Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Microbiol Spectr ; 9(3): e0048321, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34730437

ABSTRACT

Zinc (Zn) imbalance is a common single-nutrient disorder worldwide, but little is known about the short-term and long-term effects of imbalanced dietary zinc in the intestinal microbiome. Here, 3-week-old C57BL/6 mice were fed diets supplemented with Zn at the doses of 0 (low Zn), 30 (control Zn), 150 (high Zn), and 600 mg/kg of body weight (excess Zn) for 4 weeks (short term) and 8 weeks (long term). The gut bacterial composition at the phyla, genus, and species levels were changed as the result of the imbalanced Zn diet (e.g., Lactobacillus reuteri and Akkermansia muciniphila). Moreover, pathways including carbohydrate, glycan, and nucleotide metabolism were decreased by a short-term low-Zn diet. Valeriate production was suppressed by a long-term low-Zn diet. Pathways such as drug resistance and infectious diseases were upregulated in high- and excess-Zn diets over 4-week and 8-week intervals. Long-term zinc fortification doses, especially at the high-Zn level, suppressed the abundance of short-chain fatty acids (SCFAs)-producing genera as well as the concentrations of metabolites. Finally, Melainabacteria (phylum) and Desulfovibrio sp. strain ABHU2SB (species) were identified to be potential markers for Zn status with high accuracy (area under the curve [AUC], >0.8). Collectively, this study identified significant changes in gut microbial composition and its metabolite concentration in altered Zn-fed mice and the relevant microbial markers for Zn status. IMPORTANCE Zn insufficiency is an essential health problem in developing countries. To prevent the occurrence of zinc deficit, zinc fortification and supplementation are widely used. However, in developed countries, the amounts of Zn consumed often exceed the tolerable upper intake limit. Our results demonstrated that dietary Zn is an essential mediator of microbial community structure and that both Zn deficiency and Zn overdose can generate a dysbiosis in the gut microbiota. Moreover, specific microbial biomarkers of Zn status were identified and correlated with serum Zn level. Our study found that a short-term low-Zn diet (0 mg/kg) and a long-term high-zinc diet (150 mg/kg) had obvious negative effects in a mouse model. Thus, these results indicate that the provision and duration of supplemental Zn should be approached with caution.


Subject(s)
Gastrointestinal Microbiome , Zinc/metabolism , Animal Feed/analysis , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Biomarkers/metabolism , Diet , Fatty Acids, Volatile/metabolism , Male , Mice/metabolism , Mice/microbiology , Mice, Inbred C57BL , Time Factors , Zinc/analysis
2.
Genome Biol Evol ; 12(8): 1277-1301, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32531054

ABSTRACT

Island populations repeatedly evolve extreme body sizes, but the genomic basis of this pattern remains largely unknown. To understand how organisms on islands evolve gigantism, we compared genome-wide patterns of gene expression in Gough Island mice, the largest wild house mice in the world, and mainland mice from the WSB/EiJ wild-derived inbred strain. We used RNA-seq to quantify differential gene expression in three key metabolic organs: gonadal adipose depot, hypothalamus, and liver. Between 4,000 and 8,800 genes were significantly differentially expressed across the evaluated organs, representing between 20% and 50% of detected transcripts, with 20% or more of differentially expressed transcripts in each organ exhibiting expression fold changes of at least 2×. A minimum of 73 candidate genes for extreme size evolution, including Irs1 and Lrp1, were identified by considering differential expression jointly with other data sets: 1) genomic positions of published quantitative trait loci for body weight and growth rate, 2) whole-genome sequencing of 16 wild-caught Gough Island mice that revealed fixed single-nucleotide differences between the strains, and 3) publicly available tissue-specific regulatory elements. Additionally, patterns of differential expression across three time points in the liver revealed that Arid5b potentially regulates hundreds of genes. Functional enrichment analyses pointed to cell cycling, mitochondrial function, signaling pathways, inflammatory response, and nutrient metabolism as potential causes of weight accumulation in Gough Island mice. Collectively, our results indicate that extensive gene regulatory evolution in metabolic organs accompanied the rapid evolution of gigantism during the short time house mice have inhabited Gough Island.


Subject(s)
Biological Evolution , Body Size/genetics , Gene Expression , Mice/genetics , Mice/metabolism , Animals , Female , Hypothalamus/metabolism , Liver/growth & development , Liver/metabolism , Male , Mice/growth & development , Quantitative Trait Loci
3.
Gen Comp Endocrinol ; 260: 164-170, 2018 05 01.
Article in English | MEDLINE | ID: mdl-28951261

ABSTRACT

Probing previously unknown neuropeptides and/or peptide hormones is essential for our understanding of the regulation of energy homeostasis in the brain. We recently performed a cDNA subtractive screening of the chicken hypothalamus, which contained one of the feeding and energy metabolic centers. We found a gene encoding a novel protein of 182 amino acid residues, including one putative small secretory protein of 80 amino acid residues. The C-terminal amino acids of the small protein were Gly-Leu-NH2, and as a result, the small protein was termed neurosecretory protein GL (NPGL). Subcutaneous and intracerebroventricular infusions of NPGL increased body mass gain in chicks, suggesting a central role for this protein in regulating growth and energy homeostasis. A database search revealed that the Npgl gene is conserved in vertebrates, including mice and rats. This review summarizes the advances in the characterization, localization, and biological action of NPGL, in birds and rodents.


Subject(s)
Chickens , Energy Metabolism/genetics , Feeding Behavior/physiology , Mice , Nerve Tissue Proteins/physiology , Animals , Chickens/genetics , Chickens/metabolism , Homeostasis , Hypothalamus/metabolism , Mice/genetics , Mice/metabolism , Neuropeptides/physiology , Rats/genetics , Rats/metabolism , Vertebrates
4.
Food Funct ; 8(10): 3758-3768, 2017 Oct 18.
Article in English | MEDLINE | ID: mdl-28956585

ABSTRACT

During the complementary feeding (CF) period, nutritional imbalances can have negative consequences not only on a child's health in the short term but also later in adulthood, as a phenomenon known as "nutritional programming" takes place. The aim of this study was to evaluate the possible changes in body growth, gut microbiota (GM) and the immune system in mice fed with two different commercial sterilized baby foods in jars (BFJs) for CF. Mice fed with different BFJs (A and B groups) showed an accelerated growth from the fifth week of life when compared with the control (C) group. Group A showed a higher BMI, post-weaning growth rate, and IL-10 levels and a decrease in the Lactobacillus group. Group B showed a significant decrease in the total bacterial count, Lactobacillus group, Enterococcus spp. and Bacteroidetes-Prevotella. The Bifidobacterium genus tended to be lower in groups A and B. Akkermansia muciniphila was more frequently detected in group C. The results obtained from groups A and B can be attributed to the BFJ fatty acid profile, rich in UFAs. This study demonstrates for the first time that the commercial BFJ composition during CF might be a "programming" factor for body growth, GM and the immune system.


Subject(s)
Gastrointestinal Microbiome , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Infant Food/analysis , Mice/growth & development , Mice/immunology , Animals , Animals, Newborn/growth & development , Animals, Newborn/immunology , Animals, Newborn/metabolism , Animals, Newborn/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Fatty Acids/metabolism , Female , Gastrointestinal Tract/metabolism , Immune System , Immunity , Interleukin-10/genetics , Interleukin-10/immunology , Male , Mice/metabolism , Mice/microbiology
5.
J Agric Food Chem ; 65(32): 6957-6971, 2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28742334

ABSTRACT

This study aims to investigate the health effects of long-term dietary oxidized tyrosine (O-Tyr) and its main product (dityrosine) administration on mice metabolism. Mice received daily intragastric administration of either O-Tyr (320 µg/kg body weight), dityrosine (Dityr, 320 µg/kg body weight), or saline for consecutive 6 weeks. Urine and plasma samples were analyzed by NMR-based metabolomics strategies. Body weight, clinical chemistry, oxidative damage indexes, and histopathological data were obtained as complementary information. O-Tyr and Dityr exposure changed many systemic metabolic processes, including reduced choline bioavailability, led to fat accumulation in liver, induced hepatic injury, and renal dysfunction, resulted in changes in gut microbiota functions, elevated risk factor for cardiovascular disease, altered amino acid metabolism, induced oxidative stress responses, and inhibited energy metabolism. These findings implied that it is absolutely essential to reduce the generation of oxidation protein products in food system through improving modern food processing methods.


Subject(s)
Mice/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Animals , Female , Liver/chemistry , Liver/metabolism , Metabolomics , Oxidation-Reduction , Tyrosine/chemistry
6.
J Comp Neurol ; 523(8): 1258-80, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25560461

ABSTRACT

In the vestibular periphery of nearly every vertebrate, cholinergic vestibular efferent neurons give rise to numerous presynaptic varicosities that target hair cells and afferent processes in the sensory neuroepithelium. Although pharmacological studies have described the postsynaptic actions of vestibular efferent stimulation in several species, characterization of efferent innervation patterns and the relative distribution of efferent varicosities among hair cells and afferents are also integral to understanding how efferent synapses operate. Vestibular efferent markers, however, have not been well characterized in the turtle, one of the animal models used by our laboratory. Here we sought to identify reliable efferent neuronal markers in the vestibular periphery of turtle, to use these markers to understand how efferent synapses are organized, and to compare efferent neuronal labeling patterns in turtle with two other amniotes using some of the same markers. Efferent fibers and varicosities were visualized in the semicircular canal of red-eared turtles (Trachemys scripta elegans), zebra finches (Taeniopygia guttata), and mice (Mus musculus) utilizing fluorescent immunohistochemistry with antibodies against choline acetyltransferase (ChAT). Vestibular hair cells and afferents were counterstained using antibodies to myosin VIIa and calretinin. In all species, ChAT labeled a population of small diameter fibers giving rise to numerous spherical varicosities abutting type II hair cells and afferent processes. That these ChAT-positive varicosities represent presynaptic release sites were demonstrated by colabeling with antibodies against the synaptic vesicle proteins synapsin I, SV2, or syntaxin and the neuropeptide calcitonin gene-related peptide. Comparisons of efferent innervation patterns among the three species are discussed.


Subject(s)
Neurons, Efferent/cytology , Semicircular Canals/innervation , Turtles/anatomy & histology , Animals , Blotting, Western , Calbindin 2/metabolism , Cell Size , Choline O-Acetyltransferase/metabolism , Female , Finches/anatomy & histology , Finches/metabolism , Fluorescent Antibody Technique , Hair Cells, Vestibular/cytology , Hair Cells, Vestibular/metabolism , Male , Mice/anatomy & histology , Mice/metabolism , Microscopy, Confocal , Microscopy, Fluorescence , Myosin VIIa , Myosins/metabolism , Neurons, Efferent/metabolism , Semicircular Canals/metabolism , Species Specificity , Synapses/metabolism , Turtles/metabolism
7.
Horm Metab Res ; 43(13): 899-906, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22068812

ABSTRACT

Hypothalamus-pituitary-adrenal-axis activity is suggested to be involved in the pathophysiology of the metabolic syndrome. In diet-induced obesity mouse models, features of the metabolic syndrome are induced by feeding high fat diet. However, the models reveal conflicting results with respect to the hypothalamus-pituitary-adrenal-axis activation. The aim of this review was to assess the effects of high fat feeding on the activity of the hypothalamus-pituitary-adrenal-axis in mice. PubMed, EMBASE, Web of Science, the Cochrane database, and Science Direct were electronically searched and reviewed by 2 individual researchers. We included only original mouse studies reporting parameters of the hypothalamus-pituitary-adrenal-axis after high fat feeding, and at least 1 basal corticosterone level with a proper control group. Studies with adrenalectomized mice, transgenic animals only, high fat diet for less than 2 weeks, or other interventions besides high fat diet, were excluded. 20 studies were included. The hypothalamus-pituitary-adrenal-axis evaluation was the primary research question in only 5 studies. Plasma corticosterone levels were unchanged in 40%, elevated in 30%, and decreased in 20% of the studies. The effects in the peripheral tissues and the central nervous system were also inconsistent. However, major differences were found between mouse strains, experimental conditions, and the content and duration of the diets. This systematic review demonstrates that the effects of high fat feeding on the basal activity of the hypothalamus-pituitary-adrenal-axis in mice are limited and inconclusive. Differences in experimental conditions hamper comparisons and accentuate the need for standardized evaluations to discern the effects of diet-induced obesity on the hypothalamus-pituitary-adrenal-axis.


Subject(s)
Diet, High-Fat/adverse effects , Dietary Fats/metabolism , Disease Models, Animal , Hypothalamus/metabolism , Metabolic Syndrome/metabolism , Mice/metabolism , Pituitary-Adrenal System/metabolism , Animals , Dietary Fats/adverse effects , Humans
8.
Endocrinology ; 152(6): 2311-20, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21447634

ABSTRACT

The hypothalamic-pituitary-gonadal (HPG) axis exerts a profound effect on animal development, reproduction, and response to stress, and new insights into its complicated functional activities are continuously being made. In the present study, by using immunohistochemical studies and different mouse models (ovariectomy and ob/ob mice), we systemically analyzed the expression of a novel mitochondria-localized glutamic acid-rich protein (MGARP)/ovary-specific acid protein and demonstrated that MGARP is under the regulation of the HPG axis. MGARP is highly enriched in steroidogenic tissues and the visual system. Interestingly, its expression increases as mice develop. Early in development, MGARP is mainly detected in the retina and adrenal gland. At this early developmental stage, its expression is not detectable in the gonads, but its expression in the gonads dramatically increases during the first 2-4 wk after birth. Importantly, MGARP levels correlate with estrogen levels in the ovaries during the estrous cycle, and estrogen regulates the expression of MGARP in a tissue-specific manner and through a feedback regulatory mechanism. Functional inhibition of GnRH with an antagonist strongly reduces MGARP levels, and knockout of leptin (ob/ob) significantly reduces the MGARP expression in follicular granular cells. We proposed a model that elucidates the role MGARP plays in the HPG axis. Within the HPG axis loop, MGARP participates in hormone biosynthesis while being under the regulation of the hormones derived from the HPG axis.


Subject(s)
Gene Expression Regulation , Gonads/metabolism , Hypothalamus/metabolism , Membrane Proteins/genetics , Mice/metabolism , Pituitary Gland/metabolism , Animals , Eye Proteins , Female , Gene Expression Regulation, Developmental , Gonads/growth & development , Hormones/metabolism , Male , Membrane Proteins/metabolism , Mice/genetics , Mice/growth & development , Mice, Inbred ICR , Mitochondrial Proteins
9.
Biol Reprod ; 83(1): 92-101, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20357268

ABSTRACT

Injection of mammalian sperm extracts or cRNA of the sperm-specific phospholipase C zeta 1 (PLCZ1) has been shown to trigger repetitive oscillations in the concentration of free calcium ([Ca(2+)](i)), leading to oocyte activation and embryo development in all mammals studied to date. While PLCZ1 has cross-species activity, it has also been observed that species-specific differences may exist in the frequency and pattern of the resulting [Ca(2+)](i) oscillations following PLCZ1 cRNA injection into oocytes of different species. Accordingly, we used a crossover design strategy to directly investigate the activity of murine and bovine PLCZ1 in both murine and bovine oocytes. In murine oocytes, injection of murine Plcz1 cRNA induced [Ca(2+)](i) oscillations at 10-fold lower concentrations than bovine PLCZ1, although in bovine oocytes bovine PLCZ1 was more effective than murine Plcz1 at inducing [Ca(2+)](i) oscillations. Investigation of ITPR1 (IP(3)R1) down-regulation in bovine oocytes by PLCZ1 cRNA also showed that bovine PLCZ1 was more active in homologous oocytes. To determine whether these PLCZs exhibited similar cellular distribution, Venus-tagged PLCZ1 cRNA was injected into oocytes, and PLCZ1 was overexpressed. Bovine PLCZ1 failed to accumulate in the pronucleus (PN) of bovine or murine zygotes, despite possessing a putative nuclear localization signal. Conversely, murine PLCZ1 accumulated in the PN of both murine and bovine zygotes. These results demonstrate that murine PLCZ1 and bovine PLCZ1 possess species-specific differences in activity and suggest potential differences in the mode of action of the protein between the two species. Variation in sperm PLCZ1 protein content among species, along with oocyte-specific differences in the localization and availability of PLCZ1 substrates, may further contribute to optimize the activation stimulus to enhance embryo development.


Subject(s)
Calcium Signaling , Cattle/metabolism , Mice/metabolism , Oocytes/metabolism , Phosphoinositide Phospholipase C/metabolism , RNA, Complementary/metabolism , Animals , Down-Regulation , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Microinjections , Recombinant Proteins/metabolism , Species Specificity , Spermatozoa/enzymology
11.
J Nutr ; 137(5): 1132-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17449570

ABSTRACT

In our previous work with rats, plasma and tissue homocysteine concentrations were decreased by selenium deprivation. The purpose of this study was to follow up and expand on that work by determining the effects of selenium status (deficient, adequate, and supranutritional) on several aspects of homocysteine metabolism involving methionine recycling and transsulfuration. A 2nd objective was to determine whether there are differences in how selenium status affects homocysteine metabolism in rats and mice. Male weanling Fischer-344 rats and male weanling CD-1 mice were fed diets containing 0, 0.2, or 2.0 microg selenium (as sodium selenite)/g for 72 d or 60 d, respectively. Plasma homocysteine and cysteine were significantly decreased by feeding rats or mice the selenium-deficient diet compared with feeding adequate or supranutritional selenium. On the other hand, plasma glutathione was increased by selenium deficiency only in rats. Also, the specific activities of liver betaine homocysteine methyltransferase and glycine N-methyltransferase were decreased by selenium deficiency in rats, but were unaffected by selenium status in mice. Real-time RT-PCR was used to determine the expression of the subunits of glutamate-cysteine ligase, which catalyzes the rate-limiting step in glutathione biosynthesis. The expression of Gclc, the catalytic subunit of glutamate-cysteine ligase, was upregulated by selenium deprivation in both rat and mouse liver. Gclm, the modifier subunit of glutamate-cysteine ligase, was downregulated in rats fed 2 microg Se/g compared with rats fed adequate or deficient selenium. Based on these findings, it is evident that selenium deficiency has different outcomes in mice and rats. These variables are all related to methionine/methyl metabolism. Although only one strain of rat was compared with one strain of mouse, this work suggests that differences between species may prove vital in determining which animal model is used in studies of selenium deficiency or in studies that are designed to ascertain chemopreventive mechanisms of selenium.


Subject(s)
Diet , Homocysteine/metabolism , Mice/metabolism , Rats/metabolism , Selenium/administration & dosage , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Cysteine/blood , Glutamate-Cysteine Ligase/metabolism , Glutathione/blood , Glycine N-Methyltransferase/metabolism , Isoenzymes/metabolism , Liver/metabolism , Male , Methionine/metabolism , Mice, Inbred Strains , Rats, Inbred F344 , Selenium/deficiency , Selenium/metabolism , Selenium/pharmacology , Species Specificity , Sulfur/metabolism
12.
Gene Expr Patterns ; 4(5): 595-9, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15261838

ABSTRACT

We have examined the mRNA expression pattern of the murine expressed sequence tag (EST) clone in embryonic and early postnatal mice. Expression was strongly and specifically localised to developing bones and odontoblasts in teeth, therefore we have named this gene Bono1 (Bone and odontoblasts). Bono1, which has human, rat and chicken orthologues designated as FKSG28 was expressed in most ossification regions of the head including calvarial bones, skull and jaws. Expression was localised to osteoblasts derived from both intramembraneous and endochondral ossification processes. Comparative analysis of the expression of Bono1 in the mandible with Bone sialoprotein (BSP), a marker of advanced osteoblastogenesis, revealed that Bono1 expression starts later in the osteoblast cell lineage than BSP. In the tooth, Bono1 was localised in secretory odontoblasts. This expression was complementary to BSP, which was only present in early pre-odontoblasts. In secretory odontoblasts, Bono1 was shown to be co-expressed with Dentin sialophosphoprotein (DSPP). In summary, Bono1 was expressed in functional osteoblasts and odontoblasts and was associated with regions of matrix mineralization.


Subject(s)
Bone and Bones/metabolism , Gene Expression , Insulin-Like Growth Factor Binding Proteins/metabolism , Mice/embryology , Odontoblasts/metabolism , Animals , DNA Primers , Expressed Sequence Tags/metabolism , Extracellular Matrix Proteins , In Situ Hybridization , Integrin-Binding Sialoprotein , Mice/metabolism , Phosphoproteins , Protein Precursors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sialoglycoproteins/metabolism , Skull/metabolism
13.
Exp Cell Res ; 289(1): 162-73, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-12941614

ABSTRACT

The development of skeletal muscle in mammals is promoted by the muscle-specific basic helix-loop-helix transcription factors of the MyoD family. Evidence also suggests that there are basic helix-loop-helix proteins that specifically inhibit skeletal myogenesis, including Mtwist, Mist1, and the most recently described, MyoR. It has been suggested that MyoR expression is limited to the precursors of the skeletal muscle lineage and acts as a transcriptional repressor of the muscle differentiation program. However, our results demonstrate that MyoR is expressed in several different, nonmuscle adult tissues. Furthermore, MyoR is expressed in the embryonic ectoderm of blastocyst stage mouse embryos, well before skeletal muscle specification and even before delineation of the mesodermal germ layer. Using embryonic ectoderm analogous stem cells, we demonstrate that in these nonmuscle cells, as in skeletal muscle precursor cells, expression of MyoR is inversely correlated with the extent of cellular differentiation as induced by retinoic acid. Our preliminary results indicate that overexpression of exogenous MyoR inhibits retinoic-acid-induced differentiation in EC cells and is lethal to early mouse embryos. Our results suggest a much broader role for MyoR in the repression and/or determination of embryonic cell differentiation.


Subject(s)
Cell Differentiation/physiology , Ectoderm/metabolism , Gene Expression Regulation, Developmental/physiology , Mice/embryology , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Cell Lineage/physiology , DNA, Complementary/analysis , DNA, Complementary/genetics , Ectoderm/cytology , Fetus , Gene Expression Regulation, Developmental/drug effects , Genes, Lethal/drug effects , Genes, Lethal/genetics , Genes, Regulator/drug effects , Genes, Regulator/physiology , Mice/metabolism , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , Molecular Sequence Data , Muscle Proteins , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/isolation & purification , Tretinoin/pharmacology
14.
J Androl ; 24(1): 51-8, 2003.
Article in English | MEDLINE | ID: mdl-12514083

ABSTRACT

Previously we demonstrated that the murine sperm adhesion molecule 1 (Spam1 or PH-20) is synthesized by the epididymal epithelium, preferentially in the distal region, and is released into the luminal fluid. We also showed that whereas testicular and epididymal Spam1 have hyaluronidase activity at neutral pH, they are under different transcriptional regulation. The aim of this study was to further compare characteristics of the two forms of this glycosyl-phosphatidylinositol-linked protein and their transcripts, and to determine whether secreted epididymal Spam1 is released with its lipid anchor. With GeneRacer amplification of the 3' end of the complementary DNA we show that the poly(A) tails are significantly (P <.05) shorter in the epididymis than in the testis. Two-dimensional polyacrylamide gel electrophoresis with immunoblotting reveals one to three isoforms for epididymal Spam1 with the isoelectric point (pl) ranging from 7.3 to 9.0, and four isoforms ranging from 6.6 to 9.0 pl for testicular Spam1. Two isoforms with a pl ranging from 7.6 to 9.0 were observed for caudal sperm. Lectin blotting analysis shows that Phaseolus vulgaris erythroagglutinin, Lycopersicon esculentum lectin (LEL), and Solanum tuberosum lectin, which all bind to N-linked chains, recognize a 67 kd band in the epididymis and caudal sperm, but not in the testis. Treatment of the protein extracts with anti-Spam1 serum prior to blotting with LEL led to the disappearance of the banding, indicating Spam1 specificity of the staining. The lectin peanut agglutinin, which preferentially binds to O-linked side chains, recognizes a 67 kd band in all three cell types. Enzymatic deglycosylation studies confirmed the presence of an O-linked glycan in all three cell types. Ultracentrifugation of the luminal fluid reveals that epididymal Spam1 is secreted predominantly as insoluble particles, which when treated with phosphatidylinositol-specific phospholipase C or Triton X-100, reveal that the majority of epididymal Spam1 is released with its lipid anchor, a form in which it can bind to sperm.


Subject(s)
Cell Adhesion Molecules/metabolism , Epididymis/metabolism , Glycosylphosphatidylinositols/metabolism , Mice/metabolism , Animals , Blotting, Western , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Electrophoresis, Gel, Two-Dimensional , Glycosylation , Hyaluronoglucosaminidase , Immunoblotting , Male , Mice, Inbred ICR , Octoxynol/pharmacology , Oligosaccharides/metabolism , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Poly A/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , Solubility , Spermatozoa/metabolism , Testis/metabolism , Type C Phospholipases/pharmacology
15.
J Comp Neurol ; 424(1): 86-98, 2000 Aug 14.
Article in English | MEDLINE | ID: mdl-10888741

ABSTRACT

The suprachiasmatic nucleus (SCN) is the dominant circadian pacemaker in mammals. To understand better the ontogeny of mouse SCN and the role of the pacemaker in peptide expression, the authors examined the distribution of cells that were immunoreactive for vasopressin (AVP) or vasoactive intestinal polypeptide (VIP) in wild type and Clock mutant mice at two developmental stages. Clock homozygous mice failed to show the dramatic increase in the number of VIP-immunoreactive (VIP-ir) neurons from postnatal day 6 (P6) to P30 that was found in the SCN of wild type mice. The number of AVP-ir neurons was relatively constant in the postnatal SCN but was significantly reduced in Clock/Clock mice. The effects of the Clock mutation varied with position in the SCN for both peptides. Densitometry of immunolabeled brains indicated that the Clock mutation reduced AVP expression specifically in the SCN and not in other brain areas. The SCN did not significantly change shape or size with age or Clock genotype. Taken together, these results indicate that the neonatal mouse SCN has its full complement of cells, some of which are not yet mature in their neuropeptide content. Furthermore, the observation that the Clock mutation appears to act on a subset of AVP and VIP cells suggests heterogeneity within these cell classes in the SCN.


Subject(s)
Aging/metabolism , Mice/metabolism , Neuropeptides/metabolism , Suprachiasmatic Nucleus/metabolism , Trans-Activators/physiology , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Arginine Vasopressin/metabolism , CLOCK Proteins , Female , Male , Mice, Inbred Strains , Neurons/metabolism , Suprachiasmatic Nucleus/cytology , Suprachiasmatic Nucleus/growth & development , Thalamus/metabolism , Tissue Distribution , Vasoactive Intestinal Peptide/metabolism
16.
Hepatology ; 29(5): 1556-62, 1999 May.
Article in English | MEDLINE | ID: mdl-10216142

ABSTRACT

Hepatic uptake of organic cations is essential for the metabolism and secretion of numerous endobiotics and drugs. Several hepatic organic cation transporters have been kinetically defined, yet have not been isolated or cloned. We have isolated a complementary DNA (cDNA) from both murine liver and kidney cDNA libraries (mOct1/Slc22a1), and have functionally expressed it in Xenopus laevis oocytes. Although mOct1/Slc22a1 is homologous to previously cloned rat and human organic cation transporters, organic cation transport kinetics differed markedly. mOct1/Slc22a1-RNA injection of oocytes resulted in the saturable, time- and temperature-dependent uptake of the quaternary organic cation [14C]-tetraethylammonium ([14C]-TEA), with a Km of 38 micromol/L. TEA uptake was inhibited by several other organic cation drugs, but was not inhibited by the organic cation n-methyl-nicotinamide (NMN), being instead stimulated by it (fourfold). [14C]-TEA uptake was also stimulated by an inside-outside proton gradient. mOct1/Slc22a1-injected oocytes transported the organic cations [3H]-1-methyl-4-phenylpyridium and [3H]-choline chloride, but did not transport other classes of organic compounds. mOct1/Slc22a1 encodes for a hepatic and renal organic cation transporter which may be important for the uptake and secretion of cationic drugs and endobiotics.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice/genetics , Mice/metabolism , 1-Methyl-4-phenylpyridinium/metabolism , Animals , Cations/metabolism , Cations/pharmacology , Choline/pharmacokinetics , Female , Kinetics , Membrane Potentials/physiology , Molecular Sequence Data , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Oocytes/metabolism , Organic Cation Transporter 1 , Protons , Tetraethylammonium/antagonists & inhibitors , Tetraethylammonium/pharmacokinetics , Xenopus laevis
17.
Am J Physiol ; 269(3 Pt 2): F405-18, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7573490

ABSTRACT

A full-length cDNA encoding the murine renal Na-K-Cl cotransporter (NKCC2) was cloned using library screening and anchored polymerase chain reaction. The deduced protein sequence contained 1,095 amino acids and was 93.5% identical to rabbit NKCC2 and 97.6% identical to rat BSC1. Two potential sites of phosphorylation by adenosine 3',5'-cyclic monophosphate-dependent protein kinase and seven potential sites of phosphorylation by protein kinase C, which were previously identified in the rabbit and rat sequences, were phylogenetically conserved in the mouse. The expression of NKCC2 in the mouse was examined with Northern blot analysis and in situ hybridization. Expression of NKCC2 was kidney specific in both adult and embryonic mice. In the developing metanephros, NKCC2 was induced at 14.5 days post coitus and was expressed in distal limbs of immature loops of Henle but was absent from the ureteric bud, S-shaped bodies, and earlier nephrogenic structures. Similar to the rabbit, isoforms of NKCC2 that differed in the sequence of a 96-bp segment were identified in the mouse. In situ hybridization revealed that the isoforms exhibited different patterns of expression in the mature thick ascending limb of the loop of Henle as follows: isoform F was most highly expressed in the inner stripe of outer medulla, isoform A was most highly expressed in the outer stripe of the outer medulla, and isoform B was most highly expressed in the cortical thick ascending limb. To verify that the isoforms were generated by alternative splicing of mutually exclusive cassette exons, genomic clones encoding murine NKCC2 were characterized. Cassette exons were identified that corresponded to each of the three isoforms and were flanked by consensus splice donor and acceptor sequences.


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Embryo, Mammalian/metabolism , Mice/metabolism , Aging/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Embryonic and Fetal Development , Genes , Mice/embryology , Molecular Probes/genetics , Molecular Sequence Data , Rabbits , Rats , Sodium-Potassium-Chloride Symporters
18.
Am J Physiol ; 268(5 Pt 1): C1207-14, 1995 May.
Article in English | MEDLINE | ID: mdl-7762614

ABSTRACT

The H(+)-K(+)-ATPase of the gastric parietal cells is responsible for the acidification of the stomach lumen. This heterodimeric protein belongs to the family of cation-translocating P-type ATPases, which includes the closely related Na(+)-ATPase. We have cloned the alpha-subunit cDNA of the Xenopus and murine gastric H(+)-K(+)-ATPase (alpha H-K). We have expressed Xenopus and murine alpha H-K along with the previously cloned gastric H(+)-K(+)-ATPase beta-subunit of rabbit (beta H-K) in Xenopus oocytes by cRNA injection. An antibody directed against the beta H-K coimmunoprecipitates under nondenaturing conditions the alpha H-K of both species, demonstrating assembly of the alpha/beta complex. Additionally, we demonstrate the presence of K(+)-transporting H(+)-K(+)-ATPase in the plasma membrane of oocytes by 86Rb- uptake. The H(+)-K(+)-ATPase-mediated K+ uptake was inhibited by the gastric H(+)-K(+)-ATPase inhibitor Sch-28080, but not by ouabain, and shows K(+)-dependent activation (K1/2 approximately 2 mM). Furthermore, H(+)-K(+)-ATPase-expressing oocytes show a Sch-28080 inhibitable proton extrusion. Our data indicate that the expressed H(+)-K(+)-ATPase behaves functionally in oocytes as in the gastric gland.


Subject(s)
H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Mice/metabolism , Stomach/enzymology , Xenopus laevis/metabolism , Animals , Base Sequence , Biological Transport , DNA, Complementary/genetics , Mice, Inbred BALB C , Molecular Sequence Data , Oligonucleotide Probes/genetics , Oocytes/metabolism , Protons , Rubidium/metabolism , Tissue Distribution
19.
Genomics ; 27(1): 131-41, 1995 May 01.
Article in English | MEDLINE | ID: mdl-7665160

ABSTRACT

We describe the isolation and characterization of a human gene (CLCN3) and its murine homologue (Clcn3) sharing significant sequence and structural similarities with all previously identified members of the voltage-gated chloride channel (ClC) family. This gene is expressed primarily in tissues derived from neuroectoderm. Within the brain, Clcn3 expression is particularly evident in the hippocampus, olfactory cortex, and olfactory bulb. CLCN3 encodes a 760-amino-acid protein that differs by only 2 amino acid residues from the protein encoded by Clcn3. CLCN3 protein also shows a high similarity with GEF1, an integral membrane protein of the yeast Saccharomyces cerevisiae known to be involved in respiration and iron-limited cell growth, and with the predicted protein product of a DNA sequence from the mold Septoria nodorum. This high degree of sequence conservation in very distantly related species such as human and yeast indicates that this gene has retained a fundamental function throughout evolution.


Subject(s)
Genes, Fungal , Genes , Membrane Proteins/genetics , Mice/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Chloride Channels/metabolism , Chlorides/metabolism , Chromosome Mapping , Chromosomes, Human, Pair 4 , DNA, Complementary/genetics , Fetal Proteins/genetics , Gene Expression , Humans , Hybrid Cells , In Situ Hybridization , Mice/metabolism , Mitosporic Fungi/genetics , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Oocytes/metabolism , Organ Specificity , Patch-Clamp Techniques , Polymerase Chain Reaction , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Tubulin/genetics , Xenopus laevis
20.
Mol Endocrinol ; 8(4): 448-55, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8052266

ABSTRACT

The androgen receptor (AR) mediates the biological functions of androgens and is essential for normal growth and differentiation of urogenital organs as well as initiation and maintenance of spermatogenesis. Withdrawal of androgens by castration or other methods has been shown to cause a marked, although often temporary, regression of many prostate cancers. In order to gain a better understanding of the transcriptional regulation of the AR, a series of truncation mutants derived from the 5'-region of the mouse AR (mAR) were inserted into the promoter-less plasmid pBLCAT3 and transiently expressed in the mouse alpha T3-1 and GT1-7 cell lines. The results of these experiments indicate the presence of a negative regulatory element in the 5'-untranslated region of the gene, which is able to reduce chloramphenicol acetyltransferase (CAT) activity by 77-89%. We have named this element the mAR suppressor (mARS). DNase-I protection assays of the 5'-untranslated region disclosed a protected domain. Gel mobility assays using the mARS revealed the presence of three protein-DNA complexes that could specifically bind to this protected domain. Insertion of the mARS into the thymidine kinase promoter containing pBLCAT2 vector resulted in a 2- to 10-fold decrease in CAT activity, but only if the insert was 3' to the start of transcription initiation. Finally, point mutations within the mARS were able to increase transcription of the AR promoter by 2.3-fold. The results of these experiments indicate that the mAR 5'-untranslated region contains a suppressor element.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Gene Expression Regulation , Genes , Mice/metabolism , Promoter Regions, Genetic , Receptors, Androgen/genetics , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Animals , Base Sequence , Cell Line , Cell Nucleus/metabolism , Chloramphenicol O-Acetyltransferase/biosynthesis , Chloramphenicol O-Acetyltransferase/genetics , Consensus Sequence , Hypothalamus/cytology , Molecular Sequence Data , Mutagenesis, Site-Directed , Pituitary Gland/cytology , Point Mutation , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sequence Deletion , Simplexvirus/genetics , Thymidine Kinase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL