Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
Add more filters

Publication year range
1.
J Immunol ; 211(1): 34-42, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37212607

ABSTRACT

We recently discovered a (to our knowledge) new neuroimmune interaction named the gateway reflex, in which the activation of specific neural circuits establishes immune cell gateways at specific vessel sites in organs, leading to the development of tissue-specific autoimmune diseases, including a multiple sclerosis (MS) mouse model, experimental autoimmune encephalomyelitis (EAE). We have reported that peripheral-derived myeloid cells, which are CD11b+MHC class II+ and accumulate in the fifth lumbar (L5) cord during the onset of a transfer model of EAE (tEAE), play a role in the pain-mediated relapse via the pain-gateway reflex. In this study, we investigated how these cells survive during the remission phase to cause the relapse. We show that peripheral-derived myeloid cells accumulated in the L5 cord after tEAE induction and survive more than other immune cells. These myeloid cells, which highly expressed GM-CSFRα with common ß chain molecules, grew in number and expressed more Bcl-xL after GM-CSF treatment but decreased in number by blockade of the GM-CSF pathway, which suppressed pain-mediated relapse of neuroinflammation. Therefore, GM-CSF is a survival factor for these cells. Moreover, these cells were colocalized with blood endothelial cells (BECs) around the L5 cord, and BECs expressed a high level of GM-CSF. Thus, GM-CSF from BECs may have an important role in the pain-mediated tEAE relapse caused by peripheral-derived myeloid cells in the CNS. Finally, we found that blockade of the GM-CSF pathway after pain induction suppressed EAE development. Therefore, GM-CSF suppression is a possible therapeutic approach in inflammatory CNS diseases with relapse, such as MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neuroinflammatory Diseases , Endothelial Cells/metabolism , Central Nervous System , Pain/metabolism , Myeloid Cells , Recurrence
2.
Surg Today ; 52(7): 989-994, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35606618

ABSTRACT

This article translates the guidelines for cadaver surgical training (CST) published in 2012 by Japan Surgical Society (JSS) and Japanese Association of Anatomists from Japanese to English. These guidelines are based on Japanese laws and enable the usage of donated cadavers for CST and clinical research. The following are the conditions to implement the activities outlined in the guidelines. The aim is to improve medicine and to contribute to social welfare. Activities should only be carried out at medical or dental universities under the centralized control by the department of anatomy under the regulation of Japanese law. Upon the usage of cadavers, registered donors must provide a written informed-consent for their body to be used for CST and other activities of clinical medicine. Commercial use of cadavers and profit-based CST is strongly prohibited. Moreover, all the cadaver-related activities except for the commercial-based ones require the approval of the University's Institutional Review Board (IRB) before implementation. The expert committee organized at each university for the implementation of CST should summarize the implementation of the program and report the details of the training program, operating costs, and conflicts of interest to the CST Promotion Committee of JSS.


Subject(s)
Anatomists , Clinical Medicine , Cadaver , Dissection , Humans , Japan
3.
EMBO J ; 35(12): 1346-63, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27220846

ABSTRACT

A primary cilium is a microtubule-based sensory organelle that plays an important role in human development and disease. However, regulation of Akt in cilia and its role in ciliary development has not been demonstrated. Using yeast two-hybrid screening, we demonstrate that Inversin (INVS) interacts with Akt. Mutation in the INVS gene causes nephronophthisis type II (NPHP2), an autosomal recessive chronic tubulointerstitial nephropathy. Co-immunoprecipitation assays show that Akt interacts with INVS via the C-terminus. In vitro kinase assays demonstrate that Akt phosphorylates INVS at amino acids 864-866 that are required not only for Akt interaction, but also for INVS dimerization. Co-localization of INVS and phosphorylated form of Akt at the basal body is augmented by PDGF-AA Akt-null MEF cells as well as siRNA-mediated inhibition of Akt attenuated ciliary growth, which was reversed by Akt reintroduction. Mutant phosphodead- or NPHP2-related truncated INVS, which lack Akt phosphorylation sites, suppress cell growth and exhibit distorted lumen formation and misalignment of spindle axis during cell division. Further studies will be required for elucidating functional interactions of Akt-INVS at the primary cilia for identifying the molecular mechanisms underlying NPHP2.


Subject(s)
Basal Bodies/metabolism , Cilia/metabolism , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/metabolism , Animals , Cell Line , DNA Mutational Analysis , Humans , Mice , Phosphorylation , Protein Interaction Mapping , Transcription Factors/genetics , Two-Hybrid System Techniques
4.
Biochem Biophys Res Commun ; 517(1): 146-154, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31351587

ABSTRACT

The phosphinositide PtdIns(3)P plays an important role in autophagy; however, the detailed mechanism of its activity remains unclear. Here, we used a Systematic Evolution of Ligands by EXponential enrichment (SELEX) screening approach to identify an RNA aptamer of 40 nucleotides that specifically recognizes and binds to intracellular lysosomal PtdIns(3)P. Binding occurs in a magnesium concentration- and pH-dependent manner, and consequently inhibits autophagy as determined by LC3II/I conversion, p62 degradation, formation of LC3 puncta, and lysosomal accumulation of Phafin2. These effects in turn inhibited lysosomal acidification, and the subsequent hydrolytic activity of cathepsin D following induction of autophagy. Given the essential role of PtdIns(3)P as a key targeting molecule for autophagy induction, identification of this novel PtdIns(3)P RNA aptamer provides new opportunities for investigating the biological functions and mechanisms of phosphoinositides.


Subject(s)
Aptamers, Nucleotide/metabolism , Phosphatidylinositol Phosphates/metabolism , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacology , Autophagy/drug effects , Base Sequence , Cell Line , Humans , Lysosomes/drug effects , Lysosomes/metabolism , SELEX Aptamer Technique , Vesicular Transport Proteins/metabolism
5.
PLoS Genet ; 12(10): e1006349, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27736879

ABSTRACT

Zinc transporters play a critical role in spatiotemporal regulation of zinc homeostasis. Although disruption of zinc homeostasis has been implicated in disorders such as intestinal inflammation and aberrant epithelial morphology, it is largely unknown which zinc transporters are responsible for the intestinal epithelial homeostasis. Here, we show that Zrt-Irt-like protein (ZIP) transporter ZIP7, which is highly expressed in the intestinal crypt, is essential for intestinal epithelial proliferation. Mice lacking Zip7 in intestinal epithelium triggered endoplasmic reticulum (ER) stress in proliferative progenitor cells, leading to significant cell death of progenitor cells. Zip7 deficiency led to the loss of Olfm4+ intestinal stem cells and the degeneration of post-mitotic Paneth cells, indicating a fundamental requirement for Zip7 in homeostatic intestinal regeneration. Taken together, these findings provide evidence for the importance of ZIP7 in maintenance of intestinal epithelial homeostasis through the regulation of ER function in proliferative progenitor cells and maintenance of intestinal stem cells. Therapeutic targeting of ZIP7 could lead to effective treatment of gastrointestinal disorders.


Subject(s)
Cation Transport Proteins/genetics , Cell Proliferation/genetics , Endoplasmic Reticulum Stress/genetics , Zinc/metabolism , Animals , Apoptosis/genetics , Cation Transport Proteins/biosynthesis , Endoplasmic Reticulum/genetics , Epithelial Cells/metabolism , Gastrointestinal Tract/growth & development , Gastrointestinal Tract/metabolism , Gene Knockout Techniques , Homeostasis , Intestinal Mucosa/growth & development , Intestinal Mucosa/metabolism , Mice , Organoids/growth & development , Paneth Cells/metabolism , Stem Cells/metabolism
6.
Exp Eye Res ; 172: 1-9, 2018 07.
Article in English | MEDLINE | ID: mdl-29596849

ABSTRACT

The hyaloid vasculature constitutes a transitory system nourishing the internal structures of the developing eye, but the mechanism of vascular regression and its cell biological characteristics are not fully understood. The present study aimed to reveal the specificity of the hyaloid vessels by a systematic immunohistochemical approach for marker substances of myeloid cells and the extracellular matrix (ECM) in neonatal mice. Macrophages immunoreactive for F4/80, cathepsin D, and LYVE-1 gathered around the vasa hyaloidea propria (VHP), while small round cells in vascular lumen of VHP were selectively immunoreactive for galectin-3; their segmented nuclei and immunoreactivities for Ly-6G, CD11b, and myeloperoxidase indicated their neutrophilic origin. VHP possessed thick ECM and a dense pericyte envelope as demonstrated by immunostaining for laminin, type IV collagen, integrin ß1, and NG2. The galectin-3+ cells loosely aggregated with numerous erythrocytes in the lumen of hyaloid vessels in a manner reminiscent of vascular congestion. Galectin-3 is known to polymerize and form a complex with ECM and NG2 as well as recruit leukocytes on the endothelium. Observation of galectin-3 KO mice implicated the involvement of galectin-3 in the regression of hyaloid vasculature. Since macrophages may play central roles including blocking of the blood flow and the induction of apoptosis in the regression, galectin-3+ neutrophils may play a supportive role in the macrophage-mediated involution of the hyaloid vascular system.


Subject(s)
Blood Vessels/pathology , Vitreous Body/blood supply , Animals , Animals, Newborn , Antigens, Differentiation/metabolism , Antigens, Ly/metabolism , Atrophy , Biomarkers/metabolism , Blood Vessels/metabolism , CD11b Antigen/metabolism , Cathepsin D/metabolism , Extracellular Matrix Proteins/metabolism , Female , Fluorescent Antibody Technique, Indirect , Macrophages/metabolism , Mice , Microscopy, Fluorescence , Neutrophils/metabolism , Pericytes/metabolism , Peroxidase/metabolism , Pregnancy , Vesicular Transport Proteins/metabolism
7.
Am J Physiol Renal Physiol ; 313(1): F20-F29, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28298361

ABSTRACT

The natriuretic hormone CCK exhibits its gene transcripts in total kidney extracts. To test the possibility of CCK acting as an intrarenal mediator of sodium excretion, we examined mouse kidneys by 1) an in situ hybridization technique for CCK mRNA in animals fed a normal- or a high-sodium diet; 2) immuno-electron microscopy for the CCK peptide, 3) an in situ hybridization method and immunohistochemistry for the CCK-specific receptor CCKAR; 4) confocal image analysis of receptor-mediated Ca2+ responses in isolated renal tubules; and 5) metabolic cage experiments for the measurement of urinary sodium excretion in high-salt-fed mice either treated or untreated with the CCKAR antagonist lorglumide. Results showed the CCK gene to be expressed intensely in the inner medulla and moderately in the inner stripe of the outer medulla, with the expression in the latter being enhanced by high sodium intake. Immunoreactivity for the CCK peptide was localized to the rough endoplasmic reticulum of the medullary interstitial cells in corresponding renal regions, confirming it to be a secretory protein. Gene transcripts, protein products, and the functional activity for CCKAR were consistently localized to the late proximal tubule segments (S2 and S3) in the medullary rays, and the outer stripe of the outer medulla. Lorglumide significantly diminished natriuretic responses of mice to a dietary sodium load without altering the glomerular filtration rate. These findings suggest that the medullary interstitial cells respond to body fluid expansion by CCK release for feedback regulation of the late proximal tubular reabsorption.


Subject(s)
Cholecystokinin/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Proximal/metabolism , Natriuresis , Signal Transduction , Sodium, Dietary/administration & dosage , Water-Electrolyte Balance , Animals , Calcium/metabolism , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/genetics , Feedback, Physiological , Hormone Antagonists/pharmacology , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kidney Medulla/drug effects , Kidney Medulla/ultrastructure , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Male , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Natriuresis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Signal Transduction/drug effects , Time Factors , Water-Electrolyte Balance/drug effects
8.
Mol Hum Reprod ; 23(5): 271-281, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28333263

ABSTRACT

STUDY QUESTION: Do intraluteal prostaglandins (PG) contribute to luteal regulation in women? SUMMARY ANSWER: Prostaglandin E (PGE), which is produced in human granulosa-lutein cells stimulated with luteotropic hCG, exerts similar luteotropic effects to hCG, and the expression of PG synthetic and metabolic enzymes in the human CL is driven toward less PGE but more prostaglandin F (PGF) during luteolysis. WHAT IS KNOWN ALREADY: Uterine PGF is a major luteolysin in many non-primate species but not in women. Increases in the PGF synthase, aldo-ketoreductase family one member C3 (AKR1C3), have been observed in the CL of marmoset monkeys during luteolysis. PGE prevents spontaneous or induced luteolysis in domestic animals. STUDY DESIGN, SIZE, DURATION: Human CL tissues staged as the early-luteal (n = 6), mid-luteal (n = 6), late-luteal (n = 5) and menstrual (n = 3) phases were obtained at the time of hysterectomy for benign gynecological conditions. Luteinized granulosa cells (LGCs) were purified from follicular fluids obtained from patients undergoing assisted conception. PARTICIPANTS/MATERIALS, SETTING, METHODS: Upon collection, one half of the CL was snap-frozen and the other was fixed with formalin and processed for immunohistochemical analysis of a PGE synthase (PTGES). Quantitative RT-PCR was employed to examine changes in the mRNA abundance of PG synthetic and metabolic enzymes, steroidogenic enzymes, and luteolytic molecules in the staged human CL and in human LGCs in vitro treated with hCG, PGE and PGF. A PGE withdrawal experiment was also conducted in order to reveal the effects of the loss of PGE in LGCs. Progesterone concentrations in the culture medium were measured. MAIN RESULTS AND THE ROLE OF CHANCE: The key enzyme for PGE synthesis, PTGES mRNA was abundant in the functional CL during the mid-luteal phase (P < 0.01), while mRNA abundance for genes involved in PGF synthesis (AKR1B1 and AKR1C1-3) increased in the CL during the late-luteal phase and menstruation (P < 0.05-0.001). PTGES mRNA expression positively correlated with that of 3ß-hydroxysteroid dehydrogenase (HSD3B1; r = 0.7836, P < 0.001), while AKR1C3 expression inversely correlated with that of HSD3B1 (r = -0.7514, P = 0.0012) and PTGES (r = -0.6923, P = 0.0042). PGE exerted similar effects to hCG-promoting genes, such as steroidogenic acute regulatory protein (STAR) and HSD3B1, to produce progesterone and luteotropic PGE, suppress PGF synthetic enzymes and down-regulate luteolytic molecules such as ßA- and ßB-inhibin subunits (INHBA and INHBB) and bone morphogenetic proteins (BMP2, BMP4 and BMP6). PGE withdrawal resulted in reductions in the enzymes that produce progesterone (STAR; P < 0.001) and PGE (PTGES; P < 0.001), and the capacity to produce PGE decreased, while the capacity to produce PGF increased during the culture. The addition of PGF did not recapitulate the luteolytic effects of PGE withdrawal. LARGE SCALE DATA: None. LIMITATIONS, REASONS FOR CAUTION: Changes in mRNA expression of PG synthetic and metabolic enzymes may not represent actual increases in PGF during luteolysis in the CL. The effects of PGF on luteal cells currently remain unclear and the mechanisms responsible for decreases in the synthesis of PGE in vitro and at luteolysis have not been elucidated in detail. WIDER IMPLICATIONS OF THE FINDINGS: The results obtained strongly support a luteotropic function of PGE in regulation of the human CL. They suggest that the main PG produced in human luteal tissue changes from PGE to PGF during the maturation and regression of the CL, and the loss of PGE is more important than the effects of PGF during luteolysis in women. This may be accompanied by reduced effects of LH/hCG in luteal cells, particularly decreased activation of cAMP/protein kinase A; however, the underlying mechanisms remain unknown. STUDY FUNDING AND COMPETING INTEREST(S): This study was supported by the Cunningham Trust to WCD, a Postdoctoral Fellowship for Research Abroad from the Japan Society for the Promotion of Science and the Suntory Foundation for Life Sciences to J.N.-K.; W.C.D. is supported by an MRC Centre Grant G1002033 and a Scottish Senior Clinical Fellowship. The authors have nothing to disclose.


Subject(s)
Corpus Luteum/metabolism , Granulosa Cells/metabolism , Luteinization/physiology , Luteolysis/genetics , Prostaglandins E/genetics , 20-Hydroxysteroid Dehydrogenases/genetics , 20-Hydroxysteroid Dehydrogenases/metabolism , Aldehyde Reductase/genetics , Aldehyde Reductase/metabolism , Animals , Chorionic Gonadotropin/pharmacology , Corpus Luteum/cytology , Corpus Luteum/drug effects , Female , Gene Expression Regulation , Granulosa Cells/cytology , Granulosa Cells/drug effects , Humans , Inhibin-beta Subunits/genetics , Inhibin-beta Subunits/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Luteal Phase/physiology , Menstruation/physiology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Placenta Growth Factor/pharmacology , Primary Cell Culture , Progesterone/biosynthesis , Progesterone/metabolism , Progesterone Reductase/genetics , Progesterone Reductase/metabolism , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Prostaglandins E/deficiency , Prostaglandins E/pharmacology , Signal Transduction , Steroid Isomerases/genetics , Steroid Isomerases/metabolism
9.
J Physiol ; 594(12): 3339-52, 2016 06 15.
Article in English | MEDLINE | ID: mdl-26854275

ABSTRACT

KEY POINTS: Luminal short-chain fatty acids (SCFAs) influence gut physiological function via SCFA receptors and transporters. The contribution of an SCFA receptor, free fatty acid receptor (FFA)3, to the enteric nervous system is unknown. FFA3 is expressed in enteric cholinergic neurons. Activation of neural FFA3 suppresses Cl(-) secretion induced by nicotinic ACh receptor activation via a Gi/o pathway. Neural FFA3 may have an anti-secretory function by modulating cholinergic neural reflexes in the enteric nervous system. ABSTRACT: The proximal colonic mucosa is constantly exposed to high concentrations of microbially-produced short-chain fatty acids (SCFAs). Although luminal SCFAs evoke electrogenic anion secretion and smooth muscle contractility via neural and non-neural cholinergic pathways in the colon, the involvement of the SCFA receptor free fatty acid receptor (FFA)3, one of the free fatty acid receptor family members, has not been clarified. We investigated the contribution of FFA3 to cholinergic-mediated secretory responses in rat proximal colon. FFA3 was immunolocalized to enteroendocrine cells and to the enteric neural plexuses. Most FFA3-immunoreactive nerve fibres and nerve endings were cholinergic, colocalized with protein gene product (PGP)9.5, the vesicular ACh transporter, and the high-affinity choline transporter CHT1. In Ussing chambered mucosa-submucosa preparations (including the submucosal plexus) of rat proximal colon, carbachol (CCh)-induced Cl(-) secretion was decreased by TTX, hexamethonium, and the serosal FFA3 agonists acetate or propionate, although not by an inactive analogue 3-chloropropionate. Serosal application of a selective FFA3 agonist (N-[2-methylphenyl]-[4-furan-3-yl]-2-methyl-5-oxo-1,4,5,6,7,8-hexahydro-quinoline-3-carboxamide; MQC) dose-dependently suppressed the response to CCh but not to forskolin, with an IC50 of 13 µm. Pretreatment with MQC inhibited nicotine-evoked but not bethanechol-evoked secretion. The inhibitory effect of MQC was reversed by pretreatment with pertussis toxin, indicating that FFA3 acts via the Gi/o pathway. Luminal propionate induced Cl(-) secretion via the cholinergic pathway, which was reduced by MQC, as well as by TTX, hexamethonium or removal of the submucosal plexus. These results suggest that the SCFA-FFA3 pathway has a novel anti-secretory function in that it inhibits cholinergic neural reflexes in the enteric nervous system.


Subject(s)
Colon/physiology , Enteric Nervous System/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Carbachol/pharmacology , Chlorides/metabolism , Colon/drug effects , Colon/metabolism , HeLa Cells , Humans , In Vitro Techniques , Male , Neurons/physiology , Propionates/pharmacology , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/physiology
10.
Cell Tissue Res ; 364(1): 175-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26553655

ABSTRACT

Murine nasopharynx-associated lymphoid tissue (NALT), located at the base of the nasal cavity, serves as a major site for the induction of mucosal immune responses against airway antigens. The follicle-associated epithelium (FAE) covering the luminal surface of NALT is characterized by the presence of microfold cells (M cells), which take up and transport luminal antigens to lymphocytes. Glycoprotein 2 (GP2) has recently been identified as a reliable marker for M cells in Peyer's patches of the intestine. However, the expression of GP2 and other functional molecules in the M cells of NALT has not yet been examined. We have immunohistochemically detected GP2-expressing cells in the FAE of NALT and the simultaneous expression of other intestinal M-cell markers, namely Tnfaip2, CCL9, and Spi-B. These cells have been further identified as M cells because of their higher uptake capacity of luminal microbeads. Electron microscopic observations have shown that GP2-expressing cells on the FAE display morphological features typical of M cells: they possess short microvilli and microfolds on the luminal surface and are closely associated with intraepithelial lymphocytes. We have also found that the receptor activator of nuclear factor kappa-B ligand (RANKL) is expressed by stromal cells underneath the FAE, which provides its receptor RANK. The administration of RANKL markedly increases the number of GP2(+)Tnfaip2(+) cells on the NALT FAE and that of intestinal M cells. These results suggest that GP2(+)Tnfaip2(+) cells in NALT are equivalent to intestinal M cells, and that RANKL-RANK signaling induces their differentiation.


Subject(s)
Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Lymphoid Tissue/immunology , Pharynx/immunology , RANK Ligand/pharmacology , Signal Transduction/drug effects , Animals , Antigens, Differentiation/immunology , Cell Differentiation/immunology , GPI-Linked Proteins/immunology , Gene Expression Regulation/immunology , Lymphoid Tissue/cytology , Mice , Mice, Inbred BALB C , Pharynx/cytology , RANK Ligand/immunology , Signal Transduction/immunology
11.
Mol Reprod Dev ; 83(12): 1083-1091, 2016 12.
Article in English | MEDLINE | ID: mdl-27704662

ABSTRACT

Sialylation creates a negative charge on the cell surface that can interfere with blastocyst implantation. For example, α2,6-sialylation on terminal galactose, catalyzed by the sialyltransferase ST6GAL1, inhibits the binding of galectin-1, a ß-galactoside-binding lectin. We recently reported the potential involvement of galectin-1 and -3 in the pathogenesis of tubal ectopic pregnancy; however, the precise role of galectins and their ligand glycoconjugates remain unclear. Here, we investigated the expression of the genes encoding α2,3- and α2,6-galactoside sialyltransferases (ST3GAL1-6 and ST6GAL1-2) and the localization of sialic acids in the Fallopian tube of women with or without ectopic implantation. ST6GAL1 expression was higher in the mid-secretory phase than the proliferative phase of non-pregnant women (P < 0.0001), whereas ST6GAL1 (P < 0.0001), ST3GAL3 (P = 0.0029), ST3GAL5 (P = 0.0089), and ST3GAL6 (P = 0.0018) were all lower in Fallopian tubes with ectopic implantations. α2,3- and α2,6-sialic acids, however, both remained enriched on the surface of Fallopian tube epithelium. Cigarette smoking, a major risk factor for tubal ectopic pregnancy, was associated with reduced mid-secretory-phase expression of ST6GAL1 (P = 0.0298), but elevated expression of ST3GAL5 (P = 0.0006), an enzyme known to be involved in ciliogenesis. Indeed, sialic acid-containing ciliated inclusion cysts, which are associated with abnormal ciliogenesis, were observed within the epithelium at a higher frequency in women who smoked (P = 0.0177), suggesting that abnormal ciliogenesis is associated with smoking. Thus, cigarette smoking alters sialylation in the Fallopian tube epithelium, and is potentially a source of decreased tubal transport and increased receptivity for blastocyst in the human Fallopian tube. Mol. Reprod. Dev. 83: 1083-1091, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Fallopian Tubes/metabolism , Gene Expression Regulation, Enzymologic , N-Acetylneuraminic Acid/metabolism , Pregnancy, Ectopic/metabolism , Sialyltransferases/biosynthesis , Smoking/adverse effects , Adolescent , Adult , Fallopian Tubes/pathology , Female , Humans , Middle Aged , Pregnancy , Pregnancy, Ectopic/etiology , Pregnancy, Ectopic/pathology , Smoking/metabolism , Smoking/pathology
12.
Exp Eye Res ; 153: 170-177, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27793618

ABSTRACT

The blood-brain barrier in the neonatal brain expresses the monocarboxylate transporter (MCT)-1 rather than the glucose transporter (GLUT)-1, due to the special energy supply during the suckling period. The hyaloid vascular system, consisting of the vasa hyaloidea propria and tunica vasculosa lentis, is a temporary vasculature present only during the early development of mammalian eyes and later regresses. Although the ocular vasculature manifests such a unique developmental process, no information is available concerning the expression of endothelial nutrient transporters in the developing eye. The present immunohistochemical study using whole mount preparations of murine eyes found that the hyaloid vascular system predominantly expressed GLUT1 in the endothelium, in contrast to the brain endothelium. Characteristically, the endothelium in peripheral regions of the neonatal hyaloid vessels displayed a mosaic pattern of MCT1-immunoreactive cells scattered within the GLUT1-expressing endothelium. The proper retinal vessels first developed by sprouting angiogenesis endowed with filopodia, which were absolutely free from the immunoreactivities of GLUT1 and MCT1. The remodeling retinal capillary networks and veins in the surface layer of the retina mainly expressed MCT1 until the weaning period. Immunostaining of MCT1 in the retina revealed fine radicular processes projecting from the endothelium, differing from the MCT1-immunonegative filopodia. These findings suggest that the expression of nutrient transporters in the ocular blood vessels is differentially regulated at a cellular level and that the neonatal eyes provide an interesting model for research on nutrient transporters in the endothelium.


Subject(s)
Eye/growth & development , Glucose Transporter Type 1/biosynthesis , Monocarboxylic Acid Transporters/biosynthesis , Pigment Epithelium of Eye/metabolism , Pregnancy, Animal , Symporters/biosynthesis , Animals , Animals, Newborn , Biological Transport , Female , Immunohistochemistry , Mice , Microscopy, Electron , Models, Animal , Pigment Epithelium of Eye/growth & development , Pigment Epithelium of Eye/ultrastructure , Pregnancy
13.
EMBO Rep ; 15(12): 1297-304, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25378482

ABSTRACT

Intrinsic Notch signaling in intestinal epithelial cells restricts secretory cell differentiation. In gut-associated lymphoid tissue (GALT), stromal cells located beneath the follicle-associated epithelium (FAE) abundantly express the Notch ligand delta-like 1 (Dll1). Here, we show that mice lacking Rbpj-a gene encoding a transcription factor implicated in Notch signaling-in intestinal epithelial cells have defective GALT maturation. This defect can be attributed to the expansion of goblet cells, which leads to the down-regulation of CCL20 in FAE. These data demonstrate that epithelial Notch signaling maintained by stromal cells contributes to the full maturation of GALT by restricting secretory cell differentiation in FAE.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Receptors, Notch/metabolism , Stromal Cells/cytology , Animals , Cell Differentiation/physiology , In Vitro Techniques , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Signal Transduction/physiology , Stromal Cells/metabolism
14.
Am J Physiol Gastrointest Liver Physiol ; 308(3): G188-97, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25394661

ABSTRACT

Bacterial or ingested food-derived short-chain fatty acids (SCFAs) are present in the duodenal lumen. Acetate, the most abundant SCFA in the foregut lumen, is absorbed immediately after ingestion, although the mechanism by which this absorption occurs is not fully understood. We investigated the distribution and function of candidate SCFA transporters in rat duodenum. The Na(+)-coupled monocarboxylate transporter-1 (SMCT1) was localized to the brush border, whereas the pH-dependent monocarboxylate transporter (MCT) 1 and MCT4 were localized to the duodenocyte basolateral membrane. In Ussing chambered duodenal mucosa, luminal acetate dose-dependently increased short-circuit current (Isc) in the presence of serosal bumetanide and indomethacin by a luminal Na(+)-dependent, ouabain-sensitive mechanism. The Isc response was inhibited dose-dependently by the SMCT1 nonsubstrate inhibitor ibuprofen, consistent with net electrogenic absorption of acetate via SMCT1. Other SCFAs and lactate also increased Isc. Furthermore, duodenal loop perfusion of acetate increased portal venous acetate concentration, inhibited by coperfusion of ibuprofen or a MCT inhibitor. Luminal acetate perfusion increased duodenal HCO3 (-) secretion via capsaicin-sensitive afferent nerve activation and cyclooxygenase activity, consistent with absorption-mediated HCO3 (-) secretion. These results suggest that absorption of luminal SCFA via SMCT1 and MCTs increases duodenal HCO3 (-) secretion. In addition to SCFA sensing via free fatty acid receptors, the presence of rapid duodenal SCFA absorption may be important for the suppression of luminal bacterial colonization and implicated in the generation of functional dyspepsia due to bacterial overgrowth.


Subject(s)
Duodenum/metabolism , Fatty Acids, Volatile/metabolism , Intestinal Mucosa/metabolism , Sodium/metabolism , Animals , Biological Transport/drug effects , Hydrogen-Ion Concentration , Intestinal Absorption/drug effects , Intestinal Absorption/physiology , Intestinal Mucosa/drug effects , Male , Models, Animal , Propionates/pharmacology , Rats, Sprague-Dawley
15.
Mol Hum Reprod ; 21(8): 645-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26003810

ABSTRACT

Intense macrophage infiltration is observed during luteolysis in various animals including women; however, we still do not know how macrophage infiltration into the human corpus luteum (CL) during luteolysis is regulated. In this study, we examined the expression, localization and regulation of an important chemokine for the recruitment of monocyte/macrophage lineages, C-C motif ligand 2 (CCL2), in the human CL across the luteal phase and in cultured human luteinized granulosa cells (LGCs), with special reference to the number of infiltrating macrophages and luteal cell function. CCL2 mRNA increased in the non-functional regressing CL during menstruation (P < 0.01), corresponding to an elevated mRNA expression of a macrophage-derived cytokine, tumor necrosis factor (TNF), and an increased number of infiltrating macrophages positively stained with a macrophage marker, CD68. CCL2 protein was immunohistochemically localized to the cytoplasm of granulosa-lutein and theca-lutein cells, and CCL2 mRNA was significantly reduced by hCG both in vivo (P < 0.05) and in vitro (P < 0.01). CCL2 was also down-regulated by luteotrophic prostaglandin (PG) E (P < 0.0001), but up-regulated by luteolytic PGF (P < 0.05) in vitro. Administration of TNF significantly enhanced the CCL2 mRNA expression in cultured LGCs (P < 0.01). A greater abundance of infiltrating macrophages were found around granulosa-lutein cells lacking 3ß-HSD or PGE synthase (PGES) immunostaining. CCL2 mRNA expression was negatively correlated with both HSD3B1 and PGES, suggesting that locally produced progesterone and PGE suppress macrophage infiltration into the CL. Taken together, the infiltration of macrophages in the human CL is regulated by endocrine and paracrine molecules via regulation of the CCL2 expression in luteal cells.


Subject(s)
Chemokine CCL2/metabolism , Corpus Luteum/metabolism , Luteal Cells/cytology , Luteal Cells/metabolism , Luteolysis/genetics , Macrophages/cytology , Macrophages/metabolism , Apoptosis/genetics , Apoptosis/physiology , Cells, Cultured , Corpus Luteum/cytology , Female , Humans , Immunohistochemistry , In Vitro Techniques , Luteolysis/physiology
16.
Histochem Cell Biol ; 143(3): 301-12, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25249350

ABSTRACT

Information concerning the cellular localization of cholecystokinin (CCK)-1 receptors has been discrepant and remained scanty at ultrastructural levels. The present immunohistochemical study at light and electron microscopic levels revealed the distinct localization of CCK1 receptors in visceral organs. Immunohistochemistry by use of a purified antibody against mouse CCK1 receptor was applied to fixed tissue sections of the pancreas, gallbladder, stomach, and intestine of mice. A silver-intensified immunogold method revealed the subcellular localization under electron microscope. The immunoreactivity for CCK1 receptors was selectively found in the basolateral membrane of pancreatic acinar cells and gastric chief cells but was absent in pancreatic islets and gastric D cells. Another intense expression in the gut was seen in the myenteric nerve plexus of the antro-duodenal region and some populations of c-Kit-expressing pacemaker cells in the duodenal musculature. The gallbladder contained smooth muscle fibers with an intense immunoreactivity of CCK1 receptors on cell surfaces. The restricted localization of CCK1 receptors on the basolateral membrane of pancreatic acinar cells and gastric chief cells, along with their absence in the islets of Langerhans and gastric D cells, provides definitive information concerning the regulatory mechanism by circulating CCK. Especially, the subcellular localization in the acinar cells completes the investigation for the detection of circulating CCK by the basolateral membrane.


Subject(s)
Gallbladder/cytology , Pancreas/cytology , Receptor, Cholecystokinin A/analysis , Receptor, Cholecystokinin A/metabolism , Stomach/cytology , Animals , Gallbladder/ultrastructure , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred Strains , Microscopy, Electron , Molecular Sequence Data , Pancreas/ultrastructure , Receptor, Cholecystokinin A/ultrastructure , Stomach/ultrastructure
17.
Cells Tissues Organs ; 200(6): 424-34, 2015.
Article in English | MEDLINE | ID: mdl-26359845

ABSTRACT

Galectin-1 and galectin-3 are abundantly expressed at implantation sites in the uterus, suggesting their involvement in the establishment of pregnancy. In this study, we examined the expression and localization of galectin-1 and galectin-3 in fallopian tubes from nonpregnant women, and in those presenting with tubal ectopic pregnancy. There was no significant difference in the expression of either galectin-1 (LGALS1) or galectin-3 (LGALS3) transcripts in the fallopian tube across the menstrual cycle. Their expressions in the fallopian tube were inversely correlated to each other (r = -0.5134, p < 0.0001) and differentially localized. Galectin-1 protein was abundant in the stroma of nonpregnant fallopian tubes, whereas galectin-3 was mainly localized to the epithelium, notably to the cilia of ciliated cells and the apical cytoplasm of secretory cells. In ectopic pregnancies, LGALS3 expression was significantly reduced (p < 0.0001), but LGALS1 expression did not change when compared to nonpregnant fallopian tubes collected during the mid-secretory phase. The percentage of fallopian tube epithelial cells expressing galectin-3 in cilia tended to be reduced (p = 0.0685), with an accompanying loss of a normal ciliary structure, while nuclear galectin-3 increased (p < 0.05) in ectopic pregnancies. Epithelial immunostaining for galectin-1 tended to be elevated in fallopian tubes from women with ectopic pregnancy. Coculture of human trophoblast origin SW71 cells significantly increased LGALS1 expression in human fallopian tube epithelial OE-E6/E7 cells, suggesting that trophoblast-derived products regulate LGALS1 expression in the oviductal epithelium. These findings imply a differential contribution of galectin-1 and galectin-3 in the homeostasis of human fallopian tubes and in the pathophysiology of ectopic pregnancy.


Subject(s)
Fallopian Tubes/pathology , Galectin 1/analysis , Galectin 3/analysis , Gene Expression Regulation , Pregnancy, Tubal/genetics , Pregnancy, Tubal/pathology , Adolescent , Adult , Cell Line , Fallopian Tubes/metabolism , Female , Galectin 1/genetics , Galectin 3/genetics , Humans , Middle Aged , Pregnancy , Pregnancy, Tubal/blood , Young Adult
18.
Jpn J Vet Res ; 63(3): 107-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26563030

ABSTRACT

Nuclear factor of kappa light polypeptide gene enhancer in B cells (NF-κB) inhibitor zeta (Nfkbiz) is a nuclear inhibitor of NF-κB (IκB) protein that is also termed as molecule possessing ankyrin repeats induced by lipopolysaccharide, interleukin-1-inducible nuclear ankyrin repeat protein, or IκBζ. We found previously that disrupting the Nfkbiz gene resulted in atopic dermatitis-like lesions in mice, suggesting an important role for Nfkbiz in the skin. In this study, we examined the cellular function of Nfkbiz in keratinocytes. Immunohistochemical analyses for Ki-67 revealed that Nfkbiz-/- keratinocytes were hypoproliferative. In skin from Nfkbiz-/- mice, the expression of the keratinocyte differentiation markers K10 and filaggrin were reduced, although that of K14 was unchanged. The terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay revealed that the frequency of apoptosis was comparable between control and Nfkbiz-/- keratinocytes. Interestingly, the subcellular localization of the NF-κB subunits and the transcriptional activity of NF-κB were not changed in Nfkbiz-/- keratinocytes. These findings indicate a novel possible role of Nfkbiz in controlling the proliferation and differentiation of epidermal keratinocytes through NF-κB-independent mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/genetics , Epidermis/physiology , Gene Expression Regulation , Keratinocytes/physiology , Nuclear Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Proliferation , Epidermal Cells , Immunohistochemistry , Mice , Nuclear Proteins/genetics
19.
Br J Nutr ; 110(4): 733-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23308394

ABSTRACT

Brown adipose tissue (BAT) is responsible for cold- and diet-induced thermogenesis, and thereby contributes to the control of whole-body energy expenditure (EE) and body fat content. BAT activity can be assessed by fluoro-2-deoxyglucose (FDG)-positron emission tomography (PET) in human subjects. Grains of paradise (GP, Aframomum melegueta), a species of the ginger family, contain pungent, aromatic ketones such as 6-paradol, 6-gingerol and 6-shogaol. An alcohol extract of GP seeds and 6-paradol are known to activate BAT thermogenesis in small rodents. The present study aimed to examine the effects of the GP extract on whole-body EE and to analyse its relation to BAT activity in men. A total of nineteen healthy male volunteers aged 20-32 years underwent FDG-PET after 2 h of exposure to cold at 19°C with light clothing. A total of twelve subjects showed marked FDG uptake into the adipose tissue of the supraclavicular and paraspinal regions (BAT positive). The remaining seven showed no detectable uptake (BAT negative). Within 4 weeks after the FDG-PET examination, whole-body EE was measured at 27°C before and after oral ingestion of GP extract (40 mg) in a single-blind, randomised, placebo-controlled, crossover design. The resting EE of the BAT-positive group did not differ from that of the BAT-negative group. After GP extract ingestion, the EE of the BAT-positive group increased within 2 h to a significantly greater (P<0·01) level than that of the BAT-negative group. Placebo ingestion produced no significant change in EE. These results suggest that oral ingestion of GP extract increases whole-body EE through the activation of BAT in human subjects.


Subject(s)
Adipose Tissue, Brown/metabolism , Dietary Supplements , Energy Metabolism/drug effects , Plant Extracts/pharmacology , Zingiberaceae/chemistry , Adipose Tissue , Adipose Tissue, Brown/drug effects , Adult , Anthropometry , Calorimetry, Indirect , Cross-Over Studies , Diet , Fluorodeoxyglucose F18 , Guaiacol/analogs & derivatives , Guaiacol/metabolism , Humans , Ketones/chemistry , Ketones/metabolism , Male , Positron-Emission Tomography , Seeds/metabolism , Single-Blind Method , Temperature , Time Factors , Young Adult
20.
Biomed Res ; 44(3): 81-95, 2023.
Article in English | MEDLINE | ID: mdl-37258205

ABSTRACT

Gastrin and CCK (cholecystokinin), gut hormones first secreted after postprandial stages, share the C-terminal amino acids and some types of receptors to be stimulated. Both types of hormone-secreting cells are typical open-type cells which detect foods and their digested elements in the lumen and regulate the secretion of gastric acid and digestive enzymes, gut motility, and satiety. Gastrin cell granules are characterized by their heterogenous ultrastructure within the cell, while CCK cell granules show a uniform ultrastructural figure. Gastrin cells are equipped with peptone receptor GPR92, amino acid receptor GPRC6A, and a Ca-sensing receptor. In addition to nutrient receptors, the release of CCK is regulated by a unique negative feedback mechanism. Development of an antibody for CCK-specific receptor (CCK-1R) has revealed its exact localization throughout the body, but specific antibodies against CCK-2R remain unavailable. Gastrin affects differentiation and proliferation-including cancer cells, while CCK possesses trophic effects to target tissues. CCK is a peripheral satiety signal and acts either via the vagus or directly on the dorsal medulla via CCK-1R. In this review, endocrine cells secreting these unique and so-called old gut hormones are described on a morphological basis.


Subject(s)
Cholecystokinin , Gastrins , Cholecystokinin/metabolism , Cholecystokinin/pharmacology , Gastrins/metabolism , Gastrins/pharmacology , Receptors, Cholecystokinin/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL