/TMA/RAS patients, 69 GPCA-/TGA/TMA/RAS patients, 240 all autoantibodies-negative RAS patients (Abs-/RAS patients), and 342 healthy control subjects. RESULTS: GPCA+/TGA/TMA/RAS patients had significantly lower mean Hb (for men only) and vitamin B12 levels as well as significantly greater frequencies of Hb, iron, and vitamin B12 deficiencies than healthy control subjects. GPCA+/TGA/TMA/RAS patients had lower serum vitamin B12 level and higher MCV as well as a significantly greater frequency of vitamin B12 deficiency than GPCA-/TGA/TMA/RAS patients. Furthermore, both GPCA-/TGA/TMA/RAS and Abs-/RAS patients did have significantly lower mean Hb, MCV, and iron levels as well as significantly greater frequencies of Hb, iron and vitamin B12 deficiencies than healthy control subjects. There were no significant differences in blood data between GPCA-/TGA/TMA/RAS and Abs-/RAS patients CONCLUSION: Both serum GPCA positivity and RAS itself are the contributing factors causing anemia and hematinic deficiencies in GPCA+/TGA/TMA/RAS patients. RAS itself but not TGA/TMA positivity plays a significant role in causing anemia and hematinic deficiencies in GPCA-/TGA/TMA/RAS patients.


Subject(s)
Anemia, Iron-Deficiency/complications , Anemia, Pernicious/complications , Folic Acid Deficiency/blood , Stomatitis, Aphthous/blood , Adult , Aged , Aged, 80 and over , Anemia, Iron-Deficiency/immunology , Anemia, Pernicious/immunology , Autoantibodies/blood , Case-Control Studies , Erythrocyte Indices , Female , Folic Acid/blood , Folic Acid Deficiency/immunology , Hemoglobins/analysis , Humans , Iron/blood , Male , Middle Aged , Parietal Cells, Gastric/immunology , Stomatitis, Aphthous/immunology , Taiwan , Vitamin B 12/blood
SELECTION OF CITATIONS
SEARCH DETAIL