Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Cell Physiol ; 238(10): 2304-2315, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37555566

RESUMEN

Gastrointestinal epithelial cells respond to milk-born molecules throughout breastfeeding, influencing growth, and development. The rapid renewal of the small intestine depends on the proliferation in the crypt that drives cell fates. We used early weaning model to investigate immediate and late effects of breastfeeding on proliferation, differentiation of jejunal epithelial cells. Wistar rats were either allowed to suckle (S) until 21 postnatal days or submitted to early weaning (EW) at 15 days. By comparing ages (18, 60, and 120 days), we found that EW decreased Ki67 indices and villi height at 18 and 60 days (p < 0.05), and at 120 days they were similar between diets. Proliferative reduction and augmented expression of Cdkn1b (p27 gene) were parallel. In the stem cell niche, EW increased the number and activity (Defa24) of Paneth cells at 18 and 60 days (p < 0.05), and Lgr5 and Ascl2 genes showed inverted responses between ages. Among target cells, EW decreased goblet cell number at 18 and 60 days (p < 0.05) and increased it at 120 days (p < 0.05), whereas enteroendocrine marker genes were differentially altered. EW reduced enterocytes density at 18 days (p < 0.05), and at 120 days this population was decreased (vs. 60 days). Among cell fate crypt-controlling genes, Notch and Atoh1 were the main targets of EW. Metabolically, intraperitoneal glucose tolerance was immediately reduced (18 days), being reverted until 120 days (p < 0.05). Currently, we showed that breastfeeding has a lifespan influence on intestinal mucosa and on its stem cell compartment. We suggest that, although jejunum absorptive function is granted after early weaning, the long lasting changes in gene expression might prime the mucosa with a different sensitivity to gut disorders that still have to be further explored.

2.
Cell Biol Int ; 46(5): 701-710, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35032139

RESUMEN

The small intestine mucosa is lined by specialized cells that form the crypt-villus axis, which expands its surface. Among the six intestinal epithelial cell types, the Paneth cell is located at the base of the crypt, and it contains numerous granules in its cytoplasm, composed of antimicrobial peptides, such as defensins and lysozyme, and growth factors, such as epidermal growth factor, transforming growth factor-α, and Wnt ligands. Together, these elements act in the defense against microorganisms, regulation of intestinal microbiota, maintenance, and regulation of stem cell identity. Pathologies that target Paneth cells can disturb such defense activity, but they also affect the maintenance of the stem cell niche. In that way, Crohn's disease, necrotizing enterocolitis, and graft-versus-host disease promote a reduction of Paneth cell population, and, consequently, secretion of their products into the lumen of the crypts, making the affected organism predisposed to infections and dysbiosis. Additionally, the emergence of new intestinal cells is also decreased. This review aims to address the main characteristics of Paneth cells, highlighting their multiple functions and the importance of their preservation to ensure bowel homeostasis.


Asunto(s)
Mucosa Intestinal , Células de Paneth , Recuento de Células , Intestinos , Células de Paneth/metabolismo , Nicho de Células Madre
3.
Int J Mol Sci ; 21(1)2019 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-31892140

RESUMEN

Background: Gastric glands grow and cells reach differentiation at weaning in rats. By considering that early weaning (EW) can affect the timing of development, we aimed to compare molecular and cellular markers of differentiation in pups and adults. Methods: Wistar rats were separated into suckling-control (S) and EW groups at 15 days. Stomachs were collected at 15, 18, and 60 days for RNA and protein extraction, and morphology. Results: After EW, the expression of genes involved in differentiation (Atp4b, Bhlha15 and Pgc) augmented (18 days), and Atp4b and Gif were high at 60 days. EW increased the number of zymogenic cells (ZC) in pups and adults and augmented mucous neck cells only at 18 days, whereas parietal and transition cells (TC) were unchanged. Conclusions: EW affected the gastric mucosa mostly in a transient manner as the changes in gene expression and distribution of differentiated cells that were detected in pups were not fully maintained in adults, except for the size of ZC population. We concluded that though most of EW effects were immediate, such nutritional change in the infancy might affect part of gastric digestive functions in a permanent manner, as some markers were kept unbalanced in the adulthood.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/fisiología , Mucosa Gástrica/fisiología , Estómago/fisiología , Animales , Femenino , Expresión Génica/fisiología , Masculino , Ratas , Ratas Wistar , Destete
4.
Int J Mol Sci ; 14(5): 10143-61, 2013 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-23670595

RESUMEN

Mitogen-activated protein kinase (MAPK) pathways are activated by several stimuli and transduce the signal inside cells, generating diverse responses including cell proliferation, differentiation, migration and apoptosis. Each MAPK cascade comprises a series of molecules, and regulation takes place at different levels. They communicate with each other and with additional pathways, creating a signaling network that is important for cell fate determination. In this review, we focus on ERK, JNK, p38 and ERK5, the major MAPKs, and their interactions with PI3K-Akt, TGFß/Smad and Wnt/ß-catenin pathways. More importantly, we describe how MAPKs regulate cell proliferation and differentiation in the rapidly renewing epithelia that lines the gastrointestinal tract and, finally, we highlight the recent findings on nutritional aspects that affect MAPK transduction cascades.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Epiteliales/citología , Tracto Gastrointestinal/citología , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Transducción de Señal , Animales , Células Epiteliales/metabolismo , Tracto Gastrointestinal/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo
5.
Differentiation ; 79(1): 48-56, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19783352

RESUMEN

The development of the gastric mucosa is controlled by hormones, growth factors and feeding behavior. Early weaning (EW), which means the abrupt interruption of suckling, increases proliferation and differentiation in the rat gastric epithelium. Transforming growth factor alpha (TGFalpha) is secreted in the stomach, binds to the epidermal growth factor receptor (EGFR) and may control cell proliferation, differentiation and migration. Here, we investigated the influence of suckling-weaning transition on the differentiation of mucous neck cells in the stomach and its association to the expression of TGFalpha and EGFR. Fifteen-day-old Wistar rats were divided into two groups: suckling (control), in which pups were kept with the dam, and early weaning (EW), in which rats were separated from their mother and fed with hydrated powdered chow. TGFalpha and EGFR levels were increased at 18 days in EW animals compared to control ones (p<0.05). Histochemical reactions with Periodic Acid-Schiff reagent+Alcian Blue or Bandeiraea simplicifolia II lectin were used to stain the mucous neck cells and showed an increase in this cell population throughout EW, which was more pronounced at 17 days when compared to suckling pups (p<0.05). These morphological results were confirmed by RT-PCR for mucin 6. The levels of mucin 6 mRNA were higher in EW animals from the 16th to the 18th day (1-3 days post-weaning) when compared to the respective control group. Inhibition of EGFR through AG1478 administration to EW animals prevented the expansion of mucous neck cell population induced by EW (p<0.05). Therefore, early weaning up regulated TGFalpha/EGFR expression and induced differentiation of mucous neck cells. Moreover, we showed that EGFR takes part in the maturation of this cell population. We conclude that regular suckling-weaning transition is crucial to guarantee the development of the gastric mucosa.


Asunto(s)
Diferenciación Celular , Receptores ErbB/fisiología , Mucosa Gástrica/citología , Regulación del Desarrollo de la Expresión Génica , Factor de Crecimiento Transformador alfa/fisiología , Animales , Western Blotting , Células Cultivadas , Receptores ErbB/antagonistas & inhibidores , Femenino , Mucosa Gástrica/metabolismo , Técnicas para Inmunoenzimas , Mucina 6/genética , Quinazolinas , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirfostinos/farmacología , Destete
6.
Physiol Rep ; 9(3): e14744, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33580917

RESUMEN

During postnatal development, colostrum and breastmilk are sequentially the first sources of nutrition with protein components and bioactive molecules that confer protection and immunostimulatory function to the gut. Caseins, whey proteins, secretory immunoglobulin A (sIgA), mucins, tryptophan, and growth factors are among milk-borne elements that are directly important in the control of mucosa development and protection. Consequently, breastfeeding is associated with the low incidence of gastrointestinal inflammation and with the decrease in respiratory diseases during postnatal period. The novel coronavirus (SARS-CoV-2) binds to angiotensin II-converting enzyme (ACE2) on the cell membrane, allowing virus entrance, replication, and host commitment. ACE2 is expressed by different cell types, which include ciliated cells in the lungs and enterocytes in the intestine. Such cells are highly active in metabolism, as they internalize molecules to be processed and used by the organism. The disruption of ACE2 impairs leads to intestinal inflammation and decreased synthesis of serotonin, affecting motility. By reviewing the effects of SARS-CoV-2 in the gastrointestinal and respiratory tracts in infants, and gut responses to breastfeeding interruption, we suggest that it is important to maintain breastfeeding during SARS-CoV-2 infection, as it might be essential to protect newborns from gastrointestinal-associated disorders and relieve disease symptoms.


Asunto(s)
Lactancia Materna/tendencias , COVID-19/metabolismo , COVID-19/prevención & control , COVID-19/transmisión , Femenino , Humanos
7.
Front Physiol ; 12: 721242, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34588994

RESUMEN

The gastric mucosa is disturbed when breastfeeding is interrupted, and such early weaning (EW) condition permanently affects the differentiation of zymogenic cells. The aim of the study was to evaluate the immediate and long-term effects of EW on gastric cell proliferation, considering the molecular markers for cell cycle, inflammation, and metaplasia. Overall, we investigated the lifelong adaptation of gastric growth. Wistar rats were divided into suckling-control (S) and EW groups, and gastric samples were collected at 18, 30, and 60 days for morphology, RNA, and protein isolation. Inflammation and metaplasia were not identified, but we observed that EW promptly increased Ki-67-proliferative index (PI) and mucosa thickness (18 days). From 18 to 30 days, PI increased in S rats, whereas it was stable in EW animals, and such developmental change in S made its PI higher than in EW. At 60 days, the PI decreased in S, making the indices similar between groups. Spatially, during development, proliferative cells spread along the gland, whereas, in adults, they concentrate at the isthmus-neck area. EW pushed dividing cells to this compartment (18 days), increased PI at the gland base (60 days), but it did not interfere in expression of cell cycle molecules. At 18 days, EW reduced Tgfß2, Tgfß3, and Tgfbr2 and TßRII and p27 levels, which might regulate the proliferative increase at this age. We demonstrated that gastric cell proliferation is immediately upregulated by EW, corroborating previous results, but for the first time, we showed that such increased PI is stable during growth and aging. We suggest that suckling and early weaning might use TGFßs and p27 to trigger different proliferative profiles during life course.

8.
Am J Physiol Gastrointest Liver Physiol ; 298(1): G117-25, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19833863

RESUMEN

The nutritional status influences gastric growth, and interestingly, whereas cell proliferation is stimulated by fasting in suckling rats, it is inhibited in adult animals. Corticosterone takes part in the mechanisms that govern development, and its effects are regulated in particular by corticosterone-binding globulin (CBG) and glucocorticoid receptor (GR). To investigate whether corticosterone activity responds to fasting and how possible changes might control gastric epithelial cell cycle, we evaluated different parameters during the progression of fasting in 18- and 40-day-old rats. Food restriction induced higher corticosterone plasma concentration at both ages, but only in pups did CBG binding increase after short- and long-term treatments. Fasting also increased gastric GR at transcriptional and protein levels, but the effect was more pronounced in 40-day-old animals. Moreover, in pups, GR was observed in the cytoplasm, whereas, in adults, it accumulated in the nucleus after the onset of fasting. Heat shock protein (HSP) 70 and HSP 90 were differentially regulated and might contribute to the stability of GR and to the high cytoplasmic levels in pups and elevated shuttling in adult rats. As for gastric epithelial cell cycle, whereas cyclin D1 and p21 increased during fasting in pups, in adults, cyclin E slowly decreased, concomitant with higher p27. In summary, we demonstrated that corticosterone function is differentially regulated by fasting in 18- and 40-day-old rats, and such variation might attenuate any possible suppressive effects during postnatal development. We suggest that this mechanism could ultimately increase cell proliferation and allow regular gastric growth during adverse nutritional conditions.


Asunto(s)
Ayuno/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/metabolismo , Receptores de Glucocorticoides/metabolismo , Transcortina/metabolismo , Factores de Edad , Animales , Animales Lactantes , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Corticosterona/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/genética
9.
Cancer Res ; 67(3): 1007-18, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17283133

RESUMEN

Hormones and growth factors regulate endometrial cell growth. Disrupted transforming growth factor-beta (TGF-beta) signaling in primary endometrial carcinoma (ECA) cells leads to loss of TGF-beta-mediated growth inhibition, which we show herein results in lack of up-regulation of the cyclin-dependent kinase inhibitor p27(Kip1) (p27) to arrest cells in G(1) phase of the cell cycle. Conversely, in normal primary endometrial epithelial cells (EECs), TGF-beta induces a dose-dependent increase in p27 protein, with a total 3.6-fold maximal increase at 100 pmol/L TGF-beta, which was 2-fold higher in the nuclear fraction; mRNA levels were unaffected. In addition, ECA tissue lysates show a high rate of ubiquitin-mediated degradation of p27 compared with normal secretory-phase endometrial tissue (SE) such that 4% and 89% of recombinant p27 added to the lysates remains after 3 and 20 h, respectively. These results are reflected in vivo as ECA tissue lacks p27 compared with high expression of p27 in SE (P < or = 0.001). Furthermore, we show that estrogen treatment of EECs causes mitogen-activated protein kinase-driven proteasomal degradation of p27 whereas progesterone induces a marked increase in p27 in both normal EECs and ECA cells. Therefore, these data suggest that TGF-beta induces accumulation of p27 for normal growth regulation of EECs. However, in ECA, in addition to enhanced proteasomal degradation of p27, TGF-beta cannot induce p27 levels due to dysregulated TGF-beta signaling, thereby causing 17beta-estradiol-driven p27 degradation to proceed unchecked for cell cycle progression. Thus, p27 may be a central target for growth regulation of normal endometrium and in the pathogenesis of ECA.


Asunto(s)
Neoplasias Endometriales/metabolismo , Estradiol/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Progesterona/farmacología , Factor de Crecimiento Transformador beta/farmacología , Adulto , Procesos de Crecimiento Celular/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Citoplasma/metabolismo , Neoplasias Endometriales/enzimología , Neoplasias Endometriales/patología , Endotelio/citología , Endotelio/efectos de los fármacos , Endotelio/enzimología , Endotelio/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Células Tumorales Cultivadas , Ubiquitinas/biosíntesis
10.
J Cachexia Sarcopenia Muscle ; 10(5): 1116-1127, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31307125

RESUMEN

BACKGROUND: Cachexia is a multifactorial and multiorgan syndrome associated with cancer and other chronic diseases and characterized by severe involuntary body weight loss, disrupted metabolism, inflammation, anorexia, fatigue, and diminished quality of life. This syndrome affects around 50% of patients with colon cancer and is directly responsible for the death of at least 20% of all cancer patients. Systemic inflammation has been recently proposed to underline most of cachexia-related symptoms. Nevertheless, the exact mechanisms leading to the initiation of systemic inflammation have not yet been unveiled, as patients bearing the same tumour and disease stage may or may not present cachexia. We hypothesize a role for gut barrier disruption, which may elicit persistent immune activation in the host. To address this hypothesis, we analysed the healthy colon tissue, adjacent to the tumour. METHODS: Blood and rectosigmoid colon samples (20 cm distal to tumour margin) obtained during surgery, from cachectic (CC = 25) or weight stable (WSC = 20) colon cancer patients, who signed the informed consent form, were submitted to morphological (light microscopy), immunological (immunohistochemistry and flow cytometry), and molecular (quantification of inflammatory factors by Luminex® xMAP) analyses. RESULTS: There was no statistical difference in gender and age between groups. The content of plasma interleukin 6 (IL-6) and IL-8 was augmented in cachectic patients relative to those with stable weight (P = 0.047 and P = 0.009, respectively). The number of lymphocytic aggregates/field in the gut mucosa was higher in CC than in WSC (P = 0.019), in addition to those of the lamina propria (LP) eosinophils (P < 0.001) and fibroblasts (P < 0.001). The area occupied by goblet cells in the colon mucosa was decreased in CC (P = 0.016). The M1M2 macrophages percentage was increased in the colon of CC, in relation to WSC (P = 0.042). Protein expression of IL-7, IL-13, and transforming growth factor beta 3 in the colon was significantly increased in CC, compared with WSC (P = 0.02, P = 0.048, and P = 0.048, respectively), and a trend towards a higher content of granulocyte-colony stimulating factor in CC was also observed (P = 0.061). The results suggest an increased recruitment of immune cells to the colonic mucosa in CC, as compared with WSC, in a fashion that resembles repair response following injury, with higher tissue content of IL-13 and transforming growth factor beta 3. CONCLUSIONS: The changes in the intestinal mucosa cellularity, along with modified cytokine expression in cachexia, indicate that gut barrier alterations are associated with the syndrome.


Asunto(s)
Caquexia/etiología , Caquexia/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Neoplasias/complicaciones , Anciano , Biomarcadores , Caquexia/metabolismo , Citocinas/metabolismo , Femenino , Humanos , Inmunohistoquímica , Inflamación , Mediadores de Inflamación , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Proteoma , Proteómica
11.
Regul Pept ; 146(1-3): 293-302, 2008 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-18068234

RESUMEN

As the content of Transforming Growth Factor-beta (TGFbeta) wanes in the milk of lactating rat, an increase in TGFbeta is observed in the gastric epithelia concomitant with differentiation of the glands upon weaning. Whereas TGFbeta has been shown to inhibit the proliferation of gastrointestinal cells in vitro, its functional significance and mechanisms of action have not been studied in vivo. Therefore, we administered TGFbeta1 (1 ng/g body wt.) to 14-day-old rats in which the gastric epithelium was induced to proliferate by fasting, and determined the involvement of signaling through Smads and the impact on epithelial cell proliferation and apoptosis. After the gavage, we observed the progressive increase of active TGFbeta1 while TbetaRII-receptor remained constant in the gastric mucosa. By immunohistochemistry, we showed Smad2/3 increase at 60 min (p<0.05) and Smad2 phosphorylation/activation and translocation to the nucleus most prominently between 0 and 30 min after treatment (p<0.05). Importantly, TGFbeta1 inhibited cell proliferation (p<0.05), which was estimated by BrDU pulse-labeling 12 h after gavage. Lower proliferation was reflected by increased p27(kip1) at 2 h (p<0.05). Also, TGFbeta1 increased apoptosis as measured by M30 labeling at 60 and 180 min (p<0.001), and by morphological features at 12 h (p<0.05). In addition, we observed higher levels of activated caspase 3 (17 kDa) from 0 to 30 min. Altogether, these data indicate a direct effect of TGFbeta1 signaling through Smads on both inhibiting proliferation, through alteration of cycle proteins, and inducing apoptosis of gastric epithelial cells in vivo. Further, the studies suggest a potential role for both milk and tissue-expressed TGFbeta1 in gastric growth during postnatal development.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/crecimiento & desarrollo , Factor de Crecimiento Transformador beta1/farmacología , Animales , Animales Lactantes , Apoptosis/efectos de los fármacos , Western Blotting , Mucosa Gástrica/citología , Antígeno Nuclear de Célula en Proliferación/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Proteínas Smad/efectos de los fármacos , Proteínas Smad/metabolismo
12.
Sci Rep ; 8(1): 9823, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29959361

RESUMEN

Neonatal- Maternal Separation (NMS) deprives mammals from breastfeeding and maternal care, influencing growth during suckling- weaning transition. In the gastric mucosa, Mist1 (encoded by Bhlha15 gene) and moesin organize the secretory apparatus for pepsinogen C in zymogenic cells. Our current hypothesis was that NMS would change corticosterone activity through receptors (GR), which would modify molecules involved in zymogenic cell differentiation in rats. We found that NMS increased corticosterone levels from 18 days onwards, as GR decreased in the gastric mucosa. However, as nuclear GR was detected, we investigated receptor binding to responsive elements (GRE) and observed an augment in NMS groups. Next, we demonstrated that NMS increased zymogenic population (18 and and 30 days), and targeted Mist1 and moesin. Finally, we searched for evolutionarily conserved sequences that contained GRE in genes involved in pepsinogen C secretion, and found that the genomic regions of Bhlha15 and PgC contained sites highly likely to be responsive to glucocorticoids. We suggest that NMS triggers GR- GRE to enhance the expression and to prime genes that organize cellular architecture in zymogenic population for PgC function. As pepsinogen C- pepsin is essential for digestion, disturbance of parenting through NMS might alter functions of gastric mucosa in a permanent manner.


Asunto(s)
Células Principales Gástricas/metabolismo , Corticosterona/metabolismo , Mucosa Gástrica/metabolismo , Privación Materna , Pepsinógeno C/metabolismo , Receptores de Glucocorticoides/metabolismo , Destete , Animales , Animales Recién Nacidos , Diferenciación Celular , Células Cultivadas , Células Principales Gástricas/citología , Femenino , Ratas
13.
Life Sci ; 202: 35-43, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29626530

RESUMEN

AIMS: High fat diet consumes and thyroid hormones (THs) disorders may affect nutrients metabolism, but their impact on the absorptive epithelium, the first place of nutrients access, remains unknown. Our aim was to evaluate the intestinal morphology and nutrients transporters content in mice fed standard (LFD) or high fat (HFD) diets in hypo or hyperthyroidism-induced condition. MATERIAL AND METHODS: C57BL/6 male mice fed LFD or HFD diets for 12 weeks, followed by saline, PTU (antithyroid drug) or T3 treatment up to 30 days. The mice were euthanized and proximal intestine was removed to study GLUT2, GLUT5, PEPT1, FAT-CD36, FATP4, NPC1L1 and NHE3 distribution by Western blotting. Since PPAR-a is activated by fatty acids, which is abundant in the HFD, we also evaluated whether PPAR-a affects nutrients transporters. Thus, mice were treated with fenofibrate, a PPAR-a agonist. KEY FINDINGS: HFD decreased GLUT2, PEPT1, FAT-CD6 and NPC1L1, but increased NHE3, while GLUT5 and FATP4 remained unaltered. THs did not alter distribution of nutrients transporters neither in LFD nor in HFD groups, but they increased villi length and depth crypt in LFD and HFD, respectively. Fenofibrate did not affect content of nutrients transporters, excluding PPAR-a involvement on the HFD-induced changes. SIGNIFICANCE: We assume that chronic HFD consumption reduced most of the nutrients transporters content in the small intestine of mice, which might limit the entrance of nutrients and gain weight. Since NHE3 promotes sodium absorption, and it was increased in HFD group, this finding could contribute to explain the hypertension observed in obesity.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hipertiroidismo/metabolismo , Hipotiroidismo/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Transporte de Membrana/metabolismo , PPAR alfa/metabolismo , Animales , Antitiroideos/farmacología , Fenofibrato/farmacología , Prueba de Tolerancia a la Glucosa , Hipertiroidismo/inducido químicamente , Hipolipemiantes/farmacología , Hipotiroidismo/inducido químicamente , Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Intestinos/efectos de los fármacos , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR alfa/antagonistas & inhibidores , Propiltiouracilo/farmacología , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Hormonas Tiroideas/metabolismo , Triyodotironina/farmacología
14.
Sci Rep ; 7: 45867, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28361902

RESUMEN

Gastric epithelial cells differentiate throughout the third postnatal week in rats, and become completely functional by weaning time. When suckling is interrupted by early weaning (EW), cell proliferation and differentiation change in the gastric mucosa, and regulatory mechanisms might involve corticosterone activity. Here we used EW and RU486 (glucocorticoid receptor antagonist) to investigate the roles of corticosterone on differentiation of mucous neck (MNC) and zymogenic cells (ZC) in rats, and to evaluate whether effects persisted in young adults. MNC give rise to ZC, and mucin 6, Mist1, pepsinogen a5 and pepsinogen C are produced to characterize these cells. We found that in pups, EW augmented the expression of mucins, Mist1 and pepsinogen C at mRNA and protein levels, and it changed the number of MNC and ZC. Corticosterone regulated pepsinogen C expression, and MNC and ZC distributions. Further, the changes on MNC population and pepsinogen C were maintained until early- adult life. Therefore, by using EW as a model for altered corticosterone activity in rats, we demonstrated that the differentiation of secretory epithelial cells is sensitive to the type of nutrient in the lumen. Moreover, this environmental perception activates corticosterone to change maturation and reprogram cellular functions in adulthood.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Corticosterona/metabolismo , Mucosa Gástrica/metabolismo , Destete , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Células Epiteliales/metabolismo , Ratas , Estómago/crecimiento & desarrollo
15.
Endocrinology ; 158(9): 2754-2773, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28911166

RESUMEN

We discovered that pigment epithelium-derived factor (PEDF)-null mice have endometrial hyperplasia, the precursor to human type I endometrial cancer (ECA), which is etiologically linked to unopposed estrogen (E2), suggesting that this potent antiangiogenic factor might contribute to dysregulated growth and the development of type I ECA. Treatment of both ECA cell lines and primary ECA cells with recombinant PEDF dose dependently decreased cellular proliferation via an autocrine mechanism by blocking cells in G1 and G2 phases of the cell cycle. Consistent with the known opposing effects of E2 and progesterone (Pg) on endometrial proliferation, Pg increases PEDF protein synthesis and release, whereas E2 has the converse effect. Using PEDF luciferase promoter constructs containing two Pg and one E2 response elements, E2 reduced and Pg increased promoter activity due to distal response elements. Furthermore, E2 decreases and Pg increases PEDF secretion into conditioned media (CM) by both normal endometrial stromal fibroblasts (ESFs) and cancer-associated fibroblasts (CAFs), but only CM from ESFs mediated growth-inhibitory activity of primary endometrial epithelial cells (EECs). In addition, in cocultures with primary EECs, Pg-induced growth inhibition is mediated by ESFs, but not CAFs. This is consistent with reduced levels of Pg receptors on CAFs surrounding human malignant glands in vivo. Taken together, the data suggest that PEDF is a hormone-regulated negative autocrine mediator of endometrial proliferation, and that paracrine growth inhibition by soluble factors, possibly PEDF, released by ESFs in response to Pg, but not CAFs, exemplifies a tumor microenvironment that contributes to the pathogenesis of ECA.


Asunto(s)
Carcinoma Endometrioide/patología , Proliferación Celular , Neoplasias Endometriales/patología , Endometrio , Células Epiteliales/fisiología , Proteínas del Ojo/fisiología , Hormonas/farmacología , Factores de Crecimiento Nervioso/fisiología , Serpinas/fisiología , Células del Estroma/fisiología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Endometrio/citología , Endometrio/efectos de los fármacos , Endometrio/metabolismo , Endometrio/patología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Estradiol/farmacología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Progesterona/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Células Tumorales Cultivadas
16.
J Mol Histol ; 37(3-4): 95-100, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-17063384

RESUMEN

The enteric nervous and enteroendocrine systems regulate different processes in the small intestine. Ablation of myenteric plexus with benzalkonium chloride (BAC) stimulates epithelial cell proliferation, whereas endocrine serotonin cells may inhibit the process. To evaluate the connection between the systems and the influence of myenteric plexus on serotoninergic cells in rats during postnatal development, the ileal plexus was partially removed with BAC. Rats were treated at 13 or 21 days and sacrificed after 15 days. The cell bodies of myenteric neurons were stained by beta NADH-diaphorase to detect the extension of denervation. The number of enteroendocrine cells in the ileum was estimated in crypts and villi in paraffin sections immunostained for serotonin. The number of neurons was reduced by 27.6 and 45% in rats treated on the 13th and 21st days, respectively. We tried to establish a correlation of denervation and the serotonin population according to the age of treatment. We observed a reduction of immunolabelled cells in the crypts of rats treated at 13 days, whereas this effect was seen in the villi of rats denervated at 21 days. These results suggest that the enteric nervous system might control the enteroendocrine cell population and this complex mechanism could be correlated to changes in cell proliferation.


Asunto(s)
Proliferación Celular , Desnervación , Células Enteroendocrinas/citología , Plexo Mientérico/fisiología , Serotonina/análisis , Animales , Compuestos de Benzalconio , Recuento de Células , Sistema Nervioso Entérico , Crecimiento y Desarrollo , Neuronas/citología , Ratas
17.
Regul Pept ; 135(1-2): 17-22, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16644034

RESUMEN

Glucocorticoids inhibit the cell proliferation in the gastric epithelium, and induce differentiation, migration and death. The mechanism by which these effects are triggered and controlled is still discussed and can involve the transcription and activation of transforming growth factor beta (TGFbeta). The present study was conducted to evaluate the effect of hydrocortisone short-term treatment on tissue level and distribution of TGFbeta isoforms, receptors and signaling through Smad2/3. To achieve that, 18-day-old rats were injected with hydrocortisone (50 mg/Kg b.wt.) for 0, 1 and 3 h. The stomachs were collected and processed for immunohistochemistry and western blotting. We observed that the treatment for 3 h increased the number of labeled epithelial cells for TGFbeta1 (p < 0.05), decreased the distribution of TGFbeta2 (p < 0.05) and did not alter TGFbeta3, TbetaRI and TbetaRII status. The levels of TGFbeta1 and receptors were checked by western blotting and results corroborate the immunodetection. We also found that phosphorylation of Smad2/3 into Smad2P increased after 3 h (p < 0.05), indicating that the high level TGFbeta1 was active on the cells. We suggest that glucocorticoids differentially regulate the expression of TGFbeta isoforms, receptors and signaling, and so TGFbeta1 might be involved in the inhibitory pathway triggered by the hormone.


Asunto(s)
Animales Lactantes/metabolismo , Mucosa Gástrica/fisiología , Hidrocortisona/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Lactantes/anatomía & histología , Femenino , Mucosa Gástrica/citología , Hidrocortisona/administración & dosificación , Embarazo , Isoformas de Proteínas/genética , Ratas , Ratas Wistar , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética
18.
Cancer Res ; 62(10): 2778-90, 2002 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12019154

RESUMEN

Transforming growth factor beta (TGF-beta), a potent ubiquitous endogenous inhibitor of epithelial cell growth, is secreted as a latent molecule (LTGF-beta)requiring activation for function. TGF-beta signals through the type I(TbetaRI) and type II (TbetaRII) receptors, which cooperate to phosphorylate/activate Smad2/3, the transcriptional regulators of genes that induce cell cycle arrest. That carcinomas grow in vivo suggests that they are refractory to TGF-beta. However, this has been difficult to prove because of an inability to analyze the functional status of TGF-beta in vivo as well as lack of close physiological paradigms for carcinoma cells in vitro. The current studies demonstrate that whereas primary cultures of endometrial epithelial cells derived from normal proliferative endometrium (PE; n = 10) were dose-dependently and maximally growth inhibited by 55% +/- 5.3% with 10 pM TGF-beta1, endometrial epithelial cells derived from endometrial carcinomas (ECAs; n = 10) were unresponsive (P < or = 0.0066). To determine the mechanism of TGF-beta resistance in ECAs, we analyzed the TGF-beta signaling pathway in vivo by immunohistochemistry using specific antibodies to TbetaRI and TbetaRII, Smads, and to the phosphorylated form of Smad2 (Smad2P), an indicator of cells responding to bioactive TGF-beta. Smad2P was expressed in all of the normal endometria (n = 25), and was localized to the cytoplasm and nucleus in PE, and only nuclear in the secretory endometrium. In marked contrast, Smad2P immunostaining was weak or undetectable in ECA (n = 22; P < or = 0.001) and reduced in glandular hyperplasia (n = 25) compared with normal endometrium. However, total Smad2 and Smad7 (which inhibits Smad2 activation) levels were identical in ECA and normal tissue. Consistent with loss of downstream signaling, both TbetaRI (n = 19) and TbetaRII (n = 22) protein expression were significantly reduced in ECA compared with PE (n = 11; P < or = 0.05). By in situ hybridization, the mRNA levels of TbetaRI and TbetaRII were decreased in the carcinoma cells compared with normal PE glands, suggesting that receptor down-regulation occurs at the transcriptional level. Furthermore, a somatic frameshift mutation in the polyadenine tract at the 5' end of the TbetaR-II gene was detected in two of six cases examined. Finally, the ability of explants of ECA to activate endogenous LTGF-beta was deficient compared with normal tissue (23.5% versus 7.4%). Therefore, our results suggest that loss of Smad2 signaling in ECA may be because of down-regulation of TbetaRI and TbetaRII, and/or decreased activation of LTGF-beta. Because disruption of TGF-beta signaling occurred independent of grade or degree of invasion and was evident in premalignant hyperplasia, we conclude that inactivation of TGF-beta signaling leading to escape from normal growth control occurs at an early stage in endometrial carcinogenesis, thereby defining novel molecular targets for cancer prevention.


Asunto(s)
Neoplasias Endometriales/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adulto , División Celular/fisiología , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/fisiología , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Endometrio/citología , Endometrio/metabolismo , Femenino , Mutación del Sistema de Lectura , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal/fisiología , Proteína Smad2 , Transactivadores/biosíntesis , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Células Tumorales Cultivadas
19.
Nutrition ; 32(1): 101-7, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26520918

RESUMEN

OBJECTIVES: Based on previous evidence showing that early weaning disturbs the ontogenesis of rat gastric glands, which are the major site of ghrelin synthesis, we investigated the distribution of ghrelin and its receptor (GHS-R) in the rat gastric epithelium during postnatal development and evaluated the effects of early weaning on their levels. Additionally, we studied the contribution of ghrelin to gastric growth during the abrupt nutrient transition. METHODS: Wistar rats were submitted to early weaning at 15 d and suckling counterparts were taken as controls. RESULTS: By running quantitative reverse transcription polymerase chain reaction, immunoblots, and immunohistochemistry, we detected a variation of ghrelin levels and an increase of expression and number of immunolabeled cells, 3 d after treatment (P < 0.05). Through confocal microscopy, we identified GHS-R in the neck region of the gland and did not observe changes in protein levels. Growth was evaluated after ghrelin antagonist ([D-Lys-3]-GHRP-6) administration, which reduced DNA synthesis index in early-weaned rats (P < 0.05) as determined by bromodeoxyuridine incorporation. CONCLUSION: The present study demonstrated that ghrelin and GHS-R are distributed in gastric mucosa during the postnatal development, indicating that they can signal and function in epithelial cells. We concluded that early weaning increased ghrelin levels in the stomach, and it takes part of cell proliferation control that is essential for stomach growth. Therefore, among the many effects previously described for early weaning, this abrupt nutrient transition also changed ghrelin levels, which might represent an additional element in the complex mechanism that coordinates stomach development.


Asunto(s)
Mucosa Gástrica/metabolismo , Ghrelina/metabolismo , Receptores de Ghrelina/metabolismo , Destete , Animales , Proliferación Celular , Células Epiteliales/metabolismo , Mucosa Gástrica/crecimiento & desarrollo , Ratas Wistar , Transducción de Señal , Estómago/crecimiento & desarrollo
20.
Cell Cycle ; 15(7): 931-47, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26963853

RESUMEN

We previously reported that aberrant TGF-ß/Smad2/3 signaling in endometrial cancer (ECA) leads to continuous ubiquitylation of p27(kip1)(p27) by the E3 ligase SCF-Skp2/Cks1 causing its degradation, as a putative mechanism involved in the pathogenesis of this cancer. In contrast, normal intact TGF-ß signaling prevents degradation of nuclear p27 by SCF-Skp2/Cks1 thereby accumulating p27 to block Cdk2 for growth arrest. Here we show that in ECA cell lines and normal primary endometrial epithelial cells, TGF-ß increases Cdh1 and its binding to APC/C to form the E3 ligase complex that ubiquitylates Cks1 and Skp2 prompting their proteasomal degradation and thus, leaving p27 intact. Knocking-down Cdh1 in ECA cell lines increased Skp2/Cks1 E3 ligase activity, completely diminished nuclear and cytoplasmic p27, and obviated TGF-ß-mediated inhibition of proliferation. Protein synthesis was not required for TGF-ß-induced increase in nuclear p27 and decrease in Cks1 and Skp2. Moreover, half-lives of Cks1 and Skp2 were extended in the Cdh1-depleted cells. These results suggest that the levels of p27, Skp2 and Cks1 are strongly or solely regulated by proteasomal degradation. Finally, an inverse relationship of low p27 and high Cks1 in the nucleus was shown in patients in normal proliferative endometrium and grade I-III ECAs whereas differentiated secretory endometrium showed the reverse. These studies implicate Cdh1 as the master regulator of TGF-ß-induced preservation of p27 tumor suppressor activity. Thus, Cdh1 is a potential therapeutic target for ECA and other human cancers showing an inverse relationship between Cks1/Skp2 and p27 and/or dysregulated TGF-ß signaling.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Quinasas CDC2-CDC28/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias Endometriales/enzimología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Proteínas Cdh1/biosíntesis , Proteínas Cdh1/genética , Proteínas Cdh1/metabolismo , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Núcleo Celular/enzimología , Núcleo Celular/genética , Proliferación Celular , Neoplasias Endometriales/metabolismo , Endometrio/enzimología , Endometrio/crecimiento & desarrollo , Endometrio/metabolismo , Células Epiteliales/enzimología , Células Epiteliales/metabolismo , Femenino , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA