Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Br J Nutr ; 86(2): 157-62, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11502228

ABSTRACT

Iron deficiency induces thymus atrophy in laboratory animals and very likely in humans by unknown mechanisms. The atrophy is associated with impaired cell-mediated immunity. In this study, we tested the hypothesis that thymus atrophy is a result of increased apoptosis and reduced thymocyte proliferation. Thymocytes were obtained from twenty-seven control, twenty-seven pairfed, twenty-seven iron-deficient (ID) mice; twelve and fourteen ID mice that received the control diet (0.9 mmol/kg versus 0.09 mmol/kg for the ID diet) for 1 d (repletion, R1) and 3 d (R3), respectively. Cell cycle analysis and apoptosis were studied by flow cytometry using propidium iodide staining and terminal deoxyuridine nick end labeling of DNA breaks assay respectively. When mice were killed, haemoglobin, haematocrit, and liver iron stores of ID, R1, and R3 mice were 25-40 % of those of control and pairfed mice Absolute and relative thymus weights and thymocyte numbers were 19 to 68 % lower in ID, R1, and R3 than in control and pairfed groups We found no significant difference among groups in the percentage of cells undergoing apoptosis. A higher percentage of thymocytes from ID and R1 mice than those of control, pairfed, and R3 mice were in the resting phase of the normal cell cycle Conversely, a lower percentage of thymocytes from ID and R1 mice than those from control, pairfed, and R3 mice were in the DNA synthesis phase and late phase of DNA synthesis and onset of mitosis (G2-M) Indicators of iron status positively correlated (r 0.3 to 0.56) with the percentage of thymocytes in the G2-M phase Results suggest that reduced cell proliferation but not increased apoptosis is the cause of thymus atrophy associated with iron deficiency.


Subject(s)
Anemia, Iron-Deficiency/pathology , Thymus Gland/pathology , Anemia, Iron-Deficiency/metabolism , Animals , Apoptosis , Atrophy/etiology , Cell Count , Cell Cycle , Disease Models, Animal , Female , Flow Cytometry , Hematocrit , Hemoglobins/analysis , In Situ Nick-End Labeling , Iron/analysis , Liver/chemistry , Mice , Mice, Inbred C57BL
2.
J Nutr ; 131(7): 2028-33, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11435525

ABSTRACT

The influence of iron deficiency on the progression of mitogen-treated splenic lymphocytes through the cell cycle was studied in 16 control, 16 pair-fed, 15 iron-deficient (ID) and 16 ID mice that were repleted for up to 3 d (R3). The test and control diets differed only in iron concentrations (0.09 vs. 0.9 mmol/kg). When mice were killed (68 d of feeding), the hemoglobin concentration and liver iron stores of ID and R3 mice were <50% those of control mice (P < 0.05). Iron deficiency did not reduce the percentage of CD3(+) cells, but decreased CD3(+) cells/mg spleen (P < 0.05). In concanavalin A-treated and nonactivated cultures, there were no significant differences among groups in the percentages of cells in resting phase of the cell cycle (G0) to cell cycle initiation phase (G1), DNA synthesis phase (S) and exit from the S phase (G2) to mitosis phase (M) phases. In anti-CD3 and anti-CD3/anti-CD28-treated cultures, higher percentages of lymphocytes from ID and R3 mice than those from control and pair-fed mice were in the G0--G1 phase (P < 0.05). Conversely, lower percentages of activated cells from ID and R mice than those from control and pair-fed mice were in S and G2--M phases (P < 0.05). Incubation of lymphocytes with mitogens decreased the percentages of cells in G0--G1 phase from 90% to 80% in control and pair-fed but not in ID and R3 mice (P < 0.05). In activated cells, indices of iron status negatively correlated with the percentages of cells in G0--G1 (r = -0.306 to -0.597) but positively with those in S (r = 0.166--0.511) and G2--M phases (r = 0.265-0.59; P < 0.05). Data suggest that altered cell cycle progression likely contributes to impaired lymphocyte proliferation usually associated with iron deficiency.


Subject(s)
Cell Cycle/physiology , Iron Deficiencies , Lymphocyte Activation/physiology , Spleen/cytology , Animals , Cell Cycle/immunology , Cell Division , Cell Physiological Phenomena/drug effects , Cells, Cultured , Concanavalin A/pharmacology , Female , G1 Phase , G2 Phase , Hemoglobins/analysis , Mice , Mice, Inbred C57BL , Mitogens/pharmacology , Mitosis , Resting Phase, Cell Cycle , S Phase , Spleen/immunology
3.
J Cell Biochem ; 74(3): 468-78, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10412047

ABSTRACT

We investigated the effects of iron deficiency anemia, iron repletion, and iron chelation by deferoxamine on protein kinase C (PKC) activity, an enzyme that plays a crucial role on T lymphocyte proliferation. The study involved 23 control (C), 18 pairfed (PF), and 24 iron deficient (ID) mice or ID mice that were repleted for 3 (n = 14), 7 (n = 17), or 14 (n = 14) days. The low iron (0.09 mmol iron/kg) and iron-supplemented (0.9 mmol iron/kg) diets were fed to mice for 53 days. Mean hemoglobin, hematocrit, and liver iron stores of ID mice were one third of those of C mice. Lymphocyte proliferation was reduced (P < 0.05) in spleen and purified T cells in ID but not PF mice. In concanavalin A, phytohemagglutinin, and anti-CD3 antibody-treated and untreated cells that were incubated in serum-free and serum-containing medium, PKC activity was significantly (P < 0.05) reduced in ID but not PF mice and returned to normal before correction of anemia. In mitogen-treated cells, while the ratios of membrane-bound to cytosol activity increased nearly seven-fold (from 0.4-0.63 in resting cells to 1.43-7.23) in spleen cells from C, PF, and repleted mice and 11-fold in T cells (P < 0.005), they remained below 1 in ID mice suggesting reduced translocation. In vitro iron chelation by deferoxamine for 120 min prior to cell activation reduced (P < 0.05) PKC activity by 46-60% in C and PF and 28-53% in ID mice. The data suggest that: 1) it is iron-deficiency but not anemia or differences in the proportion of immunocompetent T cells that reduced PKC activity in cells from ID mice; 2) reduced PKC translocation may play an important role on altered lymphocyte proliferation and associated functions in iron-deficient individuals.


Subject(s)
Anemia, Iron-Deficiency/metabolism , Iron/physiology , Protein Kinase C/metabolism , Spleen/enzymology , T-Lymphocytes/enzymology , Anemia, Iron-Deficiency/drug therapy , Animals , Body Weight , CD3 Complex/immunology , Cell Division , Cell Membrane/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Concanavalin A/pharmacology , Culture Media, Serum-Free/pharmacology , Cytosol/drug effects , Deferoxamine/pharmacology , Female , Hemoglobins/metabolism , Iron/pharmacology , Iron, Dietary/pharmacology , Liver/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mitogens/pharmacology , Phytohemagglutinins/pharmacology , Spleen/drug effects , T-Lymphocytes/drug effects , Time Factors
4.
J Nutr ; 128(7): 1077-83, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9649588

ABSTRACT

Iron deficiency impairs lymphocyte proliferation in humans and laboratory animals by unknown mechanisms. In this study, we investigated whether this alteration can be attributed in part to impaired hydrolysis of cell membrane phosphatidyl inositol-4, 5-bisphosphate (PIP2), a required early event of T-lymphocyte activation. The study involved 46 iron-deficient (ID), 26 control (C) and 23 pair-fed (PF) mice, and ID mice that were repleted for 3 (n = 16), 7 (n = 17) or 14 d (n = 18). Mice were killed after 40-63 d (mean, 48 d) of consuming the test diet (0.09 mmol/kg iron) or the control diet (0.9 mmol/kg). The mean (+/-SEM) hemoglobin concentrations were 57 +/- 16.7, 176 +/- 2.6 and 181 +/- 9.7 g/L for ID, C and PF groups, respectively. After splenic lymphocytes were labeled in vitro with 3H-myoinositol for 3 h, PIP2 hydrolysis was estimated by measuring the radioactivity recovered as a mixture of inositol mono-, di- and triphosphate (IP) from concanavalin A (0, 1, 2.5, 5 and 10 mg/L) activated cells. Although cells from ID mice and those from mice repleted for 3 d incorporated slightly more radioactivity in cellular phospholipids than did cells from C or PF mice, less (P < 0.005) was recovered as IP than in controls, suggesting impaired conversion of the precursor to PIP2. At almost all incubation periods (10-120 min) and mitogen concentrations, the rate of PIP2 hydrolysis expressed as the ratio of radioactivity obtained in Con A-treated to untreated cells was significantly (P < 0.05) reduced in cells from ID mice compared with those obtained from C and PF mice. For cells that were activated for 60 min or less, iron repletion for 14 d significantly (P < 0.05) improved the rate of PIP2 hydrolysis. PIP2 hydrolysis positively and significantly (P < 0.05) correlated (r = 0.27-0.56) with indicators of iron status. Mitogenic response was also significantly (P < 0.05) reduced in ID but not PF mice, and it was corrected by iron repletion for 3, 7 or 14 d. Lymphocyte proliferation positively (r = 0.27-0.37, P < 0.01) correlated with indices of iron status and IP ratios. The data suggest that reduced PIP2 hydrolysis contributes to impaired blastogenesis in iron deficiency.


Subject(s)
Cell Membrane/metabolism , Iron Deficiencies , Lymphocyte Activation , Lymphocytes/ultrastructure , Phosphatidylinositol 4,5-Diphosphate/metabolism , Spleen/cytology , Animals , Concanavalin A/pharmacology , Dietary Supplements , Female , Hydrolysis , Inositol/metabolism , Iron/administration & dosage , Iron/analysis , Kinetics , Liver/chemistry , Lymphocytes/physiology , Mice , Mice, Inbred C57BL
5.
Am J Clin Nutr ; 38(2): 238-44, 1983 Aug.
Article in English | MEDLINE | ID: mdl-6410896

ABSTRACT

The capacity of spleen and peritoneal cells from iron deficient mice, ad libitum fed control mice, and pair-fed mice to kill allogenic tumor cells (mastocytoma tumor P815) has been investigated. In the first study, mice were sensitized in vivo with 10(7) viable tumor cells 51 and 56 days after weaning. The capacity of splenic cells and peritoneal cells from sensitized and nonsensitized mice to kill tumor cells was evaluated 5 days after the second dose of tumor cells. At ratios of 2.5:1 to 100:1 of attacker to target cells, the percentage 51Cr release after 4 h of incubation was significantly less in iron-deficient mice than control and/or pair-fed mice (p less than 0.05). Protein-energy undernutrition in pair-fed mice had no significant effect. In the second study, spleen cells and enriched T cell fractions were incubated in vitro for 5 days with uv irradiated Balb/C spleen cells in a 2:1 ratio. The cytotoxic capacity against the same allogenic tumor cells was again evaluated. The percentage chromium release at different attacker to target cells was less than 30% in the iron-deficient group compared to either control or pair-fed supporting the results of in vivo sensitized cells. The possible mode of impairment of the cytotoxic capacity is discussed.


Subject(s)
Anemia, Hypochromic/immunology , Cytotoxicity, Immunologic , Animals , Ascitic Fluid/cytology , Body Weight , Chromium Radioisotopes , Female , Lymphocytes/immunology , Macrophages/immunology , Mast-Cell Sarcoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Sarcoma, Experimental/immunology , Spleen/cytology , T-Lymphocytes, Cytotoxic/immunology
6.
Am J Clin Nutr ; 37(4): 557-65, 1983 Apr.
Article in English | MEDLINE | ID: mdl-6601454

ABSTRACT

Splenic lymphocytes from iron deficient C57BL/6 mice gave smaller proliferative responses to T and B cell mitogens than those from either the control of pair-fed mice. The addition of hemin to the culture medium partially restored the responses to Con A and phytohemagglutinin but not to bacterial lipopolysaccharide in unfractionated spleen cells and enriched T cell fractions. The responses of lymphocytes from the control and pair-fed mice were either unchanged or decreased. Hemin restored the blastogenic response to Con A more efficiently than to phytohemagglutinin. The blastogenic responses were increased linearly with increasing doses of hemin. Ferric chloride and iron saturated mouse transferrin did not restore the response to either Con A or lipopolysaccharide. However, both transferrin and ferric chloride partially restored the response to phytohemagglutinin. The possible mechanism of selective restoration of blastogenesis by hemin, transferrin, and ferric chloride in iron-deficient T lymphocytes is discussed.


Subject(s)
Ferric Compounds/pharmacology , Heme/analogs & derivatives , Hemin/pharmacology , Iron Deficiencies , Iron/pharmacology , Lymphocyte Activation/drug effects , Transferrin/pharmacology , Animals , Chlorides , Concanavalin A/pharmacology , DNA/biosynthesis , Dose-Response Relationship, Drug , Female , Lymphocytes/physiology , Mice , Mice, Inbred C57BL , Phytohemagglutinins/pharmacology , Spleen/cytology , T-Lymphocytes/physiology
7.
Am J Clin Nutr ; 34(12): 2635-40, 1981 Dec.
Article in English | MEDLINE | ID: mdl-6976117

ABSTRACT

In the present study, the effect of iron-deficiency anemia on delayed cutaneous hypersensitivity was measured using weanling C57BL/6 female mice which were fed either an ad libitum control diet supplemented with 25 to 30 mg Fe/kg diet (FePO4), an iron-deficient test diet (5 to 6 mg Fe/kg diet), or pairfed control diet (25 to 30 mg Fe/kg diet). When skin sensitizing agent (dinitrofluorobenzene) was applied to these animals and skin responses were measured 3 to 5 days later, anemic mice showed a significantly decreased inflammatory skin response than either control or pairfed mice. Five days after sensitization, the animals were challenged with dinitrofluorobenzene painted on the right ear and an equal dose of only the solvent on the left ear followed by 125I-deoxyuridine injected intraperitoneally. The ratio of either total or DNA associated radioactivity incorporated into the right over the left ears was significantly lower in anemic mice than either control or pairfed mice. A single dose of Imferon injected 24 h before the recall dose of dinitrofluorobenzene restored the ratio of 125I-dUR incorporated in anemic mice without having any significant effect on either the control or pairfed groups. The results suggest that iron is not required for sensitization but is required for an effective inflammatory response.


Subject(s)
Anemia, Hypochromic/immunology , Hypersensitivity, Delayed , Animals , DNA/biosynthesis , Deoxyuridine/metabolism , Diet , Dinitrofluorobenzene , Female , Hypersensitivity, Delayed/chemically induced , Immunity, Cellular/drug effects , Iron Deficiencies , Iron-Dextran Complex/pharmacology , Mice , Mice, Inbred C57BL , Skin Tests , T-Lymphocytes/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...