Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nutr Neurosci ; 23(8): 600-612, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30353787

RESUMEN

Lingonberries (LB) have been shown to have beneficial metabolic effects, which is associated with an altered gut microbiota. This study investigated whether the LB-induced improvements were associated with altered gut- and neuroinflammatory markers, as well as cognitive performance in ApoE-/- mice fed high-fat (HF) diets. Whole LB, as well as two separated fractions of LB were investigated. Eight-week-old male ApoE-/- mice were fed HF diets (38% kcal) containing whole LB (wLB), or the insoluble (insLB) and soluble fractions (solLB) of LB for 8 weeks. Inclusion of wLB and insLB fraction reduced weight gain, reduced fat deposition and improved glucose response. Both wLB and insLB fraction also changed the caecal microbiota composition and reduced intestinal S100B protein levels. The solLB fraction mainly induced weight loss in the mice. There were no significant changes in spatial memory, but significant increases in synaptic density in the hippocampus were observed in the brain of mice-fed wLB and insLB. Thus, this study shows that all lingonberry fractions counteracted negative effects of HF feedings on metabolic parameters. Also, wLB and insLB fraction showed to potentially improve brain function in the mice.


Asunto(s)
Encéfalo/efectos de los fármacos , Encefalitis/prevención & control , Gastritis/prevención & control , Microbioma Gastrointestinal/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Vaccinium vitis-Idaea , Animales , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/efectos de los fármacos , Dieta Alta en Grasa , Ácidos Grasos Volátiles , Metabolismo de los Lípidos , Masculino , Ratones Noqueados para ApoE , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Sinapsis/efectos de los fármacos
2.
Cell Tissue Res ; 376(2): 189-197, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30666535

RESUMEN

In this study, a novel subset of doublecortin-like kinase 1 (DCLK1)-immunoreactive (IR) tuft cells that also contain serotonin (5-hydroxytryptamine, 5HT) is described, in terms of their number, regional distribution, possible synthesis or reuptake of 5HT and proximity to 5-HT-containing enterochromaffin (EC) cells. The small intestine from C57BL/6J mice was divided into five segments while the large intestine was kept undivided. Double immunostaining was used to estimate numbers and topographic distribution of 5HT-IR (DCLK1/5HT) tuft cells and their possible expression of tryptophan hydroxylase (TPH) and serotonin transporter (SERT). Also, possible contacts between tuft cells and 5HT-IR EC cells were studied. In the small intestine, up to 80% of all tuft cells were identified as DCLK1/5HT-IR; in the large intestine, such cells were rare. The highest number of DCLK1/5HT-IR cells was found in the upper small intestine. The numbers of DCLK1/5HT-IR cells gradually decreased distally. DCLK1-IR tuft cells were not found to contain TPH, the rate-limiting enzyme in 5HT synthesis. SERT, the selective transporter for 5HT reuptake, could not convincingly be demonstrated in tuft cells. In villi and crypts, 3% and 10%, respectively, of all DCLK1-IR cells were in close proximity to EC cells. EC cells in close proximity to DCLK1-IR cells were, in villi and crypts, 3 and 8%, respectively. We conclude that DCLK1/5HT-IR cells constitute a novel subset of tuft cells that may have unique roles in the GI tract.


Asunto(s)
Células Enterocromafines , Mucosa Intestinal , Intestino Delgado/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Serotonina/metabolismo , Animales , Quinasas Similares a Doblecortina , Células Enterocromafines/citología , Células Enterocromafines/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Triptófano Hidroxilasa/metabolismo
3.
Exp Cell Res ; 369(1): 105-111, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29758188

RESUMEN

Tuft cells are gastrointestinal (GI) sensory cells recognized by their characteristic shape and their microvilli "tuft". Aims of the present study were to elucidate their regional distribution and spatial connections with satiety associated endocrine cells and nerve fibers throughout the intestinal tract. C57BL/6 J mice were used in the experiments. The small intestine was divided into five segments, and the large intestine was kept undivided. The segments were coiled into "Swiss rolls". Numbers and topographic distribution of tuft cells and possible contacts with endocrine cells and nerve fibers were estimated in the different segments, using immunocytochemistry. Tuft cells were found throughout the intestines; the highest number was in proximal small intestine. Five percent of tuft cells were found in close proximity to cholecystokinin-immunoreactive (IR) endocrine cells and up to 10% were in contact with peptide YY- and glucagon-like peptide-1-IR endocrine cells. Sixty percent of tuft cells in the small intestine and 40% in the large intestine were found in contact with nerve fibers. Calcitonin gene-related peptide-IR fibers constituted one-third of the fiber-contacts in the small intestine and two-thirds in the large intestine. These observations highlight the possibility of tuft cells as modulators of GI activities in response to luminal signaling.


Asunto(s)
Comunicación Celular/fisiología , Células Endocrinas/fisiología , Enterocitos/fisiología , Intestino Delgado/citología , Neuronas/fisiología , Animales , Recuento de Células , Colecistoquinina/metabolismo , Células Endocrinas/metabolismo , Enterocitos/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Intestino Delgado/inervación , Intestino Delgado/metabolismo , Intestinos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Péptido YY/metabolismo
4.
Int J Exp Pathol ; 99(1): 38-45, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29577471

RESUMEN

Reduced blood flow to the brain induces cerebral ischaemia, potentially causing central injury and peripheral complications including gastrointestinal (GI) dysfunction. The pathophysiology behind GI symptoms is suspected to be neuropathy in the enteric nervous system (ENS), which is essential in regulating GI function. This study investigates if enteric neuropathy occurs after cerebral ischaemia, by analysing neuronal survival and relative numbers of vasoactive intestinal peptide (VIP) and neuronal nitric oxide synthase (nNOS) expressing neurons in mouse ileum after three types of cerebral ischaemia. Focal cerebral ischaemia, modelled by permanent middle cerebral artery occlusion (pMCAO) and global cerebral ischaemia, modelled with either transient occlusion of both common carotid arteries followed by reperfusion (GCIR) or chronic cerebral hypoperfusion (CCH) was performed on C56BL/6 mice. Sham-operated mice for each ischaemia model served as control. Ileum was collected after 1-17 weeks, depending on model, and analysed using morphometry and immunocytochemistry. For each group, intestinal mucosa and muscle layer thicknesses, neuronal numbers and relative proportions of neurons immunoreactive (IR) for nNOS or VIP were estimated. No alterations in mucosa or muscle layer thicknesses were noted in any of the groups. Loss of myenteric neurons and an increased number of VIP-IR submucous neurons were found in mouse ileum 7 days after pMCAO. None of the global ischaemia models showed any alterations in neuronal survival or relative numbers of VIP- and nNOS-IR neurons. We conclude that focal cerebral ischaemia and global cerebral ischaemia influence enteric neuronal survival differently. This is suggested to reflect differences in peripheral neuro-immune responses.


Asunto(s)
Isquemia Encefálica/metabolismo , Íleon/inervación , Plexo Mientérico/metabolismo , Neuronas/patología , Péptido Intestinal Vasoactivo/metabolismo , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Estenosis Carotídea/fisiopatología , Muerte Celular , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Ratones Endogámicos C57BL , Plexo Mientérico/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Factores de Tiempo , Regulación hacia Arriba
5.
Exp Eye Res ; 169: 99-110, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29425879

RESUMEN

The purpose of this study was to explore retina-intrinsic neuroinflammatory reactions, effects on neuronal survival, relationship with classic gliosis, and possible role of the toll-like receptor 4 (TLR4). To isolate the adult retina from the systemic immune system, a previously described large animal explant culture model was used in which full-thickness porcine retinal sheets can be kept in vitro for extended time periods. Explants were kept for 5 days in vitro (DIV) and were treated with either; lipopolysaccharide (LPS), a Toll-like receptor-4 (TLR4) inhibitor (CLI-095), LPS + CLI-095, or solvent vehicle throughout the culture period after which retinal sections were examined with hematoxylin and eosin staining and extensive immunohistochemistry. In addition, culture medium from all explant groups was assayed for a panel of cytokines at 2 and 5DIV. Compared with in vivo controls, vehicle controls (CT) as well as CLI-095 explants displayed moderate reduction of total thickness and number of retinal neurons with upregulation of glial fibrillary acidic protein (GFAP) throughout the Müller cells. In contrast, LPS and LPS + CLI-095 treated counterparts showed extensive overall thinning with widespread neuronal degeneration but only minimal signs of classical Müller cell gliosis (limited upregulation of GFAP and no downregulation of glutamine synthetase (GS). These specimens also displayed a significantly increased expression of galectin-3 and TGF-beta activated kinase 1 (TAK1). Multiplex proteomic analysis of culture medium at 2DIV revealed elevated levels of IL-1ß, IL-6, IL-4 and IL-12 in LPS-treated explants compared to CLI-095 and CT counterparts. LPS stimulation of the isolated adult retina results in substantial neuronal cell death despite only minimal signs of gliosis indicating a retina-intrinsic neuroinflammatory response directly related to the degenerative process. This response is characterized by early upregulation of several inflammatory related cytokines with subsequent upregulation of Galectin-3, TLR4 and TAK1. Pharmacological block of TLR4 does not attenuate neuronal loss indicating that LPS induced retinal degeneration is mediated by TLR4 independent neuroinflammatory pathways.


Asunto(s)
Gliosis/fisiopatología , Inflamación/patología , Degeneración Nerviosa/patología , Degeneración Retiniana/patología , Neuronas Retinianas/patología , Receptor Toll-Like 4/fisiología , Animales , Supervivencia Celular , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Galectina 3/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Glutamato-Amoníaco Ligasa/metabolismo , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Degeneración Nerviosa/metabolismo , Proteómica , Degeneración Retiniana/metabolismo , Neuronas Retinianas/metabolismo , Porcinos , Factor de Crecimiento Transformador beta/metabolismo
6.
BMC Gastroenterol ; 18(1): 175, 2018 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-30463517

RESUMEN

BACKGROUND: The role of vitamin D in obesity and diabetes is debated. Obese and/or diabetic patients have elevated levels of free fatty acids, increased susceptibility to gastrointestinal symptoms and are suggested to have altered vitamin D balance. The enteric nervous system is pivotal in regulating gastrointestinal activity and high fat diet (HFD) has been shown to cause loss of enteric neurons in ileum and colon. This study investigates the effect of vitamin D on HFD- and palmitic acid-induced enteric neuronal loss in vivo and in vitro. METHODS: Mice were fed either a normal diet (ND) or HFD supplemented with varying levels of vitamin D (from 0x to 20x normal vitamin D level) for 19 weeks. Ileum and colon were analyzed for neuronal numbers and remodeling. Primary cultures of myenteric neurons from mouse small intestine were treated with palmitic acid (4x10-4M) and/or 1α,25-hydroxy-vitamin D3 (VD, 10-11- 10-7M) with or without modulators of lipid metabolism and VD pathways. Cultures were analyzed by immunocyto- and histochemical methods. RESULTS: Vitamin D supplementation had no effect on enteric neuronal survival in the ND group. HFD caused substantial loss of myenteric neurons in ileum and colon. Vitamin D supplementation between 0-2x normal had no effect on HFD-induced neuronal loss. Supplementation with 20x normal, prevented the HFD-induced neuronal loss. In vitro supplementation of VD prevented the palmitic acid-induced neuronal loss. The VD receptor (VDR) was not identified in enteric neurons. Enteric glia expressed the alternative VD receptor, protein disulphide isomerase family A member 3 (PDIA3), but PDIA3 was not found to mediate the VD response in vitro. Inhibition of peroxisome proliferator-activated receptor gamma (PPARγ) and immune neutralization of isocitrate lyase prevented the VD mediated neuroprotection to palmitic acid exposure. CONCLUSIONS: Results show that VD protect enteric neurons against HFD and palmitic acid induced neuronal loss. The mechanism behind is suggested to be through activation of PPARγ leading to improved neuronal peroxisome function and metabolism of neuronal lipid intermediates.


Asunto(s)
Calcifediol/farmacología , Colon/inervación , Dieta Alta en Grasa , Íleon/inervación , Plexo Mientérico/citología , Neuronas/efectos de los fármacos , Ácido Palmítico/farmacología , Animales , Calcifediol/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ratones Endogámicos C57BL , PPAR gamma/antagonistas & inhibidores , Proteína Disulfuro Isomerasas/análisis , Receptores de Calcitriol/análisis
7.
Eur J Oral Sci ; 126(2): 93-100, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29424090

RESUMEN

The human host defense peptide, LL-37, is an important player in the first line of defense against invading microorganisms. LL-37 and its precursor, hCAP18, have been detected in unstimulated whole saliva but no reports showing hCAP18/LL-37 in isolated, parotid, and/or submandibular/sublingual saliva have been presented. Here, we measured the levels of hCAP18/LL-37 in human parotid and submandibular/sublingual saliva and investigated the expression of hCAP18/LL-37 in parotid and submandibular gland tissue. Parotid and submandibular/sublingual saliva was collected from healthy volunteers, and the levels of hCAP18/LL-37 in saliva were analyzed by dot blot, ELISA, and western blotting. Cellular expression of hCAP18/LL-37 in human parotid and submandibular glands was investigated by immunohistochemistry. Immunoreactivity for hCAP18/LL-37 was detected in both parotid and submandibular/sublingual saliva of all individuals. The concentration of hCAP18/LL-37 was similar in parotid and submandibular/sublingual saliva, and was determined by densitometric scanning of each dot and normalization to the total protein concentration of each sample, and by ELISA. Double immunohistochemistry revealed that intravascular neutrophils of both parotid and submandibular glands express hCAP18/LL-37. For the first time, we demonstrate hCAP18/LL-37 in isolated human parotid and submandibular/sublingual saliva and expression of hCAP18/LL-37 in glandular intravascular neutrophils, indicating that neutrophils of the major salivary glands contribute to the LL-37 content of whole saliva.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/análisis , Neutrófilos/química , Saliva/química , Péptidos Catiónicos Antimicrobianos/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Neutrófilos/inmunología , Glándula Parótida , Glándula Submandibular , Catelicidinas
8.
Cell Tissue Res ; 366(2): 353-362, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27357804

RESUMEN

The biologically active form of vitamin D, 1α,25-dihydroxyvitamin D3 (1,25D3), has been reported to positively regulate the human cathelicidin anti-microbial peptide (CAMP) gene coding for LL-37, but the mechanisms are not completely understood. We have determined the expression of CAMP, vitamin D receptor (VDR), and the retinoid X receptor (RXR) isoforms in human skin and gingival tissue biopsies and investigated the signaling pathways involved in 1,25D3-induced upregulation of CAMP. Human skin and gingival biopsies exhibited few VDR-immunoreactive cells within the stratum basale, whereas rat colon enterocytes (positive control) possessed abundant VDR immunoreactivity. Nuclear VDR immunoreactivity was demonstrated in human skin keratinocytes (HaCaT cells). Gene analysis revealed that human skin biopsies expressed higher levels of both CAMP and RXRα mRNA than human gingival biopsies, whereas VDR and RXRß transcript levels were similar in skin and gingiva. In HaCaT cells, treatment with 1,25D3 (5 nM and 1 µM) for 4 and 24 h up-regulated CAMP mRNA several fold, and treatment with 1,25D3 for 24 h increased protein expression of the pro-form of LL-37 (hCAP-18) by about 13 times. The 1,25D3-evoked stimulation of HaCaT CAMP expression was associated with attenuated VDR mRNA and protein expression. Treatment with RXRα short interfering RNA reversed the 1,25D3-induced CAMP expression in HaCaT cells, showing that RXRα is involved in the up-regulation of CAMP by 1,25D3. We conclude that the 1,25D3-evoked stimulation of CAMP expression in human skin keratinocytes is dependent on RXRα but is not associated with the up-regulation of VDR expression.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Queratinocitos/metabolismo , Receptor alfa X Retinoide/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Vitamina D/farmacología , Péptidos Catiónicos Antimicrobianos/genética , Línea Celular , Femenino , Encía/metabolismo , Humanos , Queratinocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Receptor alfa X Retinoide/genética , Piel/metabolismo , Vitamina D/análogos & derivados , Catelicidinas
10.
Purinergic Signal ; 10(3): 455-64, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24510452

RESUMEN

Gastrointestinal symptoms have a major impact on the quality of life and are becoming more prevalent in the western population. The enteric nervous system (ENS) is pivotal in regulating gastrointestinal functions. Purinergic neurotransmission conveys a range of short and long-term cellular effects. This study investigated the role of the ADP-sensitive P2Y13 receptor in lipid-induced enteric neuropathy. Littermate P2Y13 (+/+) and P2Y13 (-/-) mice were fed with either a normal diet (ND) or high-fat diet (HFD) for 6 months. The intestines were analysed for morphological changes as well as neuronal numbers and relative numbers of vasoactive intestinal peptide (VIP)- and neuronal nitric oxide synthase (nNOS)-containing neurons. Primary cultures of myenteric neurons from the small intestine of P2Y13 (+/+) or P2Y13 (-/-) mice were exposed to palmitic acid (PA), the P2Y13 receptor agonist 2meSADP and the antagonist MRS2211. Neuronal survival and relative number of VIP-containing neurons were analysed. In P2Y13 (+/+), but not in P2Y13 (-/-) mice, HFD caused a significant loss of myenteric neurons in both ileum and colon. In colon, the relative numbers of VIP-containing submucous neurons were significantly lower in the P2Y13 (-/-) mice compared with P2Y13 (+/+) mice. The relative numbers of nNOS-containing submucous colonic neurons increased in P2Y13 (+/+) HFD mice. HFD also caused ileal mucosal thinning in P2Y13 (+/+) and P2Y13 (-/-) mice, compared to ND fed mice. In vitro PA exposure caused loss of myenteric neurons from P2Y13 (+/+) mice while neurons from P2Y13 (-/-) mice were unaffected. Presence of MRS2211 prevented PA-induced neuronal loss in cultures from P2Y13 (+/+) mice. 2meSADP caused no change in survival of cultured neurons. P2Y13 receptor activation is of crucial importance in mediating the HFD- and PA-induced myenteric neuronal loss in mice. In addition, the results indicate a constitutive activation of enteric neuronal apoptosis by way of P2Y13 receptor stimulation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Sistema Nervioso Entérico/metabolismo , Neuronas/metabolismo , Ácido Palmítico/toxicidad , Receptores Purinérgicos P2/deficiencia , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plexo Mientérico/efectos de los fármacos , Plexo Mientérico/metabolismo , Plexo Mientérico/patología , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos
11.
Cell Tissue Res ; 351(3): 521-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23254679

RESUMEN

Gonadotropin-releasing hormone (GnRH) analogs are given to women undergoing in vitro fertilization. Case reports describing the development of chronic intestinal pseudo-obstruction and auto-antibodies against GnRH after such treatment suggest a strong association between intestinal dysfunction and GnRH analogs. No experimental model for studying such a relationship is currently at hand. Our main goal was to investigate possible enteric neurodegeneration and titers of GnRH antibodies in response to repeated administration of the GnRH analog buserelin in rat. Rats were treated for 1-4 sessions with daily subcutaneous injections of buserelin or saline for 5 days, followed by 3 weeks of recovery. Buserelin treatment caused significant loss of submucous and myenteric neurons in the fundus, ileum, and colon. The loss of enteric neurons can, at least partly, be explained by increased apoptosis. No GnRH- or GnRH-receptor-immunoreactive (IR) enteric neurons but numerous luteinizing hormone (LH)-receptor-IR neurons were detected. After buserelin treatment, the relative number of enteric LH-receptor-IR neurons decreased, whereas that of nitric-oxide-synthase-IR neurons increased. No intestinal inflammation or increased levels of circulating interleukins/cytokines were noted in response to buserelin treatment. Serum GnRH antibody titers were undetectable or extremely low in all rats. Thus, repeated administrations of buserelin induce neurodegeneration in rat gastrointestinal tract, possibly by way of LH-receptor hyperactivation. The present findings suggest that enteric neurodegenerative effects of GnRH analog treatment in man can be mimicked in rat. However, in contrast to man, no production of GnRH auto-antibodies has been noted in rat.


Asunto(s)
Buserelina/farmacología , Tracto Gastrointestinal/citología , Hormona Liberadora de Gonadotropina/análogos & derivados , Neuronas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Colon/citología , Femenino , Inmunohistoquímica , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de HL/metabolismo , Receptores LHRH/metabolismo , Péptido Intestinal Vasoactivo/metabolismo
12.
Purinergic Signal ; 9(1): 67-79, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22941026

RESUMEN

While high levels of glucose and saturated fatty acids are known to have detrimental effects on beta cell function and survival, the signalling pathways mediating these effects are not entirely known. In a previous study, we found that ADP regulates beta cell insulin secretion and beta cell apoptosis. Using MIN6c4 cells as a model system, we investigated if autocrine/paracrine mechanisms of ADP and purinergic receptors are involved in this process. High glucose (16.7 mmol/l) and palmitate (100 µmol/l) rapidly and potently elevated the extracellular ATP levels, while mannitol was without effect. Both tolbutamide and diazoxide were without effect, while the calcium channel blocker nifedipine, the volume-regulated anion channels (VRAC) inhibitor NPPB, and the pannexin inhibitor carbenoxolone could inhibit both effects. Similarly, silencing the MDR1 gene also blocked nutrient-generated ATP release. These results indicate that calcium channels and VRAC might be involved in the ATP release mechanism. Furthermore, high glucose and palmitate inhibited cAMP production, reduced cell proliferation in MIN6c4 and increased activated Caspase-3 cells in mouse islets and in MIN6c4 cells. The P2Y(13)-specific antagonist MRS2211 antagonized all these effects. Further studies showed that blocking the P2Y(13) receptor resulted in enhanced CREB, Bad and IRS-1 phosphorylation, which are known to be involved in beta cell survival and insulin secretion. These findings provide further support for the concept that P2Y(13) plays an important role in beta cell apoptosis and suggest that autocrine/paracrine mechanisms, related to ADP and P2Y(13) receptors, contribute to glucolipotoxicity.


Asunto(s)
Adenosina Difosfato/farmacología , Apoptosis/efectos de los fármacos , Comunicación Autocrina/efectos de los fármacos , Ácidos Grasos no Esterificados/farmacología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Receptores Purinérgicos P2/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adenosina Trifosfato/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular Tumoral , Separación Celular , Femenino , Inmunohistoquímica , Insulina/metabolismo , Secreción de Insulina , Lentivirus/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción Genética
13.
BMC Gastroenterol ; 12: 30, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22463807

RESUMEN

BACKGROUND: Neuropathy is believed to be a common feature of functional and inflammatory intestinal diseases. Vasoactive intestinal peptide (VIP) is an acknowledged neuroprotective agent in peripheral, including enteric, and central neurons. The proglucagon-like hormones glucagon-like peptide 1 and 2 (GLP1 and GLP2) belong to the secretin/glucagon/VIP superfamily of peptides and GLP1 and GLP2 receptors are expressed in enteric neurons. Possible neuroprotective effects of these peptides were investigated in the present study. METHODS: GLP1, GLP2 and VIP were added to cultured myenteric neurons from rat small intestine or to co-cultures of myenteric neurons and rat peritoneal mast cells. Receptor selectivity was tested by the simultaneous presence of a GLP1 receptor antagonist (exendin (9-39) amide) or a VIP receptor antagonist (hybrid of neurotensin 6-11 and VIP 7-28). Neuronal survival was examined using immunocytochemistry and cell counting. RESULTS: GLP1, GLP2 and VIP significantly and concentration-dependently enhanced neuronal survival. In addition the peptides efficiently counteracted mast cell-induced neuronal cell death in a concentration-dependent manner. Exendin(9-39)amide reversed GLP1-induced neuroprotection while GLP2- and VIP-induced enhanced neuronal survival were unaffected. The VIP receptor antagonist reversed GLP1- and VIP-induced neuroprotection while the GLP2-induced effect on neuronal survival was unaffected. CONCLUSIONS: By activating separate receptors VIP, GLP1 and GLP2 elicit neuroprotective effects on rat myenteric neurons cultured with or without mast cells. This implies a powerful therapeutic potential of these peptides in enteric neuropathies with a broad spectrum of applications from autoimmunity to functional disorders.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Péptido 1 Similar al Glucagón/farmacología , Péptido 2 Similar al Glucagón/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptido Intestinal Vasoactivo/farmacología , Análisis de Varianza , Animales , Células Cultivadas , Femenino , Intestino Delgado/inervación , Mastocitos , Plexo Mientérico , Neurotensina/farmacología , Fragmentos de Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/farmacología
14.
Physiol Rep ; 10(7): e15249, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35416410

RESUMEN

Air pollution and exposure to fine airborne particles with aerodynamic diameter <2.5 µm (PM2.5 ) negatively impacts human health. Airways constitute a primary route of exposure but PM2.5 -contaminated food, drinks as well as mucociliary and hepatobiliary clearance all constitute potential entry points into the intestine. This study evaluated intestinal histopathological and inflammatory changes as well as enteric neuronal numbers after short- or long-term exposure to urban PM2.5 . Using a nebulizer, male rats were exposed to a mist with a concentration of 5.3mg PM2.5 /m3 for 8 h (short term) or 1.8 mg PM2.5 /m3 for 3 h/day, 5 days/week for 8 weeks (long-term) with controls run in parallel. Samples were taken from three regions of the small intestine as well as the colon. Results showed that short-term exposure to PM2.5 induces mucosal lesions and reduces IL1ß levels in the small intestine but not colon. No significant changes were observed after long-term exposure, suggesting the presence of intestinal adaptation to environmental stressors in the PM2.5 . To our knowledge, this is the first study to systematically characterize regional effects along the intestine.


Asunto(s)
Contaminantes Atmosféricos , Contaminación del Aire , Contaminantes Atmosféricos/análisis , Contaminantes Atmosféricos/toxicidad , Contaminación del Aire/efectos adversos , Contaminación del Aire/análisis , Animales , Masculino , Material Particulado/análisis , Material Particulado/toxicidad , Ratas
15.
Cell Tissue Res ; 346(3): 393-405, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22108805

RESUMEN

Nesfatin-1 is a novel anorexigenic regulatory peptide. The peptide is the N-terminal part of nucleobindin 2 (NUCB2) and is expressed in brain areas regulating feeding. Outside the brain, nesfatin-1 expression has been reported in adipocytes, gastric endocrine cells and islet cells. We studied NUCB2 expression in human and rodent islets using immunocytochemistry, in situ hybridization and western blot. Furthermore, we investigated the potential influence of nesfatin-1 on secretion of insulin and glucagon in vitro and in vivo in mice and in INS-1 (832/13) cells. The impact of type 2 diabetes (T2D) and glucolipotoxicity on NUCB2 gene expression in human islets and its relationship to insulin secretory capacity and islet gene expression was studied using microarray. Nesfatin-1 immunoreactivity (IR) was abundant in human and rodent beta cells but absent in alpha, delta, PP and ghrelin cells. Importantly, in situ hybridization showed that NUCB2 mRNA is expressed in human and rat islets. Western blot analysis showed that nesfatin-1 IR represented full length NUCB2 in rodent islets. Human islet NUCB2 mRNA was reduced in T2D subjects but upregulated after culture in glucolipotoxic conditions. Furthermore, a positive correlation between NUCB2 and glucagon and insulin gene expression, as well as insulin secretory capacity, was evident. Nesfatin-1 enhanced glucagon secretion but had no effect on insulin secretion from mouse islets or INS-1 (832/13) cells. On the other hand, nesfatin-1 caused a small increase in insulin secretion and reduced glucose during IVGTT in mice. We conclude that nesfatin-1 is a novel glucagon-stimulatory peptide expressed in the beta cell and that its expression is decreased in T2D islets.


Asunto(s)
Proteínas de Unión al Calcio/biosíntesis , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Glucagón/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Unión al ADN/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Glucagón/genética , Humanos , Inmunohistoquímica , Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Nucleobindinas , Ratas , Ratas Sprague-Dawley
16.
J Pineal Res ; 50(4): 412-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21355877

RESUMEN

Melatonin has multiple receptor-dependent and receptor-independent functions. At the cell membrane, melatonin interacts with its receptors MT1 and MT2, which are expressed in numerous tissues. Genome-wide association studies have recently shown that the MTNR1B/MT2 receptor may be involved in the pathogenesis of type 2 diabetes mellitus. In line with these findings, expression of melatonin receptors has been shown in mouse, rat, and human pancreatic islets. MT1 and MT2 are G-protein-coupled receptors and are proposed to exert inhibitory effects on insulin secretion. Here, we show by immunocytochemistry that these membrane melatonin receptors have distinct locations in the mouse islet. MT1 is expressed in α-cells while MT2 is located to the ß-cells. These findings help to unravel the complex machinery underlying melatonin's role in the regulation of islet function.


Asunto(s)
Receptores de Melatonina/metabolismo , Animales , Femenino , Inmunohistoquímica , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Receptor de Melatonina MT1/genética , Receptor de Melatonina MT1/metabolismo , Receptor de Melatonina MT2/genética , Receptor de Melatonina MT2/metabolismo , Receptores de Melatonina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Genesis ; 48(6): 374-81, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20533404

RESUMEN

The cadherin family of cell adhesion molecules mediates adhesive interactions that are required for the formation and maintenance of tissues. Previously, we demonstrated that N-cadherin, which is required for numerous morphogenetic processes, is expressed in the pancreatic epithelium at E9.5, but later becomes restricted to endocrine aggregates in mice. To study the role of N-cadherin during pancreas formation and function we generated a tissue-specific knockout of N-cadherin in the early pancreatic epithelium by inter-crossing N-cadherin-floxed mice with Pdx1Cre mice. Analysis of pancreas-specific ablation of N-cadherin demonstrates that N-cadherin is dispensable for pancreatic development, but required for beta-cell granule turnover. The number of insulin secretory granules is significantly reduced in N-cadherin-deficient beta-cells, and as a consequence insulin secretion is decreased.


Asunto(s)
Cadherinas/fisiología , Gránulos Citoplasmáticos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/fisiología , Células Secretoras de Insulina/metabolismo , Páncreas/crecimiento & desarrollo , Transactivadores/fisiología , Animales , Femenino , Técnica del Anticuerpo Fluorescente , Immunoblotting , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Páncreas/metabolismo
18.
Sci Rep ; 6: 32893, 2016 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-27612206

RESUMEN

In addition to brain injury stroke patients often suffer gastrointestinal complications. Neuroimmune interactions involving galectin-3, released from microglia in the brain, mediates the post-stroke pro-inflammatory response. We investigated possible consequences of stroke on the enteric nervous system and the involvement of galectin-3. We show that permanent middle cerebral artery occlusion (pMCAO) induces loss of enteric neurons in ileum and colon in galectin-3(+/+), but not in galectin-3(-/-), mice. In vitro we show that serum from galectin-3(+/+), but not from galectin-3(-/-), mice subjected to pMCAO, caused loss of C57BL/6J myenteric neurons, while myenteric neurons derived from TLR4(-/-) mice were unaffected. Further purified galectin-3 (10(-6) M) caused loss of cultured C57BL/6J myenteric neurons. Inhibitors of transforming growth factor ß-activated kinase 1 (TAK1) or AMP activated kinase (AMPK) counteracted both the purified galectin-3 and the galectin-3(+/+) pMCAO serum-induced loss in vitro. Combined we show that stroke (pMCAO) triggers central and peripheral galectin-3 release causing enteric neuronal loss through a TLR4 mediated mechanism involving TAK1 and AMPK. Galectin-3 is suggested a target for treatment of post-stroke complications.


Asunto(s)
Galectina 3/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Microglía/metabolismo , Neuronas/fisiología , Transducción de Señal , Accidente Cerebrovascular/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Colon/inervación , Colon/fisiopatología , Modelos Animales de Enfermedad , Íleon/inervación , Íleon/fisiopatología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/fisiopatología , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/fisiopatología , Receptor Toll-Like 4/metabolismo
19.
Auton Neurosci ; 193: 104-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26480825

RESUMEN

BACKGROUND: Clinical observations have suggested repeated gonadotropin-releasing hormone (GnRH) exposure to cause intestinal dysfunction and loss of enteric neurons. This has been further studied and confirmed in a rat in vivo model involving iterated GnRH treatments. Mechanisms behind are enigmatic since no GnRH receptors are found to be expressed in enteric neurons neither in man nor rat. Both species, however, harbor substantial subpopulations of luteinizing hormone (LH) receptor-immunoreactive myenteric neurons which suggests that intestinal GnRH-induced neuropathy may be mediated by LH release. AIMS: To reveal if exposures of GnRH or LH to rat myenteric neurons in vitro cause neuronal loss. METHODS: Primary cultured adult rat myenteric neurons were exposed to single or repeated treatments of the GnRH analog buserelin or the LH analog lutrotropin alpha, and neuronal survival was determined by cell counting. Possible presence of GnRH- or LH receptor -immunoreactive neurons was determined by immunocytochemistry. RESULTS: Exposure to the LH, but not the GnRH, analog caused significantly reduced neuronal survival. LH, but not GnRH, receptors were found to be expressed on cultured myenteric neurons. CONCLUSION: Myenteric neurons express LH receptors in vitro and LH exposure causes reduced neuronal survival. This suggests that GnRH-induced enteric neuropathy in vivo is mediated by way of LH release and activation of enteric neuronal LH receptors.


Asunto(s)
Plexo Mientérico/fisiología , Neuronas/fisiología , Receptores de HL/metabolismo , Animales , Buserelina/farmacología , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Hormona Liberadora de Gonadotropina/análogos & derivados , Hormonas/farmacología , Íleon/inervación , Inmunohistoquímica , Hormona Luteinizante/análogos & derivados , Masculino , Plexo Mientérico/efectos de los fármacos , Neuronas/efectos de los fármacos , Ratas Sprague-Dawley , Receptores LHRH/metabolismo
20.
Mol Cancer Ther ; 14(1): 259-67, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25381265

RESUMEN

Intestinal alkaline sphingomyelinase (alk-SMase) generates ceramide and inactivates platelet-activating factor (PAF) and was previously suggested to have anticancer properties. The direct evidence is still lacking. We studied colonic tumorigenesis in alk-SMase knockout (KO) mice. Formation of aberrant crypt foci (ACF) was examined after azoxymethane (AOM) injection. Tumor was induced by AOM alone, a conventional AOM/dextran sulfate sodium (DSS) treatment, and an enhanced AOM/DSS method. ß-Catenin was determined by immunohistochemistry, PAF levels by ELISA, and sphingomyelin metabolites by mass spectrometry. Without treatment, spontaneous tumorigenesis was not identified but the intestinal mucosa appeared thicker in KO than in wild-type (WT) littermates. AOM alone induced more ACF in KO mice but no tumors 28 weeks after injection. However, combination of AOM/DSS treatments induced colonic tumors and the incidence was significantly higher in KO than in WT mice. By the enhanced AOM/DSS method, tumor number per mouse increased 4.5 times and tumor size 1.8 times in KO compared with WT mice. Although all tumors were adenomas in WT mice, 32% were adenocarcinomas in KO mice. Compared with WT mice, cytosol expression of ß-catenin was significantly increased and nuclear translocation in tumors was more pronounced in KO mice. Lipid analysis showed decreased ceramide in small intestine and increased sphingosine-1-phosphate (S1P) in both small intestine and colon in nontreated KO mice. PAF levels in feces were significantly higher in the KO mice after AOM/DSS treatment. In conclusion, lack of alk-SMase markedly increases AOM/DSS-induced colonic tumorigenesis associated with decreased ceramide and increased S1P and PAF levels.


Asunto(s)
Azoximetano/toxicidad , Proteínas Portadoras/metabolismo , Neoplasias del Colon/genética , Sulfato de Dextran/toxicidad , Esfingomielina Fosfodiesterasa/genética , Adenocarcinoma , Animales , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/patología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Esfingolípidos/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA