Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Domest Anim Endocrinol ; 51: 1-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25460066

ABSTRACT

To investigate the impact of duration of maternal undernutrition in twin sheep pregnancies, ewes were either fed 100% (C) or 50% of their nutrient requirements from 28 to 78 d gestational age (dGA) and readjusted to 100% beginning at 79 dGA (LC) or continuously restricted from 28 to 135 dGA (LL). Weights of the fetus, empty carcass, brain, and liver were greater in the LC than LL fetuses at 135 dGA (P ≤ 0.05). Although umbilical vein (UmV) glucose concentrations did not differ, the UmV:umbilical artery (UmA) glucose gradient was smaller (0.26 ± 0.03 vs 0.38 ± 0.03 and 0.39 ± 0.04 mmol L(-1); P ≤ 0.05) in LL than C and LC fetuses, respectively. Umbilical vein concentrations of IGF-1 were less (46.7 ± 5.62 vs 74.3 ± 6.71 ng/mL; P ≤ 0.05) in LL than LC fetuses. Additionally, LL fetuses tended (P ≤ 0.10) to have lower UmA concentrations of insulin (0.24 ± 0.13 vs 0.70 ± 0.15 ng/mL) and IGF-1 (66.6 ± 7.51 vs 91.4 ± 8.97 ng/mL) than LC fetuses. Although most of the observed differences occurred between LC and LL pregnancies, LC fetuses tended (P ≤ 0.10) to have greater UmV and UmA pCO2 than C fetuses. Furthermore, the UmV:UmA O2 content gradient tended to be greater (5.02 ± 0.43 vs 3.41 ± 0.47; P ≤ 0.10) in C than LL fetuses. UmA placental lactogen also tended to be greater (46.6 ± 4.40 vs 31.1 ± 4.69 ng/mL; P ≤ 0.10) in LL than C fetuses. These data suggest that in twin pregnancies, maternal undernutrition followed by realimentation induces a different fetal outcome compared with continuous nutrient restriction, and both may differ physiologically from control fed pregnancies.


Subject(s)
Animal Nutritional Physiological Phenomena , Fetal Development/physiology , Malnutrition/veterinary , Maternal Nutritional Physiological Phenomena/physiology , Pregnancy Complications/veterinary , Sheep Diseases/physiopathology , Animals , Blood Glucose/analysis , Female , Fetal Blood/chemistry , Fetal Weight , Growth Hormone/blood , Humans , Insulin/blood , Insulin-Like Growth Factor I/analysis , Malnutrition/complications , Placental Lactogen/blood , Pregnancy , Pregnancy Complications/physiopathology , Pregnancy, Multiple , Sheep , Time Factors , Umbilical Veins
2.
J Dairy Sci ; 96(10): 6473-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23958007

ABSTRACT

Objectives of this study were to measure both daily and periprandial plasma ghrelin concentrations of postpubertal Holstein heifers during prolonged undernutrition. Following an acclimation period, Holstein heifers [n=10; 339.5 ± 8.6 kg of body weight (BW)] were fed ad libitum [well fed (WF); n=5] or restricted to 50% of ad libitum intake [underfed (UF); n=5) for 8 wk. Body condition scores (BCS) were recorded at the beginning and end of the treatment period, and weekly measurements of BW, plasma ghrelin, progesterone, and nonesterified fatty acids (NEFA) concentrations were obtained. Ovarian follicular and luteal structures were measured twice weekly via transrectal ultrasonography. Plasma ghrelin concentrations were also measured during a periprandial window bleed conducted at the end of the experiment. During the window bleed, samples were collected every 15 min between 0500 and 0900 h, with feed offered at 0700 h. Underfed heifers lost BW and BCS, whereas WF heifers gained weight and either increased or maintained BCS. Chronic underfeeding increased circulating ghrelin and NEFA concentrations. By wk 4 of the treatment period, circulating ghrelin concentrations of the UF heifers reached a plateau. Periprandial fluctuations in ghrelin concentrations were apparent as plasma ghrelin concentrations changed over time. Overall differences in periprandial plasma ghrelin concentrations were primarily due to prefeeding effects of plane of nutrition. Plasma ghrelin concentrations and change in BCS were negatively correlated such that heifers that lost the most BCS had the highest concentrations of circulating ghrelin. Two of the 5 UF heifers became anestrus by wk 3 of the treatment period. Despite being of similar age, the heifers that became anestrus had lower BW and plasma ghrelin concentrations than the UF heifers that continued to ovulate. In the current experiment, long-term undernutrition elicited ghrelin responses similar to those reported for shorter durations of nutrient restriction in cattle and other ruminants. These results demonstrate that plane of nutrition is a chronic regulator of plasma ghrelin concentrations, and that these concentrations can be experimentally manipulated in postpubertal heifers for up to 8 wk with no evidence of an adaptive response.


Subject(s)
Ghrelin/blood , Malnutrition/blood , Anestrus/blood , Animals , Body Weight/physiology , Cattle , Fatty Acids, Nonesterified/blood , Female , Malnutrition/physiopathology , Nutritional Status
3.
J Dairy Sci ; 96(1): 150-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23141832

ABSTRACT

The aim of this experiment was to localize the mRNA and protein of ghrelin and its active receptor, growth hormone secretagogue 1A (GHS-R1A), within the reproductive tract of dairy cattle. Ghrelin is an orexigenic hormone that has been identified as a potent regulator of energy homeostasis. Recent evidence suggests that ghrelin may also serve as a metabolic signal to the reproductive tract. Ghrelin and GHS-R1A have been identified in the reproductive tract of several species, including humans, mice, and rats. However, ghrelin and GHS-R1A expression have not been described within bovine reproductive tissues. Therefore, the ampulla, isthmus, uterine body, corpus luteum, and follicles were harvested from 3 Holstein heifers (15.91±0.07 mo of age) immediately following exsanguination. Duodenum and hypothalamus were collected as positive controls for ghrelin and GHS-R1A, respectively. Tissues were fixed in 10% formalin and embedded in paraffin for microscopy. Additional samples were stored at -80°C for detection of mRNA. Ghrelin and GHS-R1A mRNA and protein were observed in all tissue types within the reproductive tract of dairy heifers; however, expression appeared to be cell specific. Furthermore, ghrelin protein appeared to be localized to the cytoplasm, whereas GHS-R1A protein was found on the plasma membrane. Within the reproductive tissues, ghrelin mRNA and protein were most abundantly expressed in the ampulla of the oviduct. Concentrations of GHS-R1A were lower than those of ghrelin but differed between tissues. This is one of the first studies to provide molecular evidence for the presence of ghrelin and GHS-R1A within the entire reproductive tract. However, implications for fertility remain to be determined.


Subject(s)
Genitalia, Female/chemistry , Ghrelin/physiology , Receptors, Ghrelin/physiology , Animals , Cattle , Corpus Luteum/chemistry , Corpus Luteum/physiology , Duodenum/chemistry , Female , Fluorescent Antibody Technique/veterinary , Genitalia, Female/physiology , Ghrelin/analysis , Hypothalamus/chemistry , Ovarian Follicle/chemistry , Ovarian Follicle/physiology , Receptors, Ghrelin/analysis , Uterus/chemistry , Uterus/physiology
4.
J Biol Chem ; 276(30): 27770-7, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11356842

ABSTRACT

Accumulating evidence indicates that the beta-arrestins act as scaffold molecules that couple G-protein-coupled receptors to mitogen-activated protein (MAP) kinase signaling pathways. Recently, we identified the c-Jun N-terminal kinase 3 (JNK3) as a beta-arrestin2-interacting protein in yeast-two hybrid and co-immunoprecipitation studies. Beta-arrestin2 acts as a scaffold to enhance signaling to JNK3 stimulated by overexpression of the MAP3 kinase ASK1 or by agonist activation of the angiotensin 1A receptor. Whereas beta-arrestin2 is a very strong activator of JNK3 signaling, beta-arrestin1 is very weak in this regard. The data also indicate that the specific step enhanced by beta-arrestin2 involves phosphorylation of JNK3 by the MAP2 kinase MKK4. We reasoned that defining the region (or domain) in beta-arrestin2 responsible for high level JNK3 activation would provide insight into the mechanism by which beta-arrestin2 enhances the activity of this signaling pathway. Using chimeric beta-arrestins, we have determined that sequences in the carboxyl-terminal region of beta-arrestin2 are important for the enhancement of JNK3 phosphorylation. More detailed analysis of the carboxyl-terminal domains of the beta-arrestins indicated that beta-arrestin2, but not beta-arrestin1, contains a sequence (RRSLHL) highly homologous to the conserved docking motif present in many MAP kinase-binding proteins. Replacement of the beta-arrestin2 RRS residues with the corresponding KP residues present in beta-arrestin1 dramatically reduced both JNK3 interaction and enhancement of JNK3 phosphorylation. Conversely, replacement of the KP residues in beta-arrestin1 with RRS significantly increased both JNK3 binding and enhancement of JNK3 phosphorylation. These results delineate a mechanism by which beta-arrestin2 functions as a scaffold protein in the JNK3 signaling pathway and implicate the conserved docking site in beta-arrestin2 as an important factor in binding JNK3 and stimulating the phosphorylation of JNK3 by MKK4.


Subject(s)
Arabidopsis Proteins , Arrestins/chemistry , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , COS Cells , Dose-Response Relationship, Drug , Enzyme Activation , Immunoblotting , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Biological , Molecular Sequence Data , Phosphorylation , Plant Proteins/metabolism , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptors, Adrenergic, beta-2/metabolism , Sequence Homology, Amino Acid , Signal Transduction , beta-Arrestins
5.
J Biol Chem ; 276(25): 23155-60, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11290747

ABSTRACT

"Transactivation" of epidermal growth factor receptors (EGFRs) in response to activation of many G protein-coupled receptors (GPCRs) involves autocrine/paracrine shedding of heparin-binding EGF (HB-EGF). HB-EGF shedding involves proteolytic cleavage of a membrane-anchored precursor by incompletely characterized matrix metalloproteases. In COS-7 cells, alpha(2A)-adrenergic receptors (ARs) stimulate ERK phosphorylation via two distinct pathways, a transactivation pathway that involves the release of HB-EGF and the EGFR and an alternate pathway that is independent of both HB-EGF and the EGFR. We have developed a mixed culture system to study the mechanism of GPCR-mediated HB-EGF shedding in COS-7 cells. In this system, alpha(2A)AR expressing "donor" cells are co-cultured with "acceptor" cells lacking the alpha(2A)AR. Each population expresses a uniquely epitope-tagged ERK2 protein, allowing the selective measurement of ERK activation in the donor and acceptor cells. Stimulation with the alpha(2)AR selective agonist UK14304 rapidly increases ERK2 phosphorylation in both the donor and the acceptor cells. The acceptor cell response is sensitive to inhibitors of both the EGFR and HB-EGF, indicating that it results from the release of HB-EGF from the alpha(2A)AR-expressing donor cells. Experiments with various chemical inhibitors and dominant inhibitory mutants demonstrate that EGFR-dependent activation of the ERK cascade after alpha(2A)AR stimulation requires Gbetagamma subunits upstream and dynamin-dependent endocytosis downstream of HB-EGF shedding and EGFR activation, whereas Src kinase activity is required both for the release of HB-EGF and for HB-EGF-mediated ERK2 phosphorylation.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , GTP-Binding Proteins/metabolism , Heparin/metabolism , Mitogen-Activated Protein Kinases/metabolism , Animals , COS Cells , Coculture Techniques , Enzyme Activation , Protein Binding
6.
Proc Natl Acad Sci U S A ; 98(5): 2449-54, 2001 Feb 27.
Article in English | MEDLINE | ID: mdl-11226259

ABSTRACT

Using both confocal immunofluorescence microscopy and biochemical approaches, we have examined the role of beta-arrestins in the activation and targeting of extracellular signal-regulated kinase 2 (ERK2) following stimulation of angiotensin II type 1a receptors (AT1aR). In HEK-293 cells expressing hemagglutinin-tagged AT1aR, angiotensin stimulation triggered beta-arrestin-2 binding to the receptor and internalization of AT1aR-beta-arrestin complexes. Using red fluorescent protein-tagged ERK2 to track the subcellular distribution of ERK2, we found that angiotensin treatment caused the redistribution of activated ERK2 into endosomal vesicles that also contained AT1aR-beta-arrestin complexes. This targeting of ERK2 reflects the formation of multiprotein complexes containing AT1aR, beta-arrestin-2, and the component kinases of the ERK cascade, cRaf-1, MEK1, and ERK2. Myc-tagged cRaf-1, MEK1, and green fluorescent protein-tagged ERK2 coprecipitated with Flag-tagged beta-arrestin-2 from transfected COS-7 cells. Coprecipitation of cRaf-1 with beta-arrestin-2 was independent of MEK1 and ERK2, whereas the coprecipitation of MEK1 and ERK2 with beta-arrestin-2 was significantly enhanced in the presence of overexpressed cRaf-1, suggesting that binding of cRaf-1 to beta-arrestin facilitates the assembly of a cRaf-1, MEK1, ERK2 complex. The phosphorylation of ERK2 in beta-arrestin complexes was markedly enhanced by coexpression of cRaf-1, and this effect is blocked by expression of a catalytically inactive dominant inhibitory mutant of MEK1. Stimulation with angiotensin increased the binding of both cRaf-1 and ERK2 to beta-arrestin-2, and the association of beta-arrestin-2, cRaf-1, and ERK2 with AT1aR. These data suggest that beta-arrestins function both as scaffolds to enhance cRaf-1 and MEK-dependent activation of ERK2, and as targeting proteins that direct activated ERK to specific subcellular locations.


Subject(s)
Arrestins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Angiotensin II/pharmacology , Animals , Cell Line , Enzyme Activation , Humans , Microscopy, Confocal , beta-Arrestin 2 , beta-Arrestins
7.
Science ; 290(5496): 1574-7, 2000 Nov 24.
Article in English | MEDLINE | ID: mdl-11090355

ABSTRACT

beta-Arrestins, originally discovered in the context of heterotrimeric guanine nucleotide binding protein-coupled receptor (GPCR) desensitization, also function in internalization and signaling of these receptors. We identified c-Jun amino-terminal kinase 3 (JNK3) as a binding partner of beta-arrestin 2 using a yeast two-hybrid screen and by coimmunoprecipitation from mouse brain extracts or cotransfected COS-7 cells. The upstream JNK activators apoptosis signal-regulating kinase 1 (ASK1) and mitogen-activated protein kinase (MAPK) kinase 4 were also found in complex with beta-arrestin 2. Cellular transfection of beta-arrestin 2 caused cytosolic retention of JNK3 and enhanced JNK3 phosphorylation stimulated by ASK1. Moreover, stimulation of the angiotensin II type 1A receptor activated JNK3 and triggered the colocalization of beta-arrestin 2 and active JNK3 to intracellular vesicles. Thus, beta-arrestin 2 acts as a scaffold protein, which brings the spatial distribution and activity of this MAPK module under the control of a GPCR.


Subject(s)
Arrestins/metabolism , MAP Kinase Kinase 4 , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Angiotensin/metabolism , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Arrestins/genetics , COS Cells , Cell Line , Cell Nucleus/metabolism , Cytosol/enzymology , Cytosol/metabolism , Endosomes/enzymology , Endosomes/metabolism , Enzyme Activation , Humans , MAP Kinase Kinase Kinase 5 , Mice , Mitogen-Activated Protein Kinase 10 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Rats , Receptor, Angiotensin, Type 1 , Recombinant Fusion Proteins/metabolism , Transfection , Two-Hybrid System Techniques , beta-Arrestin 2 , beta-Arrestins
8.
Science ; 284(5417): 1161-4, 1999 May 14.
Article in English | MEDLINE | ID: mdl-10325225

ABSTRACT

RAFT1 (rapamycin and FKBP12 target 1; also called FRAP or mTOR) is a member of the ATM (ataxia telangiectasia mutated)-related family of proteins and functions as the in vivo mediator of the effects of the immunosuppressant rapamycin and as an important regulator of messenger RNA translation. In mammalian cells RAFT1 interacted with gephyrin, a widely expressed protein necessary for the clustering of glycine receptors at the cell membrane of neurons. RAFT1 mutants that could not associate with gephyrin failed to signal to downstream molecules, including the p70 ribosomal S6 kinase and the eIF-4E binding protein, 4E-BP1. The interaction with gephyrin ascribes a function to the large amino-terminal region of an ATM-related protein and reveals a role in signal transduction for the clustering protein gephyrin.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor) , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , Sirolimus/pharmacology , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Gene Expression , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Mutation , Phosphorylation , Rats , Receptors, Glycine/metabolism , Repressor Proteins/metabolism , TOR Serine-Threonine Kinases
9.
Vet Microbiol ; 54(2): 155-66, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9057259

ABSTRACT

A polymerase chain reaction (PCR) assay was developed to detect. Chlamydia psittaci DNA in faeces and tissue samples from avian species. Primers were designed to amplify a 264 bp product derived from part of the 5' non-translated region and part of the coding region of the ompA gene which encodes the major outer membrane protein. Amplified sequences were confirmed by Southern hybridization using an internal probe. The sensitivity of the combined assay was found to be between 60 to 600 fg of chlamydial DNA (approximately 6 to 60 genome copies). The specificity of the assay was confirmed since PCR product was not obtained from samples containing several serotypes of C. trachomatis, strains of C. pneumoniae, the type strain of C. pecorum, nor from samples containing microorganisms commonly found in the avian gut flora. In this study, 404 avian faeces and 141 avian tissue samples received by the Central Veterinary Laboratory over a 6 month period were analysed by PCR, antigen detection ELISA and where possible, cell culture isolation. PCR performed favourably compared with ELISA and cell culture, or with ELISA alone. The PCR assay was especially suited to the detection of C. psittaci DNA in avian faeces samples. The test was also useful when applied to tissue samples from small contact birds associated with a case of human psittacosis where ELISA results were negative and chlamydial isolation was a less favourable method due to the need for rapid diagnosis.


Subject(s)
Bird Diseases , Chlamydophila psittaci/isolation & purification , DNA, Bacterial/analysis , Parrots , Polymerase Chain Reaction/methods , Psittacosis/diagnosis , Psittacosis/veterinary , Animals , Bacterial Outer Membrane Proteins/genetics , Cell Line , Chick Embryo , Chlamydophila psittaci/genetics , Chlamydophila psittaci/growth & development , DNA Primers , Enzyme-Linked Immunosorbent Assay/methods , Feces/microbiology , Genes, Bacterial , Humans , Polymerase Chain Reaction/veterinary , Psittacosis/transmission , Sensitivity and Specificity
10.
Nature ; 226(5248): 836-7, 1970 May 30.
Article in English | MEDLINE | ID: mdl-16057543
SELECTION OF CITATIONS
SEARCH DETAIL
...