Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
J Perinatol ; 37(4): 436-440, 2017 04.
Article in English | MEDLINE | ID: mdl-27977019

ABSTRACT

OBJECTIVE: Small-for-gestational-age (SGA) neonates, infants of diabetic mothers (IDM) and very-low-birth weight premature neonates (VLBW) are reported to have increased risk for developing iron deficiency and possibly associated neurocognitive delays. STUDY DESIGN: We conducted a pilot study to assess iron status at birth in at-risk neonates by measuring iron parameters in umbilical cord blood from SGA, IDM, VLBW and comparison neonates. RESULTS: Six of the 50 infants studied had biochemical evidence of iron deficiency at birth. Laboratory findings consistent with iron deficiency were found in one SGA, one IDM, three VLBW, and one comparison infant. None of the infants had evidence of iron deficiency anemia. CONCLUSIONS: Evidence of biochemical iron deficiency at birth was found in 17% of screened neonates. Studies are needed to determine whether these infants are at risk for developing iron-limited erythropoiesis, iron deficiency anemia or iron-deficient neurocognitive delay.


Subject(s)
Anemia, Iron-Deficiency/blood , Infant, Small for Gestational Age/blood , Infant, Very Low Birth Weight/blood , Iron/blood , Case-Control Studies , Diabetes, Gestational , Female , Ferritins/blood , Fetal Blood/chemistry , Humans , Infant, Newborn , Linear Models , Male , Pilot Projects , Pregnancy , Pregnancy in Diabetics , Prospective Studies , Risk Factors , Utah
2.
J Perinatol ; 28(8): 580-3, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18668110

ABSTRACT

We describe three preterm neonates with bronchopulmonary dysplasia, pulmonary hypertension and ventricular hypertrophy who incurred subendocardial infarctions, as evidenced by electrocardiographic, laboratory or autopsy findings. We propose that cardiac hypertrophy contributed to the risk for subendocardial ischemia and infarction, and suggest diligence for this complication.


Subject(s)
Bronchopulmonary Dysplasia/complications , Hypertension, Pulmonary/complications , Myocardial Infarction/complications , Fatal Outcome , Female , Humans , Hypertrophy, Left Ventricular/complications , Hypertrophy, Right Ventricular/complications , Infant , Infant, Extremely Low Birth Weight , Infant, Newborn , Infant, Premature , Male
3.
Am J Physiol Gastrointest Liver Physiol ; 292(1): G323-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16935854

ABSTRACT

Hereditary hemochromatosis type 3 is an iron (Fe)-overload disorder caused by mutations in transferrin receptor 2 (TfR2). TfR2 is expressed highly in the liver and regulates Fe metabolism. The aim of this study was to investigate duodenal Fe absorption and hepatic Fe uptake in a TfR2 (Y245X) mutant mouse model of hereditary hemochromatosis type 3. Duodenal Fe absorption and hepatic Fe uptake were measured in vivo by 59Fe-labeled ascorbate in TfR2 mutant mice, wild-type mice, and Fe-loaded wild-type mice (2% dietary carbonyl Fe). Gene expression was measured by real-time RT-PCR. Liver nonheme Fe concentration increased progressively with age in TfR2 mutant mice compared with wild-type mice. Fe absorption (both duodenal Fe uptake and transfer) was increased in TfR2 mutant mice compared with wild-type mice. Likewise, expression of genes participating in duodenal Fe uptake (Dcytb, DMT1) and transfer (ferroportin) were increased in TfR2 mutant mice. Nearly all of the absorbed Fe was taken up rapidly by the liver. Despite hepatic Fe loading, hepcidin expression was decreased in TfR2 mutant mice compared with wild-type mice. Even when compared with Fe-loaded wild-type mice, TfR2 mutant mice had increased Fe absorption, increased duodenal Fe transport gene expression, increased liver Fe uptake, and decreased liver hepcidin expression. In conclusion, despite systemic Fe loading, Fe absorption and liver Fe uptake were increased in TfR2 mutant mice in association with decreased expression of hepcidin. These findings support a model in which TfR2 is a sensor of Fe status and regulates duodenal Fe absorption and liver Fe uptake.


Subject(s)
Duodenum/metabolism , Hemochromatosis/genetics , Intestinal Absorption , Iron/metabolism , Liver/metabolism , Receptors, Transferrin/genetics , Animals , Base Sequence , Biological Transport , Crosses, Genetic , DNA Primers , Disease Models, Animal , Female , Ferritins/metabolism , Genetic Carrier Screening , Hemochromatosis/metabolism , Iron/blood , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains
4.
Gastroenterology ; 121(6): 1489-96, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11729128

ABSTRACT

Hereditary hemochromatosis, a disease of iron overload, occurs in about 1 in 200-400 Caucasians. The gene mutated in this disorder is termed HFE. The product of this gene, HFE protein, is homologous to major histocompatibility complex class I proteins, but HFE does not present peptides to T cells. Based on recent structural, biochemical, and cell biological studies, transferrin receptor (TfR) is a ligand for HFE. This association directly links HFE protein to the TfR-mediated regulation of iron homeostasis. Although evidence is accumulating that binding of HFE to TfR is critical for the effects of HFE, the final pieces in the HFE puzzle have not been established. This review focuses on recent advances in HFE research and presents a hypothetical model of HFE function.


Subject(s)
HLA Antigens/genetics , Histocompatibility Antigens Class I/genetics , Iron/pharmacokinetics , Membrane Proteins , Mutation/physiology , Absorption , Animals , HLA Antigens/physiology , Hemochromatosis Protein , Histocompatibility Antigens Class I/physiology , Homeostasis , Humans , Iron/metabolism , Iron Overload/genetics
7.
Proc Natl Acad Sci U S A ; 98(9): 5170-4, 2001 Apr 24.
Article in English | MEDLINE | ID: mdl-11309500

ABSTRACT

Hereditary hemochromatosis (HH) is a common chronic human genetic disorder whose hallmark is systemic iron overload. Homozygosity for a mutation in the MHC class I heavy chain paralogue gene HFE has been found to be a primary cause of HH. However, many individuals homozygous for the defective allele of HFE do not develop iron overload, raising the possibility that genetic variation in modifier loci contributes to the HH phenotype. Mice deficient in the product of the beta(2)-microglobulin (beta(2)M) class I light chain fail to express HFE and other MHC class I family proteins, and they have been found to manifest many characteristics of the HH phenotype. To determine whether natural genetic variation plays a role in controlling iron overload, we performed classical genetic analysis of the iron-loading phenotype in beta(2)M-deficient mice in the context of different genetic backgrounds. Strain background was found to be a major determinant in iron loading. Sex played a role that was less than that of strain background but still significant. Resistance and susceptibility to iron overload segregated as complex genetic traits in F(1) and back-cross progeny. These results suggest the existence of naturally variant autosomal and Y chromosome-linked modifier loci that, in the context of mice genetically predisposed by virtue of a beta(2)M deficiency, can profoundly influence the severity of iron loading. These results thus provide a genetic explanation for some of the variability of the HH phenotype.


Subject(s)
Genetic Linkage/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Iron Overload/genetics , Iron Overload/pathology , Membrane Proteins , beta 2-Microglobulin/deficiency , Aging , Animals , Crosses, Genetic , Female , HLA Antigens/genetics , Heme/metabolism , Hemochromatosis/genetics , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Iron/metabolism , Iron Overload/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred Strains , Phenotype , Sex Characteristics , Sex Chromosomes/genetics , beta 2-Microglobulin/genetics
8.
Proc Natl Acad Sci U S A ; 98(5): 2707-11, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226304

ABSTRACT

Hereditary hemochromatosis (HH) is a common disorder of iron metabolism caused by mutation in HFE, a gene encoding an MHC class I-like protein. Clinical studies demonstrate that the severity of iron loading is highly variable among individuals with identical HFE genotypes. To determine whether genetic factors other than Hfe genotype influence the severity of iron loading in the murine model of HH, we bred the disrupted murine Hfe allele onto three different genetically defined mouse strains (AKR, C57BL/6, and C3H), which differ in basal iron status and sensitivity to dietary iron loading. Serum transferrin saturations (percent saturation of serum transferrin with iron), hepatic and splenic iron concentrations, and hepatocellular iron distribution patterns were compared for wild-type (Hfe +/+), heterozygote (Hfe +/-), and knockout (Hfe -/-) mice from each strain. Although the Hfe -/- mice from all three strains demonstrated increased transferrin saturations and liver iron concentrations compared with Hfe +/+ mice, strain differences in severity of iron accumulation were striking. Targeted disruption of the Hfe gene led to hepatic iron levels in Hfe -/- AKR mice that were 2.5 or 3.6 times higher than those of Hfe -/- C3H or Hfe -/- C57BL/6 mice, respectively. The Hfe -/- mice also demonstrated strain-dependent differences in transferrin saturation, with the highest values in AKR mice and the lowest values in C3H mice. These observations demonstrate that heritable factors markedly influence iron homeostasis in response to Hfe disruption. Analysis of mice from crosses between C57BL/6 and AKR mice should allow the mapping and subsequent identification of genes modifying the severity of iron loading in this murine model of HH.


Subject(s)
Hemochromatosis/metabolism , Iron/metabolism , Animals , Disease Models, Animal , Hemochromatosis/genetics , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred AKR , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Species Specificity , Spleen/metabolism , Spleen/pathology , Transferrin/metabolism
9.
Haematologica ; 85(4): 340-5, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10756356

ABSTRACT

BACKGROUND AND OBJECTIVE: Most patients with hereditary hemochromatosis are homozygous for a Cys282AETyr mutation in the HFE gene. This mutation has been shown to impair the association of the HFE gene product with b(2)-microglobulin and to prevent its cell surface presentation in transfected COS-7 and 293 cells. This study was performed to examine the expression of HFE protein in epithelial cells, macrophages, and circulating leukocytes obtained from normal subjects and patients with hereditary hemochromatosis. DESIGN AND METHODS: Antisera against two different peptides of the HFE protein were used to immunostain tissue sections and isolate granulocytes, lymphocytes and monocytes. RESULTS: Immunocytochemical staining showed that the HFE protein is expressed in gastric epithelial cells, tissue macrophages, and circulating monocytes and granulocytes. The cell surface associated signal, which was seen in normal gastric epithelial cells, monocytes and macrophages, was also present in C282Y mutant cells from patients with hereditary hemochromatosis, although at apparently reduced amounts in these cells. INTERPRETATION AND CONCLUSIONS: From these studies, it is clear that the C282Y mutation reduces but does not completely prevent presentation of the HFE protein on the cell surface of human monocytes, tissue macrophages, and gastric epithelial cells.


Subject(s)
HLA Antigens/biosynthesis , Histocompatibility Antigens Class I/biosynthesis , Membrane Proteins/metabolism , Amino Acid Substitution , Epithelial Cells/chemistry , Epithelial Cells/pathology , Genes, MHC Class I , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hemochromatosis/pathology , Hemochromatosis Protein , Humans , Immunohistochemistry , Macrophages/chemistry , Macrophages/pathology , Monocytes/chemistry , Monocytes/pathology , Point Mutation , Staining and Labeling , Stomach/pathology
10.
Proc Natl Acad Sci U S A ; 97(4): 1677-82, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10677517

ABSTRACT

A cDNA for a second mouse mitochondrial carbonic anhydrase (CA) called CA VB was identified by homology to the previously characterized murine CA V, now called CA VA. The full-length cDNA encodes a 317-aa precursor that contains a 33-aa classical mitochondrial leader sequence. Comparison of products expressed from cDNAs for murine CA VB and CA VA in COS cells revealed that both expressed active CAs that localized in mitochondria, and showed comparable activities in crude extracts and in mitochondria isolated from transfected COS cells. Northern blot analyses of total RNAs from mouse tissues and Western blot analyses of mouse tissue homogenates showed differences in tissue-specific expression between CA VB and CA VA. CA VB was readily detected in most tissues, while CA VA expression was limited to liver, skeletal muscle, and kidney. The human orthologue of murine CA VB was recently reported also. Comparison of the CA domain sequence of human CA VB with that reported here shows that the CA domains of CA VB are much more highly conserved between mouse and human (95% identity) than the CA domains of mouse and human CA VAs (78% identity). Analysis of phylogenetic relationships between these and other available human and mouse CA isozyme sequences revealed that mammalian CA VB evolved much more slowly than CA VA, accepting amino acid substitutions at least 4.5 times more slowly since each evolved from its respective human-mouse ancestral gene around 90 million years ago. Both the differences in tissue distribution and the much greater evolutionary constraints on CA VB sequences suggest that CA VB and CA VA have evolved to assume different physiological roles.


Subject(s)
Carbonic Anhydrases/metabolism , Mitochondria/enzymology , Amino Acid Sequence , Animals , COS Cells , Carbonic Anhydrases/genetics , Cloning, Molecular , Cytosol/enzymology , Evolution, Molecular , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Phylogeny , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Transfection
11.
Proc Natl Acad Sci U S A ; 97(5): 2214-9, 2000 Feb 29.
Article in English | MEDLINE | ID: mdl-10681454

ABSTRACT

Hereditary hemochromatosis (HH) is a common autosomal recessive disorder characterized by excess absorption of dietary iron and progressive iron deposition in several tissues, particularly liver. Liver disease resulting from iron toxicity is the major cause of death in HH. Hepatic iron loading in HH is progressive despite down-regulation of the classical transferrin receptor (TfR). Recently a human cDNA highly homologous to TfR was identified and reported to encode a protein (TfR2) that binds holotransferrin and mediates uptake of transferrin-bound iron. We independently identified a full-length murine EST encoding the mouse orthologue of the human TfR2. Although homologous to murine TfR in the coding region, the TfR2 transcript does not contain the iron-responsive elements found in the 3' untranslated sequence of TfR mRNA. To determine the potential role for TfR2 in iron uptake by liver, we investigated TfR and TfR2 expression in normal mice and murine models of dietary iron overload (2% carbonyl iron), dietary iron deficiency (gastric parietal cell ablation), and HH (HFE -/-). Northern blot analyses demonstrated distinct tissue-specific patterns of expression for TfR and TfR2, with TfR2 expressed highly only in liver where TfR expression is low. In situ hybridization demonstrated abundant TfR2 expression in hepatocytes. In contrast to TfR, TfR2 expression in liver was not increased in iron deficiency. Furthermore, hepatic expression of TfR2 was not down-regulated with dietary iron loading or in the HFE -/- model of HH. From these observations, we propose that TfR2 allows continued uptake of Tf-bound iron by hepatocytes even after TfR has been down-regulated by iron overload, and this uptake contributes to the susceptibility of liver to iron loading in HH.


Subject(s)
Hemochromatosis/metabolism , Liver/metabolism , Receptors, Transferrin/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , Disease Models, Animal , Gene Expression , Hemochromatosis/genetics , Humans , Iron/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Sequence Homology, Amino Acid , Tissue Distribution
12.
Biol Reprod ; 61(6): 1521-6, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10569998

ABSTRACT

Carbonic anhydrase (CA) is implicated in the acidification of epididymal fluid and thereby in the regulation of sperm maturation and motility. Among the CA isoenzymes, CA IV and II have been shown to be present in the rat epididymal duct epithelium. In the present study, we examined the expression and androgen regulation of CA IV and II mRNAs along the epididymal duct. Northern blot analysis revealed the presence of CA II mRNA in all regions of the epididymis with the strongest signal in the corpus region, while CA IV mRNA was expressed predominantly in the corpus epididymidis. Three days after bilateral castration, CA IV and II mRNAs were decreased by 80-90% in the corpus epididymidis. Testosterone (T) replacement maintained the expression of CA mRNAs at 50-60% of the control levels, indicating that circulating androgens alone are not sufficient to recover the CA expression in the corpus region. However, unilateral castration did not affect the mRNA levels of CA IV and II, suggesting that factors in testicular fluid do not play a major role in the regulation of CA expression in the corpus epididymidis. Immunoblot analysis showed that CA IV protein levels decreased 3 days after castration, while T administration maintained the protein expression virtually at the precastration levels. These data demonstrate that mRNAs for CA IV and II are predominantly expressed in the corpus region of the rat epididymis and can be regulated by androgens in that region. The present data suggest that the regulation of CA expression in the corpus epididymidis by androgens contributes to the known androgen effects on epididymal acidification.


Subject(s)
Androgens/pharmacology , Carbonic Anhydrases/genetics , Epididymis/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Isoenzymes/genetics , Animals , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
13.
Proc Natl Acad Sci U S A ; 96(6): 3143-8, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077651

ABSTRACT

Hereditary hemochromatosis (HH) is a common autosomal recessive disorder characterized by tissue iron deposition secondary to excessive dietary iron absorption. We recently reported that HFE, the protein defective in HH, was physically associated with the transferrin receptor (TfR) in duodenal crypt cells and proposed that mutations in HFE attenuate the uptake of transferrin-bound iron from plasma by duodenal crypt cells, leading to up-regulation of transporters for dietary iron. Here, we tested the hypothesis that HFE-/- mice have increased duodenal expression of the divalent metal transporter (DMT1). By 4 weeks of age, the HFE-/- mice demonstrated iron loading when compared with HFE+/+ littermates, with elevated transferrin saturations (68.4% vs. 49.8%) and elevated liver iron concentrations (985 micrograms vs. 381 micrograms). By using Northern blot analyses, we quantitated duodenal expression of both classes of DMT1 transcripts: one containing an iron responsive element (IRE), called DMT1(IRE), and one containing no IRE, called DMT1(non-IRE). The positive control for DMT1 up-regulation was a murine model of dietary iron deficiency that demonstrated greatly increased levels of duodenal DMT1(IRE) mRNA. HFE-/- mice also demonstrated an increase in duodenal DMT1(IRE) mRNA (average 7.7-fold), despite their elevated transferrin saturation and hepatic iron content. Duodenal expression of DMT1(non-IRE) was not increased, nor was hepatic expression of DMT1 increased. These data support the model for HH in which HFE mutations lead to inappropriately low crypt cell iron, with resultant stabilization of DMT1(IRE) mRNA, up-regulation of DMT1, and increased absorption of dietary iron.


Subject(s)
Carrier Proteins/biosynthesis , Cation Transport Proteins , Duodenum/metabolism , Hemochromatosis/metabolism , Iron-Binding Proteins , Iron/metabolism , Animals , Carrier Proteins/genetics , Hemochromatosis/genetics , Mice , Mutation
14.
Proc Natl Acad Sci U S A ; 96(4): 1579-84, 1999 Feb 16.
Article in English | MEDLINE | ID: mdl-9990067

ABSTRACT

In hereditary hemochromatosis (HH), intestinal absorption of dietary iron is increased, leading to excessive iron accumulation in tissues and resultant organ damage. The HFE protein, which is defective in HH, normally is expressed in crypt enterocytes of the duodenum where it has a unique, predominantly intracellular localization. In placenta, the HFE protein colocalizes with and forms a stable association with the transferrin receptor (TfR), providing a link between the HFE protein and iron transport. In the present study, we examined the relationship of the HFE protein to the TfR in enterocytes of the human duodenum and measured the uptake of transferrin-bound iron and ionic iron by isolated crypt and villus enterocytes. Immunocytochemistry showed that the HFE protein and TfR both are expressed in the crypt enterocytes. Western blots showed that, as was the case in human placenta, the HFE protein in crypt enterocytes is physically associated with the TfR and with beta2-microglobulin. The crypt cell fraction exhibited dramatically higher transferrin-bound iron uptake than villus cells. On the other hand, the villus cells showed 2-3 times higher uptake of ionic iron than crypt cells. We propose that the HFE protein modulates the uptake of transferrin-bound iron from plasma by crypt enterocytes and participates in the mechanism by which the crypt enterocytes sense the level of body iron stores. Impairment of this function caused by HFE gene mutations in HH could provide a paradoxical signal in crypt enterocytes that programs the differentiating enterocytes to absorb more dietary iron when they mature into villus enterocytes.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Intestinal Mucosa/metabolism , Membrane Proteins , Receptors, Transferrin/metabolism , Biological Transport , Duodenum , HLA Antigens/analysis , HLA Antigens/genetics , Hemochromatosis , Hemochromatosis Protein , Histocompatibility Antigens Class I/analysis , Histocompatibility Antigens Class I/genetics , Humans , Immunohistochemistry , Intestinal Mucosa/cytology , Iron/metabolism , Iron Radioisotopes , Receptors, Transferrin/analysis , beta 2-Microglobulin/analysis , beta 2-Microglobulin/metabolism
15.
Proc Natl Acad Sci U S A ; 95(5): 2492-7, 1998 Mar 03.
Article in English | MEDLINE | ID: mdl-9482913

ABSTRACT

Hereditary hemochromatosis (HH) is a common autosomal recessive disease characterized by increased iron absorption and progressive iron storage that results in damage to major organs in the body. Recently, a candidate gene for HH called HFE encoding a major histocompatibility complex class I-like protein was identified by positional cloning. Nearly 90% of Caucasian HH patients have been found to be homozygous for the same mutation (C282Y) in the HFE gene. To test the hypothesis that the HFE gene is involved in regulation of iron homeostasis, we studied the effects of a targeted disruption of the murine homologue of the HFE gene. The HFE-deficient mice showed profound differences in parameters of iron homeostasis. Even on a standard diet, by 10 weeks of age, fasting transferrin saturation was significantly elevated compared with normal littermates (96 +/- 5% vs. 77 +/- 3%, P < 0.007), and hepatic iron concentration was 8-fold higher than that of wild-type littermates (2,071 +/- 450 vs. 255 +/- 23 microg/g dry wt, P < 0.002). Stainable hepatic iron in the HFE mutant mice was predominantly in hepatocytes in a periportal distribution. Iron concentrations in spleen, heart, and kidney were not significantly different. Erythroid parameters were normal, indicating that the anemia did not contribute to the increased iron storage. This study shows that the HFE protein is involved in the regulation of iron homeostasis and that mutations in this gene are responsible for HH. The knockout mouse model of HH will facilitate investigation into the pathogenesis of increased iron accumulation in HH and provide opportunities to evaluate therapeutic strategies for prevention or correction of iron overload.


Subject(s)
HLA Antigens/genetics , HLA Antigens/physiology , Hemochromatosis/genetics , Hemochromatosis/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/physiology , Major Histocompatibility Complex , Membrane Proteins , Animals , Genes, Recessive , Hemochromatosis/blood , Hemochromatosis Protein , Humans , Iron/metabolism , Kidney/immunology , Kidney/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred Strains , Mice, Knockout , Spleen/immunology , Spleen/metabolism , Transferrin/metabolism
16.
J Pediatr ; 132(3 Pt 1): 535-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9544917

ABSTRACT

A 930 gm premature infant had Staphylococcal endocarditis with a tricuspid valvular vegetation that was unresponsive to antibiotics and not amenable to resection. Infusion of tissue plasminogen activator over a 3-day period completely lysed the vegetation. The infection cleared with continued antibiotics, and the infant recovered without sequelae.


Subject(s)
Endocarditis, Bacterial/drug therapy , Plasminogen Activators/therapeutic use , Staphylococcal Infections/drug therapy , Tissue Plasminogen Activator/therapeutic use , Anti-Bacterial Agents/therapeutic use , Echocardiography , Humans , Infant, Newborn , Male
17.
Proc Natl Acad Sci U S A ; 94(24): 13198-202, 1997 Nov 25.
Article in English | MEDLINE | ID: mdl-9371823

ABSTRACT

Hereditary hemochromatosis (HH) is a common autosomal recessive disease associated with loss of regulation of dietary iron absorption and excessive iron deposition in major organs of the body. Recently, a candidate gene for HH (also called HFE) was identified that encodes a novel MHC class I-like protein. Most patients with HH are homozygous for the same mutation in the HFE gene, resulting in a C282Y change in the HFE protein. Studies in cultured cells show that the C282Y mutation abrogates the binding of the recombinant HFE protein to beta2-microglobulin (beta2M) and disrupts its transport to the cell surface. The HFE protein was shown by immunohistochemistry to be expressed in certain epithelial cells throughout the human alimentary tract and to have a unique localization in the cryptal cells of small intestine, where signals to regulate iron absorption are received from the body. In the studies presented here, we demonstrate by immunohistochemistry that the HFE protein is expressed in human placenta in the apical plasma membrane of the syncytiotrophoblasts, where the transferrin-bound iron is normally transported to the fetus via receptor-mediated endocytosis. Western blot analyses show that the HFE protein is associated with beta2M in placental membranes. Unexpectedly, the transferrin receptor was also found to be associated with the HFE protein/beta2M complex. These studies place the normal HFE protein at the site of contact with the maternal circulation where its association with transferrin receptor raises the possibility that the HFE protein plays some role in determining maternal/fetal iron homeostasis. These findings also raise the question of whether mutations in the HFE gene can disrupt this association and thereby contribute to some forms of neonatal iron overload.


Subject(s)
HLA Antigens/metabolism , Hemochromatosis/metabolism , Histocompatibility Antigens Class I/metabolism , Membrane Proteins , Placenta/metabolism , Receptors, Transferrin/metabolism , Chromatography, Affinity/methods , Female , HLA Antigens/genetics , HLA Antigens/immunology , Hemochromatosis/genetics , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunohistochemistry , Precipitin Tests , Pregnancy , Protein Binding , beta 2-Microglobulin/immunology
18.
Cytogenet Cell Genet ; 79(3-4): 176-8, 1997.
Article in English | MEDLINE | ID: mdl-9605844

ABSTRACT

We have mapped 11 novel, anonymous genetic markers to rat chromosome 2. The rat ceruloplasmin gene (Cp) had been previously mapped to chromosomes 2 and 7q11-->q13 by two different methods. To resolve the assignment and to localize the Cp gene on the rat genetic linkage map, we used linkage analysis to confirm that rat Cp lies on chromosome 2.


Subject(s)
Ceruloplasmin/genetics , Genetic Linkage , Genetic Markers , Rats/genetics , Animals , Female , Male , Rats, Inbred F344 , Rats, Wistar
19.
J Clin Invest ; 96(6): 2907-13, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8675662

ABSTRACT

Carbonic anhydrase IV (CA IV) is a glycosylphosphatidylinositol-linked isozyme previously identified on the surface of renal tubular epithelium and certain populations of vascular endothelium. This report identifies the regional, cellular, and subcellular localization of CA IV in the rat gut. Northern blot and RT-PCR analyses demonstrated little CA IV expression in stomach or proximal small intestine, but abundant expression in distal small and large intestine. In contrast, CA II mRNA was abundant in stomach, decreased in proximal small intestine, low in distal small intestine, and abundant in large intestine. CA I mRNA was detected only in large intestine. The regional distribution of CA IV activity correlated with distribution of CA IV mRNA. Immunohistochemistry localized CA IV to the apical plasma membrane of the mucosal epithelium in distal small intestine and large intestine. Signal intensity was greatest in colon. CA IV was additionally found in submucosal capillary endothelium of all gastrointestinal regions. Immunohistochemical findings in human stomach and colon paralleled those in the rat. These studies demonstrate pre-translational isozyme-specific regulation of CA expression along the cranial-caudal axis of the gastrointestinal tract. The regional, cellular, and subcellular localizations are consistent with participation of CA IV in the extensive ion and fluid transport in the distal small and large intestine.


Subject(s)
Carbonic Anhydrases/biosynthesis , Digestive System/enzymology , Gastric Mucosa/enzymology , Gene Expression , Intestinal Mucosa/enzymology , Isoenzymes/biosynthesis , Animals , Capillaries , Carbonic Anhydrases/analysis , Colon/enzymology , Endothelium, Vascular , Epithelium/enzymology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Intestinal Mucosa/blood supply , Intestine, Large/enzymology , Intestine, Small/enzymology , Isoenzymes/analysis , Organ Specificity , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Species Specificity , Stomach/enzymology , Subcellular Fractions/enzymology
20.
Am J Respir Cell Mol Biol ; 10(5): 499-505, 1994 May.
Article in English | MEDLINE | ID: mdl-7514010

ABSTRACT

Pulmonary carbonic anhydrase (CA) activity plays important roles in carbon dioxide exchange, fluid secretion, and pH regulation. This study reports the use of molecular and immunologic techniques to characterize expression of the high-activity cytosolic isoenzyme CA II in rat lung tissue. Northern blot analysis of RNA isolated from various rat tissues revealed that the lung is a site of abundant tissue-specific CA II gene expression. The cell type primarily responsible for CA II expression in the lung was identified by immunohistochemistry as the alveolar type II pneumocyte. RNA blot and immunoblot analyses of isolated rat type II cells in culture confirmed CA II expression by this cell type. Little immunoreactive CA I and no CA IV was detected in these cells. Inhibition studies confirmed that the majority of CA activity in isolated type II cells is attributable to CA II. CA II expression was found to continue in these cells beyond 72 h in culture, a timeframe during which these cells had dedifferentiated. The ontologic pattern of CA II expression in the lung was found by RNA blot analysis to be disparate from that of the surfactant-associated proteins. These observations suggest roles for CA II in alveolar pneumocytes independent of (or in addition to) participation in surfactant biology. Such roles may include the regulation of fluid secretion or facilitation of carbon dioxide elimination.


Subject(s)
Carbonic Anhydrases/biosynthesis , Lung/enzymology , Animals , Blotting, Northern , Blotting, Western , Carbonic Anhydrases/genetics , Cells, Cultured , Gene Expression Regulation , Kinetics , Lung/cytology , RNA/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...