Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gen Comp Endocrinol ; 331: 114167, 2023 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-36402245

RESUMEN

Ghrelin is a gut-derived peptide with several physiological functions, including feeding, gastrointestinal motility, and hormonal secretion. Recently, a host defense peptide, liver-expressed antimicrobial peptide-2 (LEAP2), was reported as an endogenous antagonist of growth hormone secretagogue receptor (GHS-R). The physiological relevance of the molecular LEAP2-GHS-R interaction in mammals has been explored; however, studies on non-mammals are limited. Here, we report the identification and functional characterization of ghrelin and its related molecules in Western clawed frog (Xenopus tropicalis), a known model organism. We first identified cDNA encoding X. tropicalis ghrelin and GHS-R. RT-qPCR revealed that ghrelin mRNA expression was most abundant in the stomach. GHS-R mRNA was widely distributed in the brain and peripheral tissues, and a relatively strong signal was observed in the stomach and intestine. In addition, LEAP2 was mainly expressed in intestinal tissues at higher levels than in the liver. In functional analysis, X. tropicalis ghrelin and human ghrelin induced intracellular Ca2+ mobilization with EC50 values in the low nanomolar range in CHO-K1 cells expressing X. tropicalis GHS-R. Furthermore, ghrelin-induced GHS-R activation was antagonized with IC50 values in the nanomolar range by heterologous human LEAP2. We also validated the expression of ghrelin and feeding-related factors under fasting conditions. After 2 days of fasting, no changes in ghrelin mRNA levels were observed in the stomach, but GHS-R mRNA levels were significantly increased, associated with significant downregulation of nucb2. In addition, LEAP2 upregulation was observed in the duodenum. These results provide the first evidence that LEAP2 functions as an antagonist of GHS-R in the anuran amphibian X. tropicalis. It has also been suggested that the ghrelin/GHS-R/LEAP2 system may be involved in energy homeostasis in X. tropicalis.


Asunto(s)
Ghrelina , Receptores de Ghrelina , Animales , Cricetinae , Humanos , Ghrelina/genética , Ghrelina/metabolismo , Xenopus/metabolismo , Receptores de Ghrelina/metabolismo , Cricetulus , Clonación Molecular , ARN Mensajero
2.
PLoS Genet ; 16(3): e1008664, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32203505

RESUMEN

Motile cilia/flagella are essential for swimming and generating extracellular fluid flow in eukaryotes. Motile cilia harbor a 9+2 arrangement consisting of nine doublet microtubules with dynein arms at the periphery and a pair of singlet microtubules at the center (central pair). In the central system, the radial spoke has a T-shaped architecture and regulates the motility and motion pattern of cilia. Recent cryoelectron tomography data reveal three types of radial spokes (RS1, RS2, and RS3) in the 96 nm axoneme repeat unit; however, the molecular composition of the third radial spoke, RS3 is unknown. In human pathology, it is well known mutation of the radial spoke head-related genes causes primary ciliary dyskinesia (PCD) including respiratory defect and infertility. Here, we describe the role of the primary ciliary dyskinesia protein Rsph4a in the mouse motile cilia. Cryoelectron tomography reveals that the mouse trachea cilia harbor three types of radial spoke as with the other vertebrates and that all triplet spoke heads are lacking in the trachea cilia of Rsph4a-deficient mice. Furthermore, observation of ciliary movement and immunofluorescence analysis indicates that Rsph4a contributes to the generation of the planar beating of motile cilia by building the distal architecture of radial spokes in the trachea, the ependymal tissues, and the oviduct. Although detailed mechanism of RSs assembly remains unknown, our results suggest Rsph4a is a generic component of radial spoke heads, and could explain the severe phenotype of human PCD patients with RSPH4A mutation.


Asunto(s)
Cilios/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Axonema/genética , Axonema/metabolismo , Cilios/genética , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/metabolismo , Proteínas del Citoesqueleto/genética , Dineínas/metabolismo , Femenino , Flagelos/genética , Flagelos/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microtúbulos/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética
3.
Gen Comp Endocrinol ; 323-324: 114031, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35331740

RESUMEN

Motilin, a peptide hormone consisting of 22 amino acid residues, was identified in the duodenum of pigs in the 1970s. It is known to induce gastrointestinal contractions during the interdigestive state in mammals. Although the motilin gene has been identified in various animal species, it has not been studied in amphibians. Here, we identified the motilin gene in the Japanese fire bellied newt (Cynops pyrrhogaster), and conducted an analysis of tissue distribution, morphological observations, and physiological experiments. The deduced mature newt motilin comprises 22 amino acid residues, like in mammals and birds. The C-terminus of the newt motilin showed high homology with motilin from other species compared to the N-terminus region, which is considered the bioactive site. Motilin mRNA expression in newts was abundant in the upper small intestine, with notably high motilin mRNA expression found in the pancreas. Motilin-producing cells were found in the mucosal layer of the upper small intestine and existed as two cell types: open-and closed-type cells. Motilin-producing cells in the pancreas were also found to produce insulin but not glucagon. Newt motilin stimulated gastric contractions but not in other parts of the intestines in vitro, and motilin-induced gastric contraction was significantly inhibited by treatment with atropine, a muscarinic acetylcholine receptor antagonist. These results indicate that motilin is also present in amphibians, and that its gastrointestinal contractile effects are conserved in mammals, birds, and amphibians. Additionally, we demonstrated for the first time the existence of pancreatic motilin, suggesting that newt motilin has an additional unknown physiological role.


Asunto(s)
Motilina , Salamandridae , Aminoácidos , Animales , Aves/metabolismo , Motilidad Gastrointestinal , Mamíferos/metabolismo , Motilina/farmacología , Contracción Muscular , ARN Mensajero/metabolismo , Salamandridae/genética , Salamandridae/metabolismo , Porcinos
4.
Gen Comp Endocrinol ; 327: 114074, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35700795

RESUMEN

Cholecystokinin (CCK) is a peptide hormone mainly secreted by small intestinal endocrine I-cells and functions as a regulator of gallbladder contraction, gastric emptying, gastrointestinal (GI) motility, and satiety. The cellular effects of CCK in these peripheral tissues are predominantly mediated via CCK-A receptors which are found in smooth muscles, enteric neurons, and vagal afferent neurons in humans and animal models. Although various functions of CCK have been reported to be neurally mediated, it can also stimulate contraction via the CCK receptor on the smooth muscle. However, the entire underlying neural and cellular mechanisms involved in CCK-induced GI contractions are not clearly understood. Here, we first determined the cDNA and amino acid sequences of CCK and CCK-A receptor along with the distributions of cck mRNA and CCK-producing cells in house musk shrew (Suncus murinus, the laboratory strain named as suncus) and examined the mechanism of CCK-induced contraction in the GI tract. Mature suncus CCK-8 was identical to other mammalian species tested here, and suncus CCK-A receptor presented high nucleotide and amino acid homology with that of human, dog, mouse, and rat, respectively. Suncus CCK mRNA and CCK-producing cells were found mainly in small intestine and colon. In the organ bath study, CCK-8 induced dose-dependent contractions in the suncus stomach, duodenum, and jejunum, and these contractions were inhibited by atropine and CCK-A receptor antagonist. These results suggest that CCK-8-induced contraction is mediated in the myenteric cholinergic neural network and that CCK-A receptor is partly responsible for CCK-8-induced contractions. This study indicates that suncus is a useful animal model to study the functions of CCK involved in GI motility.


Asunto(s)
Colecistoquinina , Receptor de Colecistoquinina A , Musarañas , Animales , Colecistoquinina/genética , Clonación Molecular , Perros , Motilidad Gastrointestinal , Humanos , Ratones , Contracción Muscular , ARN Mensajero/genética , Ratas , Receptor de Colecistoquinina A/genética , Musarañas/genética , Sincalida/farmacología
5.
Mol Biol Rep ; 48(7): 5513-5518, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34302584

RESUMEN

Pyridoxine (PN), one of the vitamers of vitamin B6, plays an important role in the maintenance of epidermal function and is used to treat acne and rough skin. Clinical studies have revealed that PN deficiency causes skin problems such as seborrheic dermatitis and stomatitis. However, the detailed effects of PN and its mechanism of action in epidermal function are poorly understood. In this study, we examined the effects of PN on epidermal function in normal human epidermal keratinocytes and found that PN specifically causes an increase in the expression of profilaggrin mRNA, among marker genes of terminal epidermal differentiation. In addition, PN treatment caused an increase in the production of filaggrin protein in a concentration-dependent manner. Treatment with P2x purinoceptor antagonists, namely, pyridoxal phosphate-6-azo (benzene-2,4-disulfonic acid) tetrasodium salt hydrate and TNP-ATP hydrate, induced an increase in the filaggrin protein levels. Moreover, we showed that elevated filaggrin production induced upon PN treatment was suppressed by ATP (known as P2x purinoceptor agonist). This study is the first to report that PN causes an increase in filaggrin transcription and production, and these results suggest that PN-induced filaggrin production may be a useful target as a daily care component in atopic dermatitis, wherein filaggrin levels are specifically reduced.


Asunto(s)
Proteínas de Filamentos Intermediarios/genética , Queratinocitos/metabolismo , Piridoxina/metabolismo , Células Cultivadas , Epidermis/metabolismo , Proteínas Filagrina , Regulación de la Expresión Génica , Humanos , Piridoxina/farmacología
6.
Cell Biol Int ; 44(1): 62-69, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31293061

RESUMEN

Motilin, a 22-amino-acid peptide produced in the upper small intestine, induces strong gastric contraction in fasted state. In many rodents, motilin and its cognate receptors exist as pseudogenes, which has delayed motilin research in the past decades. Recently, the house musk shrew (Suncus murinus) was developed as a useful model for studying motilin and gastrointestinal motility. However, due to a lack of motilin-producing cell lines and difficulties in culturing small intestinal cells, the regulatory mechanisms of motilin secretion and its messenger RNA (mRNA) transcription have remained largely unclear. In this study, we generated small intestinal organoids from S. murinus for the first time. Using methods similar to mouse organoid generation, we found crypt-like budding structures 3 days after isolating intestinal tissues. The organoids grew gradually with time. In addition, the generated organoids were able to be passaged and maintained for 6 months or longer. Motilin messenger RNA (mRNA) and immunopositive cells were observed in both S. murinus intestinal organoids and primary tissues. This is the first report of intestinal organoids in S. murinus, and our results suggest that S. murinus intestinal organoids could be useful for analyzing motilin secretion and transcription.

7.
Sensors (Basel) ; 20(5)2020 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-32143461

RESUMEN

Magnetization reversal in a Wiegand wire induces a pulse voltage in the pickup coil around the wire, called the Wiegand pulse. The Wiegand sensor features the Wiegand wire and the pickup coil. The amplitude and width of the Wiegand pulse are independent of the frequency of the magnetic-field change. The pulse is generated by the Wiegand sensor, which facilitates the use of the Wiegand sensor as a power supply for equipment without batteries. In order to meet the power consumption requirements, it is necessary to maximize the energy of the pulse signal from the Wiegand sensor, without changing the external field conditions. The distributions of the magnetic field generated from the applied magnet in air and in the Wiegand wire were simulated before the experiments. Simulation predicted an increase in the magnetic flux density through the center of the Wiegand wire. This study determined that the magnetic flux density through the center of the Wiegand wire, the position of the pickup coil, and the angle between the Wiegand sensor and the magnetic induction line were the main factors that affected the energy of a Wiegand pulse. The relationship between these factors and the energy of the Wiegand pulse were obtained.

8.
Cell Tissue Res ; 376(3): 401-412, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30680460

RESUMEN

Lysozyme is one of the most prominent antimicrobial peptides and has been identified from many mammalian species. However, this enzyme has not been studied in the order Insectivora, which includes the most primitive placental mammals. Here, we done the lysozyme cDNA from Suncus murinus (referred to as suncus, its laboratory name) and compare the predicted amino acid sequence to those from other mammalian species. Quantitative PCR analysis revealed a relatively higher expression of this gene in the spleen and gastrointestinal tract of suncus. The lysozyme-immunopositive (ip) cells were found mainly in the red pulp of the spleen and in the mucosa of the whole small intestine, including the follicle-associated epithelium and subepithelial dome of Peyer's patches. The lysozyme-ip cells in the small intestine were mostly distributed in the intestinal crypt, although lysozyme-expressing cells were found not only in the crypt but also in the villi. On the other hand, only a few lysozyme-ip cells were found in the villi and some granules showing intense fluorescence were located toward the lumen. As reported for other mammals, Ki67-ip cells were localized in the crypt and did not co-localize with the lysozyme-ip cells. Moreover, fasting induced a decrease in the mRNA levels of lysozyme in the intestine of suncus. In conclusion, we firstly identified the lysozyme mRNA sequence, clarified expression profile of lysozyme transcripts in suncus and found a unique distribution of lysozyme-producing cells in the suncus intestine.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/química , Muramidasa/química , Musarañas/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/aislamiento & purificación , Péptidos Catiónicos Antimicrobianos/metabolismo , Mucosa Intestinal/enzimología , Muramidasa/genética , Muramidasa/aislamiento & purificación , Muramidasa/metabolismo , Ganglios Linfáticos Agregados/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Bazo/metabolismo , Distribución Tisular , Transcriptoma
9.
Can J Physiol Pharmacol ; 97(10): 909-915, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31100203

RESUMEN

Molecular hydrogen (H2) showed protection against various kinds of oxidative-stress-related diseases. First, it was reported that the mechanism of therapeutic effects of H2 was antioxidative effect due to inhibition of the most cytotoxic reactive oxygen species, hydroxy radical (•OH). However, after chronic administration of H2 in drinking water, oxidative-stress-induced nerve injury is significantly attenuated even in the absence of H2. It suggests indirect signaling of H2 and gastrointestinal tract is involved. Indirect effects of H2 could be tested by giving H2 water only before nerve injury, as preconditioning. For example, preconditioning of H2 for certain a period (∼7 days) in Parkinson's disease model mice shows significant neuroprotection. As the mechanism of indirect effect, H2 in drinking water induces ghrelin production and release from the stomach via ß1-adrenergic receptor stimulation. Released ghrelin circulates in the body, being transported across the blood-brain barrier, activates its receptor, growth-hormone secretagogue receptor. H2-induced upregulation of ghrelin mRNA is also shown in ghrelin-producing cell line, SG-1. These observations help with understanding the chronic effects of H2 and raise intriguing preventive and therapeutic options using H2.


Asunto(s)
Ghrelina/metabolismo , Hidrógeno/administración & dosificación , Enfermedades Neurodegenerativas/terapia , Neuroprotección/efectos de los fármacos , Traumatismos de los Nervios Periféricos/terapia , Animales , Barrera Hematoencefálica/metabolismo , Modelos Animales de Enfermedad , Ingestión de Líquidos , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Ghrelina/sangre , Humanos , Enfermedades Neurodegenerativas/sangre , Estrés Oxidativo/efectos de los fármacos , Traumatismos de los Nervios Periféricos/sangre , Receptores de Ghrelina/metabolismo , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Agua/química
10.
Gen Comp Endocrinol ; 274: 106-112, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30677392

RESUMEN

Motilin (MLN), a 22-amino-acid peptide hormone, is generally present in the mucosa of the upper gastrointestinal (GI) tract, mainly the duodenum of mammals, and it regulates GI motility, especially that related to interdigestive migrating contraction. However, MLN and its receptor are absent in mice and rats, and MLN does not cause any mechanical responses in the rat and mouse GI tracts. The guinea-pig is also a rodent, but expression of the MLN gene in the guinea-pig has been reported. In the present study, two guinea-pig MLNs, FIPIFTYSELRRTQEREQNKGL found in the Ensemble Genome Database (gpMLN-1) and FVPIFTYSELRRTQEREQNKRL reported by Xu et al. (2001) (gpMLN-2), were synthesized, and their biological activities were evaluated in the rabbit duodenum and guinea-pig GI tract in vitro. Both gpMLNs showed contractile activity in longitudinal muscle strips of the rabbit duodenum. The EC50 values of gpMLN-1 and gpMLN-2 were slightly higher than that of human MLN (hMLN), but the maximum contractions were as same as that of hMLN. Treatment with GM109 and hMLN-induced receptor desensitization decreased the contractile activity of both gpMLNs, indicating that the two gpMLN candidates are able to activate the MLN receptor (MLN-R) of the rabbit duodenum. In guinea-pig GI preparations, hMLN and gpMLNs did not show any mechanical responses in circular muscle strips from the gastric antrum or in longitudinal strips of the duodenum, ileum and colon although acetylcholine and 1,1-dimethyl-4-phenylpiperazinium (DMPP) caused definite mechanical responses. The DMPP-induced neural responses in the gastric circular muscle and ileal longitudinal muscles were not modified by gpMLN-1. Even in the gastric and ileal strips with intact mucosa, no mechanical responses were seen with either of the gpMLNs. Furthermore, RT-PCR using various primer sets failed to amplify the gpMLN-2 mRNA. In conclusion, gpMLNs including one that was already reported and the other that was newly found in a database were effective to the rabbit MLN-R, whereas they did not cause any contractions or modification of neural responses in the guinea-pig GI tract, indicating that the MLN system is vestigial and not functional in regulation of GI motility in the guinea-pig as well as in other rodents such as rats and mice.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/fisiología , Motilina/farmacología , Acetilcolina/farmacología , Animales , Duodeno/efectos de los fármacos , Duodeno/fisiología , Femenino , Tracto Gastrointestinal/efectos de los fármacos , Cobayas , Humanos , Técnicas In Vitro , Masculino , Motilina/genética , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Conejos , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de Neuropéptido/metabolismo
11.
Cell Biol Int ; 41(3): 320-327, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28067437

RESUMEN

Ghrelin is abundantly produced in the stomach. Here, we found that glutamate decreased ghrelin expression and release in ghrelin-producing cells, and decreased levels of food intake and plasma acyl-ghrelin in mice. Treatment with siRNA of G protein-coupled receptor, family C, group 5, member B (GPRC5B) in ghrelin-producing cell lines completely blocked the effect of glutamate-induced ghrelin suppression. In addition, glutamate inhibited ghrelin release via the extracellular signal-regulated kinase (ERK) activity pathway, and stimulated CREB2 mRNA expression in ghrelin-producing cell lines. These results suggest that glutamate inhibits ghrelin release via ERK-CREB2 pathway. These results suggest that the GPRC5B-ERK-CREB2 pathway is involved in the inhibition of ghrelin expression and secretion in ghrelin cells.


Asunto(s)
Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/metabolismo , Ghrelina/metabolismo , Ácido Glutámico/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
12.
Endocr J ; 64(Suppl.): S27-S29, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28652540

RESUMEN

Ghrelin is a peptide hormone with a unique structure comprising a medium chain fatty acid modification. Ghrelin cells are known to be abundantly localized in the gastric mucosa and are released into the blood stream to exert their multifunctional physiological effects. To elucidate the regulatory mechanisms of ghrelin secretion and acyl-modification, we developed novel ghrelin-producing cell lines. Using ghrelinoma cell lines, we focused on the mechanisms of ghrelin secretion and found that several GPCRs were highly expressed in ghrelin cells. Then, we showed that noradrenaline treatment stimulated ghrelin secretion via ß1-adrenergic receptor, and fasting-induced ghrelin elevation was completely inhibited by the ß1-adrenergic receptor antagonist in mice. In addition, we demonstrated that long chain fatty acids, glucose, and L-glutamate significantly inhibited ghrelin secretion. Furthermore, we recently revealed that the genes involved in fatty acid synthesis and long chain fatty acid metabolism were expressed in ghrelin cells, and that CPT-1 inhibitor treatment dramatically decreased the levels of acyl-modified ghrelin. Here, we introduce the current knowledge of the mechanisms involving ghrelin secretion and its acyl-modification.


Asunto(s)
Ghrelina/metabolismo , Acilación , Animales , Línea Celular Tumoral , Mucosa Gástrica/metabolismo , Ratones , Ratones Transgénicos , Transducción de Señal
13.
Endocr J ; 64(Suppl.): S11-S14, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28652536

RESUMEN

Ghrelin, a peptide hormone produced in the stomach, has been known to be involved in the regulation of gastric contraction in humans and rodents. To elucidate the detailed mechanisms of ghrelin on gastric contractions, we used Suncus murinus, a recently established small animal model for gastrointestinal motility. S. murinus produces motilin, a family peptide of ghrelin, and its stomach anatomy and physiological patterns of gastric contractions, in fed and fasted states, are closely similar to humans. Ghrelin administration in phase II, and latter half of phase I, of the migrating motor contractions (MMC) enhanced gastric motility in S. murinus. In addition, we showed that ghrelin and motilin coordinately stimulated strong gastric contractions in vitro and in vivo. We also demonstrated that a pretreatment with a ghrelin antagonist, D-Lys3-GHRP6, inhibited the effects of motilin-induced gastric contractions, and a γ-aminobutyric acid (GABA) antagonist reversed this inhibition. Our results suggest that ghrelin is essential for motilin-induced gastric contractions and that ghrelin-mediated GABAergic neurons are involved in this neural pathway.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Ghrelina/farmacología , Musarañas , Estómago/efectos de los fármacos , Animales , Antagonistas del GABA/farmacología , Motilina/farmacología , Contracción Muscular/efectos de los fármacos , Oligopéptidos/farmacología
14.
Appl Environ Microbiol ; 82(19): 5806-14, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27451451

RESUMEN

UNLABELLED: Asymptomatic infant carriers of toxigenic Clostridium difficile are suggested to play a role in the transmission of C. difficile infection (CDI) in adults. However, the mode of C. difficile carriage in infants remains to be fully elucidated. We investigated longitudinal changes in carriage rates, counts, and strain types of toxigenic C. difficile in infants. Stools collected from 111 healthy infants in Belgium periodically from birth until the age of 6 months were examined by quantitative PCR targeting 16S rRNA and toxin genes. Toxigenic C. difficile was detected in 18 of 111 infants (16%) in the period up to the age of 6 months. The carriage rate of toxigenic C. difficile remained below 5% until the age of 3 months. The carriage rate increased to 13% 1 week after weaning (average age, 143 days) and reached 16% at the age of 6 months. Counts of toxigenic C. difficile bacteria ranged from 10(4) to 10(8) cells/g of stool. Notably, two infants retained >10(8) cells/g of stool for at least several weeks. Average counts in the 18 infants hovered around 10(7) cells/g of stool from the age of 3 days until the age of 6 months, showing no age-related trend. Genotyping of toxigenic C. difficile isolates from the 18 infants revealed that 11 infants each retained a particular monophyletic strain for at least a month. The genotype most frequently identified was the same as that frequently identified in symptomatic adult CDI patients. Thus, toxigenic C. difficile strains-potential causes of CDI in adults-colonized the infants' intestines. IMPORTANCE: Our study provides longitudinal data on counts and strain types of toxigenic C. difficile in infants. We found that considerable numbers of toxigenic C. difficile bacteria colonized the infants' intestines. The results of strain typing suggest that toxigenic C. difficile carried by healthy infants could be potentially pathogenic to adults. These results and findings are informative not only for ecological studies but also for efforts to prevent or control the spread of CDI in adults.


Asunto(s)
Portador Sano/epidemiología , Clostridioides difficile/fisiología , Genotipo , Toxinas Bacterianas/genética , Bélgica/epidemiología , Portador Sano/microbiología , Clostridioides difficile/clasificación , Clostridioides difficile/genética , Recuento de Colonia Microbiana , Electroforesis Capilar , Heces/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Estudios Longitudinales , Masculino , ARN Ribosómico 16S/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Tiempo
15.
Cell Tissue Res ; 366(3): 721-731, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27590887

RESUMEN

The adenohypophysis is formed from the oral ectoderm and consists of the pars distalis (PD), pars intermedia, and pars tuberalis (PT). The mechanisms of PD development have been extensively studied, and the cellular differentiation of the PD is well understood. However, the morphogenesis and differentiation of the PT are still unclear, and the genes expressed during PT development remain largely unknown. We have explored genes specifically expressed in the PT during embryonic development and analyzed their spatiotemporal expression patterns. Microarray analysis of laser-captured PT and PD tissues obtained from chick embryos on embryonic day 10 (E10.0) has shown high expression of Cytokine-like 1 (CYTL1) and Gap junction protein alpha 5 (GJA5) genes in the PT. Detailed analysis of these spatiotemporal expression patterns during chick embryo development by in situ hybridization has revealed that CYTL1 mRNA first appears in the lateral head ectoderm and ventral head ectoderm at E1.5. The expression of CYTL1 moves into Rathke's pouch at E2.5 and is then localized in the PT primordium where it is continuously expressed until E12.0. GJA5 mRNA is transiently detected in the PT primordium from E6.0 to E12.0, whereas its expression is not detected in the PD during development. Thus, these genes might be involved in the regulation mechanisms of PT development and could be useful markers for PT development.


Asunto(s)
Biomarcadores/metabolismo , Conexinas/genética , Citocinas/genética , Ectodermo/embriología , Ectodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Morfogénesis/genética , Animales , Embrión de Pollo , Conexinas/metabolismo , Citocinas/metabolismo , Desarrollo Embrionario/genética , Estudios de Asociación Genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína alfa-5 de Unión Comunicante
16.
Zoolog Sci ; 33(5): 497-504, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27715422

RESUMEN

Ghrelin was first isolated from human and rat as an endogenous ligand for the growth hormone secretagogue receptor (GHS-R). In the present study, we determined the ghrelin cDNA sequence of the common marmoset (Callithrix jacchus), a small-bodied New World monkey, and investigated the distribution of ghrelin-producing cells in the gastrointestinal tract and localization profiles with somatostatin-producing cells. The marmoset ghrelin cDNA coding region was 354 base pairs, and showed high homology to that in human, rhesus monkey, and mouse. Marmoset ghrelin consists of 28 amino acids, and the N-terminal region is highly conserved as found in other mammalian species. Marmoset preproghrelin and mature ghrelin have 86.3% and 92.9% homology, respectively, to their human counterparts. Quantitative RT-PCR analysis showed that marmoset ghrelin mRNA is highly expressed in the stomach, but it is not detected in other tissues of the gastrointestinal tract. In addition, a large number of ghrelin mRNA-expressing cells and ghrelin-immunopositive cells were detected in the mucosal layer of the stomach, but not in the myenteric plexus. Moreover, all the ghrelin cells examined in the stomach were observed to be closed-type. Double staining showed that somatostatin-immunopositive cells were not co-localized with ghrelin-producing cells; however, a subset of somatostatin-immunopositive cells is directly adjacent to ghrelin-immunopositive cells. These findings suggest that the distribution of ghrelin cells in marmoset differs from that in rodents, and thus the marmoset may be a more useful model for the translational study of ghrelin in primates. In conclusion, we have clarified the expression and cell distribution of ghrelin in marmoset, which may represent a useful model in translational study.


Asunto(s)
Callithrix/metabolismo , Clonación Molecular , Tracto Gastrointestinal/citología , Ghrelina/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Callithrix/genética , ADN/genética , ADN Complementario/química , ADN Complementario/genética , ADN Complementario/metabolismo , Tracto Gastrointestinal/metabolismo , Regulación de la Expresión Génica/fisiología , Ghrelina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Especificidad de la Especie
17.
Dig Dis Sci ; 61(6): 1501-11, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26860510

RESUMEN

BACKGROUND: Gastric acidification inhibits motilin-induced gastric phase III contractions. However, the underlying mechanism has not been thoroughly investigated. Here, we studied the inhibitory mechanism by gastric acidification on motilin-induced contraction in Suncus murinus (S. murinus). METHODS: We measured interdigestive gastric phase III contractions in conscious, freely moving S. murinus, and examined the inhibitory effect of gastric acidification on motilin action and the involvement of the vagus nerve and transient receptor potential vanilloid receptor 1 (TRPV1) in the inhibitory mechanism. RESULTS: A bolus injection of motilin evoked phase III-like contractions during intravenous infusion of saline. Intragastric acidification (pH 1.5-2.5) inhibited motilin-induced phase III contractions in a pH-dependent manner and significantly decreased the motility index at a pH below 2.0. In contrast, intraduodenal acidification (pH 2.0) failed to inhibit motilin-induced contractions. Vagotomy significantly alleviated the suppression of motilin-induced gastric contractions under acidic conditions (pH 2.0), suggesting vagus nerve involvement. Moreover, intragastric acidification (pH 2.0) significantly increased the number of c-Fos-positive cells in the nucleus tractus solitarii. In vagotomized S. murinus, the number of c-Fos-positive cells did not change, even under gastric acidification conditions. TRPV1 mRNA was highly expressed in the muscle and mucosal regions of the antrum and the nodose ganglion, whereas was not detected in the upper small intestine. Capsazepin, a TRPV1 antagonist, completely rescued the inhibitory effect of gastric acidification. CONCLUSIONS: Gastric acidification in S. murinus inhibits motilin-induced contractions, a finding similar to results observed in humans, while TRPV1-expressing vagus nerves play a role in the inhibitory mechanism.


Asunto(s)
Motilidad Gastrointestinal/efectos de los fármacos , Motilina/farmacología , Estómago/fisiología , Canales Catiónicos TRPV/metabolismo , Nervio Vago/metabolismo , Animales , Femenino , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Musarañas , Canales Catiónicos TRPV/genética
18.
Gen Comp Endocrinol ; 233: 53-62, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27179882

RESUMEN

Motilin, a peptide hormone produced in the upper intestinal mucosa, plays an important role in the regulation of gastrointestinal (GI) motility. In the present study, we first determined the cDNA and amino acid sequences of motilin in the Japanese quail and studied the distribution of motilin-producing cells in the gastrointestinal tract. We also examined the motilin-induced contractile properties of quail GI tracts using an in vitro organ bath, and then elucidated the mechanisms of motilin-induced contraction in the proventriculus and duodenum of the quail. Mature quail motilin was composed of 22 amino acid residues, which showed high homology with chicken (95.4%), human (72.7%), and dog (72.7%) motilin. Immunohistochemical analysis showed that motilin-immunopositive cells were present in the mucosal layer of the duodenum (23.4±4.6cells/mm(2)), jejunum (15.2±0.8cells/mm(2)), and ileum (2.5±0.7cells/mm(2)), but were not observed in the crop, proventriculus, and colon. In the organ bath study, chicken motilin induced dose-dependent contraction in the proventriculus and small intestine. On the other hand, chicken ghrelin had no effect on contraction in the GI tract. Motilin-induced contraction in the duodenum was not inhibited by atropine, hexamethonium, ritanserin, ondansetron, or tetrodotoxin. However, motilin-induced contractions in the proventriculus were significantly inhibited by atropine and tetrodotoxin. These results suggest that motilin is the major stimulant of GI contraction in quail, as it is in mammals and the site of action of motilin is different between small intestine and proventriculus.


Asunto(s)
Coturnix/genética , Motilidad Gastrointestinal/genética , Motilina/genética , Animales , Clonación Molecular , Coturnix/fisiología , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Motilidad Gastrointestinal/fisiología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Ghrelina/farmacología , Íleon/efectos de los fármacos , Íleon/metabolismo , Motilina/farmacología , Motilina/fisiología , Contracción Muscular/efectos de los fármacos , Contracción Muscular/genética , Proventrículo/efectos de los fármacos , Proventrículo/metabolismo , Proventrículo/fisiología , Homología de Secuencia
20.
Biochem Biophys Res Commun ; 462(3): 263-8, 2015 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-25957475

RESUMEN

Motilin and ghrelin are gastrointestinal hormones that stimulate the migrating motor complex (MMC) of gastrointestinal motility during the fasting state. In this study, we examined the effect of motilin and ghrelin on pepsinogen secretion in anesthetized suncus (house musk shrew, Suncus murinus), a ghrelin- and motilin-producing mammal. By using a gastric lumen-perfusion system, we found that the intravenous administration of carbachol and motilin stimulated pepsinogen secretion, the latter in a dose-dependent manner, whereas ghrelin had no effect. We then investigated the pathways of motilin-induced pepsinogen secretion using acetylcholine receptor antagonists. Treatment with atropine, a muscarinic acetylcholine receptor antagonist, completely inhibited both carbachol and motilin-induced pepsinogen secretion. Motilin-induced pepsinogen secretion was observed in the vagotomized suncus. This is the first report demonstrating that motilin stimulates pepsinogen secretion, and suggest that this effect occurs through a cholinergic pathway in suncus.


Asunto(s)
Motilina/farmacología , Motilina/fisiología , Pepsinógeno A/metabolismo , Musarañas/fisiología , Animales , Atropina/farmacología , Carbacol/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Ghrelina/administración & dosificación , Inyecciones Intravenosas , Masculino , Motilina/administración & dosificación , Antagonistas Muscarínicos/farmacología , Vagotomía
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA