Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Radiol Case Rep ; 19(7): 2816-2819, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38689806

ABSTRACT

Ovarian carcinoid is a rare well-differentiated neuroendocrine tumor resembling those arising in the gastrointestinal tract. We present a case of ovarian carcinoid with magnetic resonance imaging (MRI) findings. A 50-year-old woman with genital bleeding and severe constipation was referred to our hospital. On MR imaging, a left ovarian tumor showed iso to high signal intensity on T1-weighted images (T1WI), relatively low signal intensity on T2WI, and slightly high signal intensity on diffusion-weighted images. Additionally, the tumor demonstrated early and delayed strong contrast enhancement on dynamic contrast-enhanced images. The tumor was pathologically diagnosed with ovarian strumal carcinoid. High signal intensity on T1WI should be recognized as the MRI findings in ovarian carcinoids.

2.
Acta Radiol Open ; 10(6): 20584601211022504, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34178378

ABSTRACT

Endosalpingiosis is characterized by the presence of glands lined by benign tubal-type epithelium outside the fallopian tube. It is usually an incidental finding and rarely occurs as a tumor-like mass lesion. Here, we describe the magnetic resonance imaging findings of endosalpingiosis that presented as a paraovarian multicystic lesion. It exhibited iso to low intensity on T1-weighted images and inhomogeneous high intensity on T2-weighted images. The septa presented relatively iso to slight high intensity on T2-weighted images and strong contrast enhancement on dynamic contrast-enhanced imaging. Endosalpingiosis should be considered as a differential diagnosis in cases of paraovarian multicystic lesions along the uterine serosa.

3.
Jpn J Radiol ; 39(6): 527-539, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33517507

ABSTRACT

Uterine sarcomas account for less than 1% of gynecological malignancies and 2-5% of all uterine malignancies. Such sarcomas mainly include leiomyosarcoma (LMS) and endometrial stromal sarcoma (ESS). Additionally, inflammatory myofibroblastic tumor (IMT) and endometrial carcinoma arising in adenomyosis can occur as uterine myometrial tumors. Their differentiation from leiomyoma (LM), particularly degenerated LM and the malignant tumors, is challenging, but preoperative diagnosis is very important for the patient's management. We demonstrate the useful and compulsory findings to differentiate between uterine myometrial malignant tumors and degenerated LM with an unusual appearance.


Subject(s)
Leiomyoma/diagnostic imaging , Magnetic Resonance Imaging/methods , Uterine Neoplasms/diagnostic imaging , Adult , Diagnosis, Differential , Female , Humans , Leiomyoma/pathology , Middle Aged , Myometrium/diagnostic imaging , Myometrium/pathology , Radiologists , Uterine Neoplasms/pathology , Uterus/diagnostic imaging , Uterus/pathology
4.
Endocr J ; 64(8): 777-785, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28659539

ABSTRACT

It is well documented that estrogen is predominant inducer of hepatocyte growth factor (HGF) in a variety of cell types. However, the effect of progesterone (P) remains to be elusive. Thus, in the present study, we examined the effect of P and combined effect of P and 17ß-estradiol (E2) on HGF expression and production in 3T3-L1 fibroblastic preadipocytes and mature adipocytes, as a model of stromal cells. Northern blot analysis showed that hgf mRNA expressed in preadipocytes was notably higher than that of mature adipocytes, and increased by treatment of preadipocytes with E2 or 10 nM P, but not with 1,000 nM P. The E2-induced hgf mRNA expression was enhanced by 10 nM P, but suppressed by 1,000 nM P. Western blot analysis revealed that biological active forms of HGF protein was found in the preadipocyte culture medium, while the lesser amount of HGF precursor protein was detected in the mature adipocyte culture medium. The amounts of HGF were changed dependently on the hgf mRNA expression levels. These results indicate that HGF production is intricately regulated by E2 and P at the transcriptional levels in 3T3-L1 cells, and may explain the changes in the HGF production during the mammary gland development, especially decrease in HGF expression during pregnancy when P concentration is high.


Subject(s)
Adipocytes/drug effects , Hepatocyte Growth Factor/metabolism , Progesterone/administration & dosage , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Gene Expression/drug effects , Mice
5.
Mol Endocrinol ; 28(5): 758-67, 2014 May.
Article in English | MEDLINE | ID: mdl-24678731

ABSTRACT

Mammary-specific genetic programs are activated during pregnancy by the common transcription factor signal transducer and activator of transcription (STAT) 5. More than one third of these genes carry nuclear factor I/B (NFIB) binding motifs that coincide with STAT5 in vivo binding, suggesting functional synergy between these two transcription factors. The role of NFIB in this governance was investigated in mice from which Nfib had been inactivated in mammary stem cells or in differentiating alveolar epithelium. Although NFIB was not required for alveolar expansion, the combined absence of NFIB and STAT5 prevented the formation of functional alveoli. NFIB controlled the expression of mammary-specific and STAT5-regulated genes and chromatin immunoprecipitation-sequencing established STAT5 and NFIB binding at composite regulatory elements containing histone H3 lysine dimethylation enhancer marks and progesterone receptor binding. By integrating previously published chromatin immunoprecipitation-sequencing data sets, the presence of NFIB-STAT5 modules in other cell types was investigated. Notably, genomic sites bound by NFIB in hair follicle stem cells were also occupied by STAT5 in mammary epithelium and coincided with enhancer marks. Many of these genes were under NFIB control in both hair follicle stem cells and mammary alveolar epithelium. We propose that NFIB-STAT5 modules, possibly in conjunction with other transcription factors, control cell-specific genetic programs.


Subject(s)
NFI Transcription Factors/physiology , STAT5 Transcription Factor/physiology , Animals , Base Sequence , Binding Sites , Consensus Sequence , Female , Gene Expression Regulation , Gene Ontology , Lactation , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice, Nude , Mice, Transgenic , Pregnancy , Transcriptome
6.
Endocr Relat Cancer ; 21(3): 443-57, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24692510

ABSTRACT

Transformation-related protein 63 (Trp63), the predominant member of the Trp53 family, contributes to epithelial differentiation and is expressed in breast neoplasia. Trp63 features two distinct promoters yielding specific mRNAs encoding two major TRP63 isoforms, a transactivating transcription factor and a dominant negative isoform. Specific TRP63 isoforms are linked to cell cycle arrest, apoptosis, survival, and epithelial mesenchymal transition (EMT). Although TRP63 overexpression in cultured cells is used to elucidate functions, little is known about Trp63 regulation in normal and cancerous mammary tissues. This study used ChIP-seq to interrogate transcription factor binding and histone modifications of the Trp63 locus in mammary tissue and RNA-seq and immunohistochemistry to gauge gene expression. H3K4me2 and H3K4me3 marks coincided only with the proximal promoter, supporting RNA-seq data showing the predominance of the dominant negative isoform. STAT5 bound specifically to the Trp63 proximal promoter and Trp63 mRNA levels were elevated upon deleting Stat5 from mammary tissue, suggesting its role as a negative regulator. The dominant negative TRP63 isoform was localized to nuclei of basal mammary epithelial cells throughout reproductive cycles and retained in a majority of the triple-negative cancers generated from loss of full-length Brca1. Increased expression of dominant negative isoforms was correlated with developmental windows of increased progesterone receptor binding to the proximal Trp63 promoter and decreased expression during lactation was correlated with STAT5 binding to the same region. TRP63 is present in the majority of triple-negative cancers resulting from loss of Brca1 but diminished in less differentiated cancer subtypes and in cancer cells undergoing EMT.


Subject(s)
Cell Differentiation , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Phosphoproteins/physiology , STAT5 Transcription Factor/metabolism , Trans-Activators/physiology , Animals , BRCA1 Protein/physiology , Blotting, Western , Cells, Cultured , Female , High-Throughput Nucleotide Sequencing , Histones/metabolism , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reproduction , Reverse Transcriptase Polymerase Chain Reaction
7.
Mol Cell Biol ; 34(3): 464-73, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24277936

ABSTRACT

Differentiation of mammary secretory epithelium during pregnancy is characterized by sequential activation of genes over several orders of magnitude. Although the transcription factor STAT5 is key to alveolar development, it is not clear to what extent it controls temporal activation of genetic programs in secretory epithelium. To uncover molecular mechanisms effecting progressive differentiation, we explored genome-wide STAT5 binding and H3K4me3 (i.e., trimethylated histone H3 at K4) marks in mammary tissues at early and midpregnancy and at parturition. STAT5 binding to genes induced during pregnancy was low in immature mammary tissue but increased with epithelial differentiation. Increased STAT5 binding was associated with the establishment of H3K4me3 marks and transcriptional activation. STAT5 binding preceded the formation of H3K4me3 marks in some mammary-specific genes. De novo STAT5 binding was also found at distal sites, indicating enhancers. Furthermore, we established an exhaustive mammary transcriptome. Through integration of RNA-seq and STAT5 and H3K4me4 ChIP-seq data, we discovered novel mammary-specific alternative promoters and genes, including noncoding RNAs. Our findings suggest that STAT5 is an early step in establishing transcription complexes on genes specifically expressed in mammary epithelium. This is the first study in an organ that links progressive chromatin occupancy of STAT5 to the acquisition of H3K4me3 marks and transcription during hormone-induced differentiation.


Subject(s)
Histones/metabolism , Mammary Glands, Animal/metabolism , STAT5 Transcription Factor/genetics , Transcriptional Activation , Animals , Animals, Newborn , Chromatin/genetics , Chromatin/metabolism , Epithelium/growth & development , Epithelium/metabolism , Female , Gene Expression Regulation, Developmental , Gene Ontology , Lysine/metabolism , Mammary Glands, Animal/growth & development , Methylation , Mice , Oligonucleotide Array Sequence Analysis , Phosphorylation , Pregnancy , Promoter Regions, Genetic/genetics , Protein Binding , STAT5 Transcription Factor/metabolism , Transcriptome
8.
Nucleic Acids Res ; 41(3): 1622-36, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23275557

ABSTRACT

The transcription factors Signal Transducer and Activator of Transcription (STAT) 5A/B mediate prolactin-induced mammary development during pregnancy. However, it is not clear how the different processes, expansion and maturation of alveolar precursor cells and the differential induction of milk protein genes are regulated on a molecular level. We have used mouse genetics and genome-wide analyses to determine how altering concentrations of STAT5A and STAT5B impacts mammary epithelial development during pregnancy and the regulation of target genes. The presence of only a single Stat5a or Stat5b allele was sufficient for the establishment of histologically undifferentiated alveolar units and two alleles permitted the execution of a differentiation program similar to that found with all four alleles. While one copy of Stat5 induced limited expression of target genes, two copies activated a lactation-like gene signature. Using ChIP-seq analyses on intact tissue under physiological conditions, we found that highly expressed and regulated genes were bound by STAT5 in their promoter proximal regions, whereas upstream binding had minor biological consequences. Remarkably, 80% of the genes bound by STAT5 in vivo were not under STAT5 control. RNA polymerase II intensity was directly proportional to STAT5 concentration only on STAT5 regulated genes providing mechanistic insight by which STAT5 activates mammary specific genes.


Subject(s)
Gene Expression Regulation, Developmental , Lactation/genetics , Mammary Glands, Animal/metabolism , STAT5 Transcription Factor/metabolism , Animals , Binding Sites , Epithelium/anatomy & histology , Epithelium/growth & development , Epithelium/metabolism , Female , Genotype , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/growth & development , Mice , Mice, Knockout , Mice, Nude , Pregnancy , RNA Polymerase II/metabolism , STAT5 Transcription Factor/genetics
9.
Genesis ; 50(9): 665-71, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22389215

ABSTRACT

Genome wide analysis revealed the miR-17/92 cluster as a STAT5 target. This cluster encodes six microRNAs, which predictably target genes that play a role in mammary gland development. In this study, we have deleted the miR-17/92 cluster in mammary stem cells and evaluated in the mouse its function during mammary gland development. Loss of the miR-17/92 cluster did not affect mammary development from prepuberty to lactation. Our studies demonstrated that, while expression of the miR-17/92 cluster is under control of the key mammary transcription factor STAT5, its presence is not required for normal mammary development or lactation.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Mammary Glands, Animal/growth & development , MicroRNAs/genetics , Multigene Family/physiology , STAT5 Transcription Factor/genetics , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Epithelial Cells/metabolism , Female , Gene Deletion , High-Throughput Nucleotide Sequencing , Lactation/genetics , Mammary Glands, Animal/cytology , Mice , Mice, Nude , Pregnancy , RNA, Messenger/genetics , STAT5 Transcription Factor/metabolism , Sequence Analysis, DNA , Stem Cells
10.
Neoplasia ; 12(11): 899-905, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21076615

ABSTRACT

Type I and type II classes of interferons (IFNs) signal through the JAK/STAT1 pathway and are known to be important in adaptive and innate immune responses and in protection against tumors. Although STAT1 is widely considered a tumor suppressor, it remains unclear, however, if this function occurs in tumor cells (cell autonomous) or if STAT1 acts primarily through immune cells. Here, the question of whether STAT1 has a cell autonomous role in mammary tumor formation was addressed in a mouse model of ERBB2/neu-induced breast cancer in the absence and presence of STAT1. For this purpose, mice that carry floxed Stat1 alleles, which permit cell-specific removal of STAT1, were generated. To induce tumors only in mammary cells lacking STAT1, Stat1 floxed mice were crossed with transgenic mice that express cre recombinase and the neu oncogene under the mouse mammary tumor virus LTR (Stat1fl/fl NIC). Stat1 was effectively deleted in mammary epithelium of virgin Stat1fl/fl NIC females. Time-to-tumor onset was significantly shorter in Stat1fl/fl NIC females than in WT NIC (Wilcoxon rank sum test, P = .02). The median time-to-tumor onset in the Stat1fl/fl NIC mice was 49.4 weeks, whereas it was 62.4 weeks in the WT NIC mice. These results suggest that STAT1 in mammary epithelial cells may play a role in suppressing tumorigenesis. The Stat1 floxed allele described in this study is also a unique resource to determine the cellular targets of IFNs and STAT1 action, which should aid our understanding and appreciation of these pathways.


Subject(s)
Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Receptor, ErbB-2/genetics , STAT1 Transcription Factor/genetics , Animals , Blotting, Western , Cell Nucleus/metabolism , Cells, Cultured , Embryo, Mammalian/cytology , Epithelium/metabolism , Female , Fibroblasts/metabolism , Kaplan-Meier Estimate , Male , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Receptor, ErbB-2/metabolism , STAT1 Transcription Factor/metabolism , Tumor Burden , Tumor Cells, Cultured
11.
Genes Dev ; 23(20): 2382-7, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19833766

ABSTRACT

Mammary alveologenesis is abrogated in the absence of the transcription factors STAT5A/5B, which mediate cytokine signaling. To reveal the underlying causes for this developmental block, we studied mammary stem and progenitor cells. While loss of STAT5A/5B did not affect the stem cell population and its ability to form mammary ducts, luminal progenitors were greatly reduced and unable to form alveoli during pregnancy. Temporally controlled expression of transgenic STAT5A in mammary epithelium lacking STAT5A/5B restored the luminal progenitor population and rescued alveologenesis in a reversible fashion in vivo. Thus, STAT5A is necessary and sufficient for the establishment of luminal progenitor cells.


Subject(s)
Mammary Glands, Animal/cytology , STAT5 Transcription Factor/metabolism , Stem Cells/cytology , Animals , Female , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , STAT5 Transcription Factor/genetics
12.
Dev Dyn ; 238(5): 1092-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19384959

ABSTRACT

Mammary glands develop postnatally in response to the hypothalamic-pituitary-gonadal axis. Obesity-induced changes in the local environment, however, retard mammary gland development during late pregnancy and lactation. To clarify the effects of obesity on fundamental duct development, we compared the mammary glands of nulliparous nonpregnant obese mice fed a high-fat diet with those of lean mice fed a normal diet. Obese mice had enlarged mammary glands, reflecting fat pad size, whereas the ducts in obese mice showed a less dense distribution with less frequent branching. Additionally, the ducts were surrounded by thick collagen layers, and were incompletely lined with myoepithelium. Because leptin receptors were localized in the epithelium region and leptin that was highly expressed in the obese glands suppressed mammary epithelial cell proliferation in vitro, the present results suggest that obesity disrupts mammary ductal development, possibly by remodeling the mammary microenvironment and promoting the expression of such paracrine factors as leptin.


Subject(s)
Dietary Fats/adverse effects , Leptin/metabolism , Mammary Glands, Animal/abnormalities , Mammary Glands, Animal/physiopathology , Obesity/physiopathology , Receptors, Leptin/metabolism , Adiponectin/blood , Animals , Diet , Dietary Fats/administration & dosage , Fatty Acids, Nonesterified/blood , Female , Insulin/blood , Leptin/blood , Mammary Glands, Animal/pathology , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/pathology
13.
J Exp Med ; 206(4): 819-31, 2009 Apr 13.
Article in English | MEDLINE | ID: mdl-19332876

ABSTRACT

The molecular mechanisms underlying the development of hepatocellular carcinoma are not fully understood. Liver-specific signal transducer and activator of transcription (STAT) 5A/B-null mice (STAT5-LKO) were treated with carbon tetrachloride (CCl(4)), and histological analyses revealed liver fibrosis and tumors. Transforming growth factor (TGF)-beta levels and STAT3 activity were elevated in liver tissue from STAT5-LKO mice upon CCl(4) treatment. To define the molecular link between STAT5 silencing and TGF-beta up-regulation, as well as STAT3 activation, we examined STAT5-null mouse embryonic fibroblasts and primary hepatocytes. These cells displayed elevated TGF-beta protein levels, whereas messenger RNA levels remained almost unchanged. Protease inhibitor studies revealed that STAT5 deficiency enhanced the stability of mature TGF-beta. Immunoprecipitation and immunohistochemistry analyses demonstrated that STAT5, through its N-terminal sequences, could bind to TGF-beta and that retroviral-mediated overexpression of STAT5 decreased TGF-beta levels. To confirm the in vivo significance of the N-terminal domain of STAT5, we treated mice that expressed STAT5 lacking the N terminus (STAT5-DeltaN) with CCl(4). STAT5-DeltaN mice developed CCl(4)-induced liver fibrosis but no tumors. In conclusion, loss of STAT5 results in elevated TGF-beta levels and enhanced growth hormone-induced STAT3 activity. We propose that a deregulated STAT5-TGF-beta-STAT3 network contributes to the development of chronic liver disease.


Subject(s)
Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Mice, Knockout/genetics , STAT3 Transcription Factor/physiology , STAT5 Transcription Factor/deficiency , Transforming Growth Factor beta/physiology , Animals , Carbon Tetrachloride/pharmacology , Disease Progression , Fibrinogens, Abnormal/genetics , Gene Deletion , Haptoglobins/genetics , Hepatocytes/cytology , Hepatocytes/pathology , Liver/physiopathology , Mice
14.
Vet J ; 176(3): 361-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-17449305

ABSTRACT

This study examined the mitogenic response of bovine peripheral T lymphocytes to leptin, a pleiotropic hormone regulating food intake and energy expenditure. Leptin alone slightly suppressed proliferation of T lymphocytes in the presence of concanavalin A (ConA). Leptin also inhibited proliferation of T lymphocytes induced by anti-CD3 antibody. ConA treatment activated some protein kinases, including p44/p42(MAPK) and Akt/PKB, while anti-CD3 antibody treatment increased mRNA expression of suppressor of cytokine signalling (SOCS) 3, interferon (IFN)gamma, interleukin (IL) 2 and IL4 in T lymphocytes. Leptin alone increased only SOCS3 mRNA expression. Simultaneous treatment with mitogens and leptin enhanced IFNgamma mRNA expression but decreased IL2 mRNA expression, without any synergistic effect on phosphorylation of protein kinases or mRNA expression of SOCS3 and IL4. These results suggest that leptin modulates bovine T lymphocyte functions.


Subject(s)
Cell Proliferation/drug effects , Cytokines/biosynthesis , Leptin/pharmacology , Protein Kinases/metabolism , T-Lymphocytes/cytology , Animals , Cattle , Concanavalin A , Cytokines/genetics , Cytokines/immunology , Female , Lymphocyte Activation , Mitogens/antagonists & inhibitors , Protein Kinases/genetics , Protein Kinases/immunology , Signal Transduction , T-Lymphocytes/drug effects
15.
J Vet Med Sci ; 69(5): 509-14, 2007 May.
Article in English | MEDLINE | ID: mdl-17551224

ABSTRACT

The proliferation of peripheral blood mononuclear cells (PBMC) containing both monocyte/macrophages and T lymphocytes increased after treatment with T-cell mitogen (concanavalin A: Con A). PBMC treated with either leptin alone or combination of leptin and ConA showed enhanced proliferative activity by 10-40%, compared with those treated with ConA alone. In contrast, isolated T lymphocytes treated with leptin and ConA showed lowered proliferative activity than the ConA-treated alone, indicating that leptin induced production of some cytokines from monocyte/macrophages, that subsequently resulted in enhancement of T lymphocytes proliferation in PBMC. Among the cytokines examined, monocyte/monocytes constitutively expressed interleukin (IL)-1beta, IL-12p35, IL-18 mRNA, and faintly expressed tumor necrosis factor (TNF)-alpha and IL-12p40 mRNA. Leptin treatment augmented the monocyte/macrophages mRNA expression of only TNF-alpha and IL-12p40 to comparable levels of cells treated with lipopolysaccharide (LPS). However, leptin treatment increased monocyte/macrophages production of IL-1beta as well as TNF-alpha, and induced the mRNA expression of caspase-1, which is shown to mediate the conversion of latent pro-IL-1beta and pro-IL-18 to active forms. These results suggest that leptin directly acts on monocyte/macrophages to produce factors that induce T lymphocytes proliferation such as IL-12p35/p40 complex through IL-12p40 induction and IL-1beta/IL-18 production through caspase-1 induction.


Subject(s)
Caspases/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Leptin/pharmacology , Macrophages/metabolism , Monocytes/metabolism , Analysis of Variance , Animals , Cattle , Concanavalin A/pharmacology , DNA Primers/genetics , Enzyme Induction/drug effects , Female , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
16.
Jpn J Vet Res ; 54(4): 183-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17405355

ABSTRACT

We examined the effect of stroma-derived factors, hepatocyte growth factor (HGF) and leptin, on morphological differentiation of bovine mammary epithelial cells (BMEC) in collagen gel three-dimensional culture in vitro. BMEC treated with HGF, but not leptin, formed duct-like organoids. The formation of organoids by HGF was enhanced by treatment with a mixture of insulin, cortisol and prolactin, while BMEC treated with the mixture alone did not produce the organoid. In contrast, the formation of organoids by HGF was dose-dependently inhibited by simultaneous addition of leptin, regardless of the presence or absence of the hormone mixture. These results suggest that stroma-derived factors intricately regulate mammary epithelial morphogenesis.


Subject(s)
Hepatocyte Growth Factor/antagonists & inhibitors , Leptin/pharmacology , Mammary Glands, Animal/drug effects , Animals , Cattle , Cell Differentiation/drug effects , Drug Interactions , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Hepatocyte Growth Factor/pharmacology , In Vitro Techniques , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Morphogenesis/drug effects , Morphogenesis/physiology
17.
J Vet Med Sci ; 69(2): 125-31, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17339755

ABSTRACT

Leptin, a pleiotropic hormone regulating food intake and energy expenditure, has been shown to directly modulate human polymorphonuclear neutrophil (PMN) functions or indirectly through the action of tumor necrosis factor-alpha (TNF-alpha). Bovine PMN have considerable different characteristics from human PMN. For example, it does not respond to N-formyl-Methionyl-Leucyl-phenylalanine, a well known human PMN activator. In the present study, we tested the effects of leptin and TNF-alpha on superoxide production and degranulation of bovine peripheral PMN, in which both long isoform of leptin receptor (Ob-Rb) and TNF receptor 1 were expressed. Human leptin, human TNF-alpha, phorbol myristate acetate (PMA) and opsonized zymosan particles (OZP) did not stimulate degranulation responses, while zymosan-activated serum (ZAS) did. Neither leptin nor TNF-alpha enhanced the ZAS-induced degranulation responses. TNF-alpha, PMA, OZP and ZAS increased superoxide production in different magnitudes, whereas leptin did not. TNF-alpha, but not leptin, enhanced OZP- and ZAS-induced superoxide production, possibly, in part due to facilitating translocation of p47(phox), a component of NADPH oxidase. These results indicate that, unlike in human PMN, leptin does not have any direct effect on degranulation and superoxide production in bovine PMN, although TNF-alpha influences superoxide production.


Subject(s)
Cattle/blood , Cell Degranulation/drug effects , Leptin/pharmacology , Neutrophils/drug effects , Neutrophils/physiology , Superoxides/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Drug Synergism , Female , Neutrophil Activation/drug effects , Neutrophils/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Receptors, Leptin , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Tetradecanoylphorbol Acetate/pharmacology , Zymosan/pharmacology
18.
Domest Anim Endocrinol ; 33(4): 400-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17011156

ABSTRACT

Leptin is an adipose tissue-derived cytokine plays key roles in the regulation of food intake and energy expenditure. However, regulatory mechanisms of leptin gene expression are not fully elucidated in ruminants that utilize short-chain fatty acids (SCFA), known as volatile fatty acids, as principal energy sources. In this study, we determined effects of SCFA and long-chain fatty acids (LCFA) on leptin expression in bovine adipocytes. Bovine stromal vascular cells isolated from subcutaneous adipose tissue of Holstein cows were cultured to confluence and treated sequentially with dexamethasone and isobutylmethylxanthine for 2 days and insulin and troglitazone for 12 days to achieve full differentiation to adipocytes. The cells started to accumulate lipids 4 days after the onset of treatment, with increased mRNA expression of leptin, as well as aP2, adiponectin, and PPAR-gamma. Removal of fetal calf serum and reduction of glucose in the culture medium of differentiated adipocytes decreased leptin mRNA expression. Subsequent addition of acetate, butyrate, or propionate dose-dependently restored and rather increased leptin expression, while addition of LCFA suppressed it. The stimulatory effect of acetate was abolished by prior treatment of the cells with pertussis toxin and by addition of LCFA. Furthermore, cows fasted for 48h and fed thereafter, elaborate reduced and increased plasma leptin levels, respectively. Thus, these results suggest that plasma leptin levels in cows are inversely controlled at the transcription level by VFA and LCFA, and that the effects of SCFA possibly act through a G protein-coupled receptor for SCFA.


Subject(s)
Adipocytes/metabolism , Fatty Acids, Volatile/pharmacology , Fatty Acids/pharmacology , Gene Expression Regulation/drug effects , Leptin/genetics , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Cattle , Cells, Cultured , Dexamethasone/pharmacology , Fasting , Food , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/physiology , Leptin/blood , Pertussis Toxin/pharmacology , RNA, Messenger , Reverse Transcriptase Polymerase Chain Reaction
19.
Domest Anim Endocrinol ; 30(3): 239-46, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16207523

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) is a pleiotropic cytokine that plays a crucial role in the embryonic and postnatal development of various organs including the mammary gland. We cloned bovine HGF and its c-Met receptor cDNAs, and examined their expression during mammary gland development in dairy cows. The 2.5-kbp HGF cDNA clone contained a 2190 bp open reading frame coding a 730 amino acid protein, while the 4.8-kbp c-Met cDNA clone contained a 4152 bp open reading frame coding a 1384 amino acid protein. The bovine HGF and c-Met sequences exhibited more than 87% identity with those of other mammals. RT-PCR analysis revealed ubiquitous expression of both HGF and c-Met mRNAs in various bovine tissues tested. HGF mRNA was detected only in the inactive stage of bovine mammary gland development and not in the developing, lactating, and involuting stages, while c-Met mRNA was detected in the inactive and involuting stages. Immunohistochemical analysis demonstrated that the c-Met protein was found on mammary epithelial cells in the inactive, developing, and involuting stages, and on myoepithelial cells in all stages. These results suggest pivotal roles of HGF and c-Met in the development of bovine mammary gland.


Subject(s)
Hepatocyte Growth Factor/genetics , Mammary Glands, Animal/physiology , Proto-Oncogene Proteins c-met/genetics , Animals , Base Sequence , Cattle , Female , Gene Expression , Hepatocyte Growth Factor/biosynthesis , Immunohistochemistry , Lactation , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary
20.
Vet Immunol Immunopathol ; 98(3-4): 175-84, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15010226

ABSTRACT

Molecule possessing ankyrin-repeats induced by lipopolysaccharide (MAIL) is known as an IkappaB protein induced after administration of bacterial lipopolysaccharide (LPS) to mice. In the present study, we cloned bovine MAIL cDNA and examined its mRNA expression in white blood cells isolated from Holstein cows. Bovine MAIL had more than 80% amino acid identities with murine and human MAILs, highly conserved ankyrin-repeat motifs and PEST-like sequences. Bovine MAIL mRNA was undetectable in isolated peripheral white blood cells, but rapidly induced (<1h) after stimulation by LPS and lipid A in vitro in a dose-dependent manner. The lipid A-induced MAIL mRNA expression was found in polymorphonuclear cells, monocytes/macrophages and total lymphocytes, but not in T-lymphocytes. MAIL mRNA was also induced in vivo in peripheral blood leukocytes of cows after intramammary injection of Escherichia coli derived from coliform mastitis. Thus, bovine MAIL, as rodent MAILs, is induced by inflammatory stimuli in specific immune cells in vitro and in vivo, suggesting a role in inflammatory responses to bacterial infection in cattle.


Subject(s)
Cattle/immunology , Nuclear Proteins/genetics , RNA, Messenger/biosynthesis , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern/veterinary , Cloning, Molecular , Conserved Sequence , Female , Gene Expression Regulation/immunology , Leukocytes, Mononuclear/immunology , Molecular Sequence Data , Nuclear Proteins/biosynthesis , Nuclear Proteins/immunology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sequence Alignment , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...