Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
Add more filters










Publication year range
2.
J Pharmacol Exp Ther ; 313(1): 121-6, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15640399

ABSTRACT

Bradykinin (BK) is an endogenous vasoactive peptide that promotes vasodilation by stimulating the release of nitric oxide (NO) from endothelial cells via activation of endothelial NO synthase (eNOS). Although the role of BK in modulation of eNOS activity is well understood, its possible effect on eNOS expression remains uncertain. Several studies have demonstrated negative feedback regulation of eNOS by NO. Therefore, we hypothesized that sustained stimulation with BK may down-regulate eNOS expression in endothelial cells. Human coronary endothelial cells were incubated for 24 h with either BK alone or BK plus BK receptor type 1 or type 2 blockers. NO production and eNOS abundance (Western analysis) were determined. In separate experiments, cells were incubated with either an NOS inhibitor alone or in combination with BK. Incubation with BK caused a concentration-dependent rise in NO production and a dose-dependent decline in eNOS protein expression. These effects were abrogated by BK-2 blockade but unaffected by BK-1 blockade. In contrast, NOS inhibitors lowered NO production and raised eNOS abundance in a dose-dependent fashion. The effects of BK on NO production and eNOS expression were abrogated by the NOS inhibitor. Thus, sustained activation of eNOS by BK results in a compensatory down-regulation of eNOS, whereas its sustained inhibition leads to a compensatory up-regulation of eNOS. The observed modulations of eNOS expression are mediated by NO and represent an adaptive physiologic response.


Subject(s)
Arginine/analogs & derivatives , Arginine/pharmacology , Bradykinin/pharmacology , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Nitric Oxide Synthase/biosynthesis , Blotting, Western , Cells, Cultured , Coronary Vessels/cytology , Coronary Vessels/drug effects , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Enzyme Inhibitors/pharmacology , Humans , Nitric Oxide/biosynthesis , Nitric Oxide/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III , Receptor, Bradykinin B1/drug effects , Receptor, Bradykinin B2/drug effects , Up-Regulation/drug effects , omega-N-Methylarginine/pharmacology
3.
Biochem Soc Trans ; 32(Pt 6): 1008-10, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15506949

ABSTRACT

Nramp1 (murine natural resistance-associated macrophage protein 1 gene)/Slc11a1 (solute carrier family 11 member a1 gene) encodes a bivalent-metal/iron transporter that is expressed within late endosomes/lysosomes of macrophages. A functionally null Nramp1 allele that exhibits impaired bivalent cation transport enables excessive growth of intracellular pathogens. Iron is important for many cellular activities, including defence against pathogens; however, redox-active/free iron can participate in Fenton chemistry that generates reactive oxygen species. Using Raw264.7 cells, non-functional for Nramp1, and stable Nramp1 transfectants, we have examined the effects of impaired bivalent cation transport on macrophage function using glutathione depletion as OS (oxidant stress). Our results demonstrate that OS itself is a signal for increasing Nramp1 transcription and that Nramp1 expression protects against OS. We suggest that OS-mediated protection by Nramp1 function may arise from direct removal of redox-active bivalent cations from a cytosolic pool. We show that OS transcriptional responses are probably mediated by the Sp1 transcription factor.


Subject(s)
Cation Transport Proteins/genetics , Actins/genetics , Animals , Cation Transport Proteins/metabolism , Cell Line , Cytosol/metabolism , Humans , Macrophages , Mice , Oxidation-Reduction , Oxidative Stress , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Transfection
4.
Biochem Soc Trans ; 32(Pt 6): 1084-6, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15506972

ABSTRACT

Slc11a1/Nramp1 (solute carrier family 11 member a1/murine natural resistance-associated macrophage protein 1 gene) encodes a divalent cation transporter that resides within lysosomes/late endosomes of macrophages. Nramp1 modulates the cellular distribution of divalent cations in response to cell activation by intracellular pathogens. Nramp1 expression is repressed and activated by the proto-oncogene c-Myc and Miz-1 (c-Myc-interacting zinc finger protein 1) respectively. Here we demonstrate, using a c-Myc mutant (V394D, Val(394)-->Asp) that is incapable of binding Miz-1, that c-Myc repression of Nramp1 transcription is dependent on its interaction with Miz-1. An oligo pull-down assay demonstrates specific binding of recombinant Miz-1 to the Nramp1 Miz-1-binding site or initiator element(s), and Miz-1-dependent c-Myc recruitment.


Subject(s)
Cation Transport Proteins/genetics , Genes, myc , Transcription, Genetic , Base Sequence , Cations, Divalent , Cell Physiological Phenomena , Genetic Vectors , Models, Genetic , Restriction Mapping
5.
J Biol Chem ; 278(20): 17646-54, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12637578

ABSTRACT

Iron chelators inhibit endotoxin-induced NF-kappaB activation in hepatic macrophages (HMs), suggesting a role for the intracellular chelatable pool of iron in NF-kappaB activation. The present study tested this hypothesis. Analysis of Fe(59)-loaded HMs stimulated with lipopolysaccharide (LPS), revealed a previously unreported, transient rise in intracellular low molecular weight (LMW).Fe(59) complex ([LMW.Fe](i)) at /=15 min) and NF-kappaB (>/=30 min) activation. Iron chelators (1,2-dimethyl-3-hydroxypyridin-4-one and N,N'-bis-2-hydroxybenzylethylenediamine-N,N'-diacetic acid) abrogated the [LMW.Fe](i) response and IKK and NF-kappaB activation. The [LMW.Fe](i) response was also observed in tumor necrosis factor alpha (TNFalpha)-stimulated HMs and RAW264.7 cells treated with LPS and interferon-gamma but not in primary rat hepatocytes or myofibroblastic cells exposed to LPS or TNFalpha. Both [LMW.Fe](i) response and IKK activation in LPS-stimulated HMs were inhibited by diphenylene iodonium (nonspecific inhibitor for flavin-containing oxidases), l-N(6)-(1-iminoethyl)lysine (selective iNOS inhibitor), and adenoviral-mediated expression of a dominant negative mutant of Rac1 or Cu,Zn-superoxide dismutase, suggesting the role of (.)NO and O(2)() in mediating the iron signaling. In fact, this inhibition was recapitulated by a cell-permeable scavenger of ONOO(-), 5,10,15,20-tetrakis (4-sulfonatophenyl)porphyrinato iron (III) chloride. Conversely, ONOO(-) alone induced both [LMW.Fe](i) response and IKK activation. Finally, direct addition of ferrous iron to cultured HMs activated IKK and NF-kappaB. These results support a novel signaling role for [LMW.Fe](i) in IKK activation, which appears to be induced by ONOO(-) and selectively operative in macrophages.


Subject(s)
Iron/metabolism , NF-kappa B/metabolism , Signal Transduction , Animals , Cation Transport Proteins/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Chelating Agents/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Hepatocytes/metabolism , I-kappa B Kinase , Immunoblotting , Iodine/pharmacology , Iron/pharmacology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Male , Mice , Onium Compounds/pharmacology , Oxidation-Reduction , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Wistar , Time Factors , Tumor Necrosis Factor-alpha/metabolism
6.
Biochem Soc Trans ; 30(4): 774-7, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12196193

ABSTRACT

The Nramp1 (natural resistance-associated macrophage protein 1) gene modulates the growth of intracellular pathogens and encodes a divalent cation transporter within lysosomes/late endosomes of macrophages. Nramp1 modulates the cytoplasmic iron pool. Wu, Polack and Dalla-Favera [(1999) Science 283, 676-679] showed reciprocal control of H-ferritin and IRP2 by c-Myc, and suggest that c-Myc regulates genes to increase cytoplasmic iron. A role for c-Myc in Nramp1 regulation was evaluated. Co-transfection studies show that c-Myc represses Nramp1 promoter function. Five non-canonical Myc-max binding sites (E-box) identified within the Nramp1 5'-flanking sequence are not responsible for the inhibitory effects of c-Myc on Nramp1 expression. An initiator(s) adjacent to the transcription-initiation site is a candidate for the inhibition observed. Results are consistent with a role for Nramp1 removing iron from the cytosol and antagonizing c-Myc function.


Subject(s)
Cation Transport Proteins/genetics , Gene Expression Regulation , Genes, myc , Promoter Regions, Genetic , Animals , Binding Sites , Mice , Recombinant Proteins/metabolism , Restriction Mapping , Transcription Factors/metabolism , Transfection
7.
Kidney Int ; 60(3): 1083-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11532103

ABSTRACT

BACKGROUND: Abdominal aortic coarctation above the renal arteries leads to severe hypertension (HTN) above the stenotic site. We have recently shown marked up-regulations of endothelial nitric oxide synthase (eNOS) in heart and thoracic aorta and of neuronal NOS (nNOS) in the brain of rats with severe aortic coarctation above the renal arteries. We hypothesize that the presence of severe regional HTN in the face of marked up-regulation of NO system may be partly due to enhanced NO inactivation by reactive oxygen species (ROS) leading to functional NO deficiency. METHODS: Tissue nitrotyrosine (which is the footprint of NO interaction with ROS) was determined by Western blot in sham-operated control and aortic-banded (above renal arteries) rats three weeks postoperatively. Intra-arterial pressure and tissue nitrotyrosine (Western blot) were measured. RESULTS: The banded group showed a marked rise in arterial pressure measured directly through a carotid cannula (203 +/- 9 vs. 131 +/- 2 mm Hg, P < 0.01). Compared with the sham-operated controls, the banded animals exhibited significant increases in nitrotyrosine abundance in the heart, brain, and the aorta segment above the stricture. In contrast, nitrotyrosine abundance was unchanged in the abdominal aorta segment below the stricture wherein blood pressure was not elevated. CONCLUSION: Coarctation-induced HTN is associated with increased nitrotyrosine abundance in all tissues exposed to high arterial pressure, denoting enhanced ROS-mediated inactivation and sequestration of NO in these sites. This can, in part, account for severe regional HTN in this model. The normality of nitrotyrosine abundance in the abdominal aorta wherein blood pressure is not elevated points to the role of baromechanical factors as opposed to circulating humoral factors that were necessarily similar in both segments.


Subject(s)
Aortic Coarctation/complications , Hypertension/metabolism , Nitric Oxide/metabolism , Tyrosine/analogs & derivatives , Animals , Aorta, Abdominal/metabolism , Aorta, Thoracic/metabolism , Baroreflex , Blotting, Western , Brain/metabolism , Disease Models, Animal , Hypertension/etiology , Male , Myocardium/metabolism , Oxidative Stress , Rats , Rats, Sprague-Dawley , Tyrosine/analysis
8.
Eur J Immunol ; 31(7): 2060-70, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11449359

ABSTRACT

Nramp1 controls responses to infection and encodes a biallelic (G169D) macrophage-restricted divalent-cation transporter. Nramp1(D169) is phenotypically null. We demonstrate Nramp1 is implicated in iron regulation in vivo. In spleen, expression is exclusive to Nramp1(G169) strains within the red pulp. By morphometric analysis, the distribution of splenic iron, following systemic overload, correlates with Nramp1 genotype. More iron is located within the red pulp in Nramp1(D169) strains, whereas in Nramp1(G169) strains iron deposits are localized within the marginal-zone metallophilic cells. Nramp1 immunoreactive protein is not present in control brain, but inducible within a hemorrhagic lesion model in Nramp1(G169) strains. Nramp1 protein expression demonstrates an inverse correlation to the presence of iron. Nramp1(G169) strains show no Perl's stain-reactive iron within the lesion. In contrast, Nramp1(D169) strains display iron-staining cells. The process of cellular iron regulation was investigated in vitro in Nramp1(G169) transfectant Raw264.7 macrophages. Greater (30-50%) iron efflux from Nramp1(G169) compared with Nramp1(D169) cells was determined. The extent of Nramp1-dependent iron-release was influenced by bafilomycin A1, and endogenous nitric oxide synthesis, both inhibitors of vacuolar-ATPase. This study demonstrates that Nramp1 regulates macrophage iron handling, and probably facilitates iron release from macrophages undergoing erythrophagocytosis in vivo.


Subject(s)
Carrier Proteins/physiology , Cation Transport Proteins , Iron/metabolism , Macrophages/metabolism , Membrane Proteins/physiology , Alleles , Animals , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Cytoplasm/metabolism , Cytoplasmic Vesicles/metabolism , Erythrocytes/physiology , Homeostasis , Hydrogen-Ion Concentration , Intracranial Hemorrhages/metabolism , Kupffer Cells/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Spleen/metabolism , Transfection
9.
Kidney Int ; 59(2): 654-61, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11168947

ABSTRACT

BACKGROUND: Severe aortic stenosis above the renal arteries leads to a reduction in renal perfusion, increased renin secretion, and elevation of arterial blood pressure above the stenotic site. Nitric oxide (NO) plays an important role in regulation of renal and systemic vascular resistance, renal blood flow, and Na(+) handling. Abdominal aortic banding provides an excellent model for simultaneous testing of the effects of increased and decreased pressure, flow, and shear stress in the same animal. METHODS: We studied protein expressions of endothelial NO synthase (eNOS), inducible NOS (iNOS), and neuroneal NOS (nNOS) isotypes in the renal cortex, renal medulla, heart, brain, and aorta segments above and below the stenosis site three weeks after abdominal aortic banding above the renal arteries. The results were compared with those obtained in the sham-operated controls. NOS isotype proteins were measured by Western blot. RESULTS: Compared with the control group, the banded group showed significant up-regulations of eNOS, iNOS, and nNOS in renal cortex and medulla. Likewise, heart eNOS, brain nNOS, and thoracic aorta eNOS proteins were significantly increased in the banded group. However, eNOS and iNOS expressions were unchanged in the aorta segment below the stenotic site. Likewise, iNOS expression in the heart and thoracic aorta remained unchanged in the banded animals. No significant difference was found in creatinine clearance or urinary protein excretion between the two groups. CONCLUSIONS: These findings clearly demonstrate the up-regulatory action of increased pressure on eNOS expression in the thoracic aorta and heart and of nNOS expression in the brain. These data further show up-regulation of all NOS isotypes in the kidney, which must have helped to mitigate the associated hypoperfusion.


Subject(s)
Aortic Valve Stenosis/physiopathology , Nitric Oxide Synthase/metabolism , Renal Artery/physiopathology , Animals , Aorta, Thoracic/enzymology , Brain/enzymology , Kidney/enzymology , Male , Myocardium/enzymology , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Rats , Rats, Sprague-Dawley
10.
J Leukoc Biol ; 67(4): 501-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10770282

ABSTRACT

Nramp1 (natural resistance-associated macrophage protein) controls resistance to infection by intracellular pathogens in mice. Nramp1 regulates the microenvironment of the invading pathogen by increasing the luminal iron that participates in the Haber-Weiss reaction, producing radicals that attack the pathogen. We have studied the effect of inflammatory stimuli, iron, and sodium nitroprusside on Nramp1 expression in bone marrow macrophages. Investigations show all three up-regulate Nramp1 expression with a parallel increase in immunoreactivity to an amino-terminal antibody and Nramp1 mRNA. Growth rates are reduced in macrophage cell lines expressing Nramp1. This is through a decrease in iron availability, shown by an increase in IRP2 activity and a reciprocal decrease in conventional protein kinase Cbeta-1 expression. We propose that Nramp1 activity may control its own expression via a negative autoregulatory loop that is important for iron homeostasis and maintenance of low cytoplasmic redox active iron levels in the macrophage.


Subject(s)
Autocrine Communication/immunology , Carrier Proteins/immunology , Cation Transport Proteins , Immunity, Innate , Macrophages/immunology , Membrane Proteins/immunology , Animals , Humans , Mice
11.
J Leukoc Biol ; 66(5): 757-62, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10577506

ABSTRACT

Nramp1 (natural resistance-associated macrophage protein one) regulates intracellular pathogen proliferation and macrophage inflammatory responses. Murine Nramp1 exhibits a natural polymorphism with alleles termed resistant and susceptible. Alleles restrict or allow the proliferation of intracellular pathogens, respectively. Structural predictions suggest that Nramp1 encodes the prototypic member of a transporter family. Nramp1 exhibits sequence identity to Nramp2, which regulates intestinal and reticulocyte iron uptake. Based on this sequence identity we have initiated experiments for Nramp1 to investigate its role in macrophage iron homoeostasis and using a transfection approach in the RAW264.7 murine macrophage-like cell line, which lacks a functional Nramp1 gene. Nramp1 expression supports increased acute cytoplasmic influx of iron, detected using the fluorescent iron sensor dye calcein. Analysis of the endogenous iron sensors, iron regulatory protein 1 and 2, reveals a greater flux of iron in Nramp1-expressing cells and in its exclusion from the cytoplasm. Other work supports the prediction that Nramp1 is a phosphoprotein and the extent of phosphorylation changes in response to inflammatory cytokines. Together these data support the hypothesis that control of intracellular iron homoeostasis is a vital element used by phagocytes to control the proliferation of intracellular pathogens.


Subject(s)
Carrier Proteins/physiology , Cation Transport Proteins , Iron/metabolism , Macrophages/immunology , Membrane Proteins/physiology , Amino Acid Sequence , Animals , Biological Transport , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Homeostasis , Humans , Immunity, Innate , Intracellular Fluid , Macrophages/metabolism , Macrophages/microbiology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Molecular Sequence Data
12.
AIDS ; 13(12): 1443-52, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10465066

ABSTRACT

OBJECTIVES: There is now a strong consensus that the neurotoxic properties of HIV-1 are likely to be mediated by an indirect mechanism in which neurones are damaged by infected mononuclear cells. The aim of this study was to determine the ability of HIV-1 Tat to induce neurotoxic properties in a murine macrophage cell line RAW264.7. DESIGN: Simple culture systems using dissociated neurones may not provide the appropriate microenvironment in which to observe the complex cell-cell interactions that occur in the brain. We have therefore developed a more physiological model in which rat organotypic hippocampal slices are co-cultured with the murine macrophage cell line RAW264.7. Effects of Tat were studied by using a stable Tat expressing RAW264.7 cell line or by addition of recombinant Tat protein to co-cultures. METHODS: Organotypic hippocampal slices prepared from 8-10 day rat pups were grown on membrane inserts that were placed into six-well plates on which RAW264.7 cells were growing as an adherent monolayer. Cell death in the slices was assessed using propidium iodide. Specific astrocytic (glial fibrillary acidophilic protein; GFAP) and neuronal (microtubule-associated protein; MAP2) markers were visualized by immunocytochemistry. RESULTS: RAW264.7 cells that either expressed or were exposed to HIV-1 Tat protein, produced a soluble factor that caused profound degeneration in brain slice cultures involving loss of both glial cells and neurones. By contrast treatment of slice cultures with Tat in the absence of RAW264.7 cells was not neurotoxic. CONCLUSIONS: The neurotoxic properties previously attributed to HIV-1 Tat are likely to be mediated via induction of macrophage derived soluble factor(s).


Subject(s)
Gene Products, tat/toxicity , HIV-1/physiology , Macrophages/metabolism , Macrophages/virology , Neurotoxins/metabolism , Animals , Cell Line , Coculture Techniques , Culture Media, Conditioned , Gene Products, tat/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus , Immunohistochemistry , Mice , Microtubule-Associated Proteins/metabolism , Organ Culture Techniques , Rats , Recombinant Proteins/metabolism , tat Gene Products, Human Immunodeficiency Virus
13.
J Mol Biol ; 290(1): 21-35, 1999 Jul 02.
Article in English | MEDLINE | ID: mdl-10388555

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) Nef is important for viral infectivity and pathogenicity. HIV-1 infection is associated with inappropriate activation and defects in the function of monocytes/macrophages. We have studied the effects of HIV-1 Nef in the murine (RAW264.7) and human (THP-1) monocyte-macrophage cell lines. Investigation of the activator protein-1 (AP-1) transcription factor showed that Nef expression induced both its DNA binding and transcriptional activities. Increased AP-1 DNA binding activity in RAW264.7 cells was associated with raised levels of c-Fos expression and induction of mRNA for the AP-1 responsive tissue inhibitor of metalloproteinases-1 (TIMP-1) gene. Mutagenesis and kinase inhibition studies were employed to determine signaling pathways used by Nef to induce AP-1. Data from these studies indicated that induction of AP-1 by Nef is likely to be mediated through the MAPK (ERK1 and 2) signaling pathway and requires the proline-rich PxxP motif of Nef, suggesting the involvement of upstream protein kinases belonging to the Src family. Effects of Nef on AP-1 induction were cell lineage-specific, being stimulatory in macrophages, inhibitory in T cells and without effect in HeLa cells. These latter two observations led us to test the possibility that cell-specific interactions of Nef with Src family proteins may modulate AP-1 activity. To this end we demonstrated that a dominant-negative Hck mutant caused inhibition of Nef-mediated AP-1 DNA binding activity in RAW cells. In conclusion, induction of AP-1 by Nef is a specific feature of human and murine macrophage cell lines that requires signal transduction events involving Hck and MAPKs.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Products, nef/metabolism , HIV-1/metabolism , Macrophages/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factor AP-1/biosynthesis , Base Sequence , Cell Line , DNA Primers , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Genes, fos , Humans , Proto-Oncogene Proteins c-hck , Signal Transduction , Transcription Factor AP-1/metabolism , Transfection , nef Gene Products, Human Immunodeficiency Virus
14.
Immunology ; 96(4): 656-62, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10233755

ABSTRACT

Nramp1 (natural resistance-associated macrophage protein) was positionally cloned as the defective biallelic locus in inbred mouse strains associated with uncontrolled proliferation of obligate intracellular macrophage pathogens. The causative defect was described as G169D within membrane spanning domain 4 of a transporter. The biochemical activity of Nramp1 is implied from sequence conservation with Nramp2. Nramp2 encodes a divalent cation transporter and is the carrier of a defect in models of microcytic anaemia, associated with impaired intestinal iron uptake. Iron sequestration has been proposed as an antimicrobial mechanism. Therefore, such an activity for Nramp1 is consistent with model systems. Here we showed that Nramp1 directs iron transport within the macrophage. We describe stable, high-level Nramp1G169 allele-derived polypeptide expression in Balb/c Nramp1D169 RAW264.7 cells. Transfectants express levels, comparable to those in Nramp1G169-resistant macrophages, of a 90-100x103 MW Nramp1 polypeptide. Expression of the Nramp1 polypeptide correlates with lower cellular iron loads and a reduced chelatable iron pool following challenge with iron: nitrilotriacetate. Pulse chase experiments support an enhanced iron flux in expressing cells. These data are supported using the fluorescent iron probe calcein. In Nramp1G169-expressing cells we observed an increased iron flux into the cytoplasm from a calcein-inaccessible cellular location. These data suggest Nramp1, in resting macrophage cells, mobilizes iron, from an intracellular vesicle, which is destined for cell secretion. We propose that under these conditions Nramp1 plays a role in a salvage pathway of iron recycling.


Subject(s)
Carrier Proteins/metabolism , Cation Transport Proteins , Iron/metabolism , Macrophages, Peritoneal/metabolism , Membrane Proteins/metabolism , Animals , Biological Transport/physiology , Blotting, Western , Carrier Proteins/physiology , Fluoresceins , Indicators and Reagents , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Transfection
15.
Biochim Biophys Acta ; 1436(3): 577-84, 1999 Jan 04.
Article in English | MEDLINE | ID: mdl-9989287

ABSTRACT

Fatty acids are a major source of fuel for energy production by myocytes. Lipoprotein lipase (LPL) and very low density lipoprotein (VLDL) receptor are abundantly expressed by the heart and skeletal muscles. LPL and possibly VLDL receptor represent the primary route of access to fatty acids contained in circulating triglyceride-rich lipoproteins. Physical exercise and thyroid hormone, which promote energy consumption, upregulate LPL expression in skeletal muscles. This study tested the hypothesis that increased cardiac workload might modulate myocardial LPL and/or VLDL receptor expressions. Accordingly, cardiac tissue LPL activity, LPL and VLDL receptor proteins and mRNA abundance were studied in Sprague-Dawley rats 4 weeks after induction of severe thoracic aorta constriction or sham operation. Elevation of afterload with thoracic aortic constriction led to a significant cardiomegaly and a marked upregulation of cardiac LPL activity, LPL mRNA and LPL protein abundance, but did not modify VLDL receptor mRNA or protein abundance. Thus, increased cardiac workload in this model results in upregulation of myocardial LPL expression which can enhance fatty acid availability to accommodate the heart's increased energy requirement.


Subject(s)
Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Myocardium/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Animals , Aorta, Thoracic , Aortic Coarctation/complications , Aortic Coarctation/genetics , Aortic Coarctation/metabolism , Disease Models, Animal , Energy Metabolism , Fatty Acids/metabolism , Hypertrophy, Left Ventricular/etiology , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Up-Regulation
17.
J Cell Sci ; 111 ( Pt 19): 2855-66, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9730978

ABSTRACT

The murine natural resistance-associated macrophage protein, Nramp1, has multiple pleiotropic effects on macrophage activation and regulates survival of intracellular pathogens including Leishmania, Salmonella and Mycobacterium species. Nramp1 acts as an iron transporter, but precisely how this relates to macrophage activation and/or pathogen survival remains unclear. To gain insight into function, anti-Nramp1 monoclonal and polyclonal antibodies are used here to localise Nramp1 following activation and infection. Confocal microscope analysis in uninfected macrophages demonstrates that both the mutant (infection-susceptible) and wild-type (infection-resistant) forms of the protein localise to the membranes of intracellular vesicular compartments. Gold labelling and electron microscopy defines these compartments more precisely as electron-lucent late endosomal and electron-dense lysosomal compartments, with Nramp1 colocalizing with Lamp1 and cathepsins D and L in both compartments, with macrosialin in late endosomes, and with BSA-5 nm gold in pre-loaded lysosomes. Nramp1 is upregulated with interferon-(gamma) and lipopolysaccaride treatment, coinciding with an increase in labelling in lysosomes relative to late endosomes and apparent dispersion of Nramp1-positive vesicles from a perinuclear location towards the periphery of the cytoplasm along the microtubular network. In both control and activated macrophages, expression of the protein is 3- to 4-fold higher in wild-type compared to mutant macrophages. In Leishmania major-infected macrophages, Nramp1 is observed in the membrane of the pathogen-containing phagosomes, which retain a perinuclear localization in resting macrophages. In Mycobacterium avium-infected resting and activated macrophages, Nramp1-positive vesicles migrated to converge, but not always fuse, with pathogen-containing phagosomes. The Nramp1 protein is thus located where it can have a direct influence on phagosome fusion and the microenvironment of the pathogen, as well as in the more general regulation of endosomal/lysosomal function in macrophages.


Subject(s)
Carrier Proteins/immunology , Carrier Proteins/metabolism , Cation Transport Proteins , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/immunology , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Carrier Proteins/genetics , Cell Line , Endosomes/metabolism , Epitope Mapping , Leishmania major/immunology , Leishmania major/pathogenicity , Lysosomes/metabolism , Macrophage Activation , Macrophages/parasitology , Membrane Proteins/genetics , Mice , Microscopy, Confocal , Microscopy, Immunoelectron , Molecular Sequence Data , Mutation , Mycobacterium avium/immunology , Mycobacterium avium/pathogenicity
18.
FEBS Lett ; 425(2): 239-42, 1998 Mar 27.
Article in English | MEDLINE | ID: mdl-9559656

ABSTRACT

Nramp1 (natural resistance-associated macrophage protein) controls innate immunity and encodes a transporter of unknown function. Here we describe an antibody to Nramp1 displaying immunoreactivity towards a mature heavily glycosylated polypeptide of 90-100 kDa and a precursor form of 45 kDa in macrophages. Ectopic expression of the Nramp1 cDNA in COS-1 cells demonstrates that Nramp1 modulates cellular iron levels following loading with low molecular weight iron chelates. Surprisingly, Nramp1 does not enhance iron uptake, but expression is associated with reduced cellular iron loads. We propose Nramp1 may play a role in a salvage pathway of iron recycling.


Subject(s)
Carrier Proteins/metabolism , Cation Transport Proteins , Iron/metabolism , Membrane Proteins/metabolism , Animals , Antibodies/immunology , COS Cells , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line , Gene Expression , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Rabbits
19.
Biochem J ; 325 ( Pt 3): 779-86, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9271100

ABSTRACT

The murine Nramp1 (natural-resistance-associated macrophage protein) locus, formerly known as Ity/Lsh/Bcg, was isolated previously on the basis of chromosomal location, and as conferring natural resistance to infection against intracellular macrophage pathogens. The gene encodes a transporter molecule of unknown function. We have prepared polyclonal antisera against the C-terminal 35 amino acids of murine Nramp1. This serum is reactive towards a 65 kDa protein, expressed in murine macrophage cells from resistant or susceptible mice stimulated with interferon-gamma and lipopolysaccharide, but not in non-macrophage cells. Evidence indicates that Nramp1 is localized in a subcellular membrane rather than at the cell surface. This evidence includes: the identification of conserved endocytic targeting motifs following inspection of human and murine Nramp sequences; the enrichment of Nramp1, following magnetic selection of phagolysosomal vesicles from activated macrophages that were allowed to phagocytose magnetic, IgG-coated beads; confocal microscopy. These studies place Nramp1 on a membrane in close proximity to obligate intracellular pathogens. A link between Nramp1 and divalent-cation transport is suggested by sequence similarity with yeast SMF1. Evidence showing modulation of Nramp1 protein levels by iron chelation provides a direct link with Nramp1 function and divalent-cation metabolism.


Subject(s)
Carrier Proteins/genetics , Cation Transport Proteins , Interferon-gamma/pharmacology , Macrophages/metabolism , Membrane Proteins/genetics , Amino Acid Sequence , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/immunology , Humans , Immune Sera , Iron Chelating Agents/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Mice , Microglia/metabolism , Molecular Sequence Data , Sequence Homology, Amino Acid
20.
J Gen Virol ; 78 ( Pt 2): 381-92, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9018061

ABSTRACT

The nef gene product of both human and simian immunodeficiency viruses is critically important for virus replication and disease progression in vivo. However, the precise biological function of Nef remains poorly characterized in vitro, with previous reports suggesting that Nef might be either cytotoxic or cytostatic. As a result of difficulties encountered by several groups in establishing cell lines constitutively expressing Nef, we have developed two inducible systems resulting in stable Nef expression in various mammalian cell lines. Tetracycline-regulated Nef expression was achieved in HeLa cells but could not be established in human T cell lines. Jurkat E6-1 T cell and RAW264.7 murine macrophage cell lines expressing a regulated nef gene were generated using a system in which Nef expression was controlled by a mutated version of the heavy metal-inducible human metallothionein IIA promoter. Induction of high levels of Nef expression in HeLa-Nef and Jurkat-Nef cells resulted in a moderate (2-fold) and a dramatic (10-fold) retardation of cell growth respectively, supporting the contention that Nef may be a cytotoxic or cytostatic factor. This property was also observed at low basal levels of Nef expression in RAW264.7-Nef macrophage clones (5-fold reduction in growth) and was associated with an altered morphological phenotype suggesting that different cell types may be more susceptible to the cytostatic activity of Nef. The regulated Nef-expression systems provide tools for investigating the molecular basis of Nef function, including Nef-mediated cytopathogenicity, CD4 down-regulation and enhancement of virus infectivity.


Subject(s)
Gene Expression Regulation, Viral , Gene Products, nef/genetics , Genetic Vectors , HIV-1/genetics , Animals , Cell Death , Cell Division , Cell Line , Gene Expression Regulation, Viral/drug effects , Gene Products, nef/physiology , HIV-1/pathogenicity , HeLa Cells , Humans , Jurkat Cells , Macrophages , Metallothionein/genetics , Metals, Heavy/pharmacology , Mice , Tetracycline/pharmacology , nef Gene Products, Human Immunodeficiency Virus
SELECTION OF CITATIONS
SEARCH DETAIL
...