Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Cell Physiol ; 238(10): 2304-2315, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37555566

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-35032139

ABSTRACT

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.


Subject(s)
Intestinal Mucosa , Paneth Cells , Cell Count , Intestines , Paneth Cells/metabolism , Stem Cell Niche
3.
Front Physiol ; 12: 721242, 2021.
Article in English | MEDLINE | ID: mdl-34588994

ABSTRACT

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.

4.
Physiol Rep ; 9(3): e14744, 2021 02.
Article in English | MEDLINE | ID: mdl-33580917

ABSTRACT

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.


Subject(s)
Breast Feeding/trends , COVID-19/metabolism , COVID-19/prevention & control , COVID-19/transmission , Female , Humans
5.
J Cachexia Sarcopenia Muscle ; 10(5): 1116-1127, 2019 10.
Article in English | MEDLINE | ID: mdl-31307125

ABSTRACT

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.


Subject(s)
Cachexia/etiology , Cachexia/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Neoplasms/complications , Aged , Biomarkers , Cachexia/metabolism , Cytokines/metabolism , Female , Humans , Immunohistochemistry , Inflammation , Inflammation Mediators , Male , Middle Aged , Neoplasms/metabolism , Proteome , Proteomics
6.
Int J Mol Sci ; 21(1)2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31892140

ABSTRACT

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.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/physiology , Gastric Mucosa/physiology , Stomach/physiology , Animals , Female , Gene Expression/physiology , Male , Rats , Rats, Wistar , Weaning
7.
Sci Rep ; 8(1): 9823, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29959361

ABSTRACT

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.


Subject(s)
Chief Cells, Gastric/metabolism , Corticosterone/metabolism , Gastric Mucosa/metabolism , Maternal Deprivation , Pepsinogen C/metabolism , Receptors, Glucocorticoid/metabolism , Weaning , Animals , Animals, Newborn , Cell Differentiation , Cells, Cultured , Chief Cells, Gastric/cytology , Female , Rats
8.
Life Sci ; 202: 35-43, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29626530

ABSTRACT

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.


Subject(s)
Diet, High-Fat/adverse effects , Hyperthyroidism/metabolism , Hypothyroidism/metabolism , Intestinal Mucosa/metabolism , Membrane Transport Proteins/metabolism , PPAR alpha/metabolism , Animals , Antithyroid Agents/pharmacology , Fenofibrate/pharmacology , Glucose Tolerance Test , Hyperthyroidism/chemically induced , Hypolipidemic Agents/pharmacology , Hypothyroidism/chemically induced , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestines/drug effects , Intestines/pathology , Male , Mice , Mice, Inbred C57BL , PPAR alpha/antagonists & inhibitors , Propylthiouracil/pharmacology , Sodium-Hydrogen Exchanger 3/metabolism , Thyroid Hormones/metabolism , Triiodothyronine/pharmacology
9.
Endocrinology ; 158(9): 2754-2773, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28911166

ABSTRACT

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.


Subject(s)
Carcinoma, Endometrioid/pathology , Cell Proliferation , Endometrial Neoplasms/pathology , Endometrium , Epithelial Cells/physiology , Eye Proteins/physiology , Hormones/pharmacology , Nerve Growth Factors/physiology , Serpins/physiology , Stromal Cells/physiology , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endometrium/cytology , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Estradiol/pharmacology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Progesterone/pharmacology , Stromal Cells/drug effects , Stromal Cells/pathology , Tumor Cells, Cultured
10.
Sci Rep ; 7: 45867, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28361902

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Corticosterone/metabolism , Gastric Mucosa/metabolism , Weaning , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Epithelial Cells/metabolism , Rats , Stomach/growth & development
11.
Cell Cycle ; 15(7): 931-47, 2016.
Article in English | MEDLINE | ID: mdl-26963853

ABSTRACT

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.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , CDC2-CDC28 Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endometrial Neoplasms/enzymology , S-Phase Kinase-Associated Proteins/metabolism , Transforming Growth Factor beta/physiology , Cdh1 Proteins/biosynthesis , Cdh1 Proteins/genetics , Cdh1 Proteins/metabolism , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cell Proliferation , Endometrial Neoplasms/metabolism , Endometrium/enzymology , Endometrium/growth & development , Endometrium/metabolism , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Female , Humans , Proteasome Endopeptidase Complex/metabolism
12.
Nutrition ; 32(1): 101-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26520918

ABSTRACT

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.


Subject(s)
Gastric Mucosa/metabolism , Ghrelin/metabolism , Receptors, Ghrelin/metabolism , Weaning , Animals , Cell Proliferation , Epithelial Cells/metabolism , Gastric Mucosa/growth & development , Rats, Wistar , Signal Transduction , Stomach/growth & development
13.
Rev. Soc. Bras. Clín. Méd ; 14(4): 225-229, 2016.
Article in Portuguese | LILACS | ID: biblio-827322

ABSTRACT

A linfo-histiocitose hematofagocítica é uma síndrome pouco comum, caracterizada por descontrolada ativação e proliferação imunopatológica, levando a evidências clínicas e laboratoriais de inflamação extrema. Pode ser causada primariamente por mutações genéticas (linfo-histiocitose hematofagocítica familiar) ou secundariamente, por uma condição esporádica (linfo--histiocitose hematofagocítica adquirida), como infecções e malignidades.O objetivo deste trabalho foi chamar a atenção para a hinfo-histiocitose hematofagocítica em sua forma secundária (adquirida), com discussão de relato de caso e breve revisão da literatura. Em razão da forma secundária da linfo-histiocitose hematofagocítica ser rara e letal, pouco difundida no meio médico-acadêmico, ter apresentação variável e possuir testes que exigem tempo necessário para o diagnóstico, ela constitui desafio para a realização do diagnóstico precoce e do pronto início da imunoquimioterapia necessária à sobrevivência. O tratamento é complicado por curso clínico dinâmico, alto risco de morbidade e recorrência da doença. O prognóstico geralmente é muito ruim, com evolução potencialmente letal em curto período de tempo se não tratada.


Hemophagocytic Lymphohistiocytosis (HLH) is an uncommon syndrome, characterized by uncontrolled immunopathologic activation and proliferation, leading to clinical and laboratory evidence of severe inflammation. It can be primarily caused by genetic mutations (familial HLH), or secondarily, by a sporadic condition (acquired HLH), such as an infection or malignancy. The purpose of the study is to draw the attention to hematophagocytic Lymphohistiocytosis in its secondary (acquired) form, discussing a case report and briefly reviewing the literature. Because the secondary form of hematophagocytic lymphohistiocytosis is rare and lethal, and poorly widespread in the medical-academic area, with variable appearance, and requiring time-consuming diagnostic tests, it represents a challenge for getting an early diagnosis, and immediately starting immunochemotherapy necessary for survival. Treatment is complicated by the dynamic clinical course, high morbidity risk and recurrence. The prognosis is generally very poor, with potentially fatal outcomes in short time if not treated.


Subject(s)
Humans , Female , Aged , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/therapy , Ferritins , Lymphohistiocytosis, Hemophagocytic/blood , Prognosis
14.
PLoS One ; 9(7): e101965, 2014.
Article in English | MEDLINE | ID: mdl-25000203

ABSTRACT

Throughout postnatal development, the gastric epithelium expresses Transforming Growth Factor beta1 (TGFß1), but it is also exposed to luminal peptides that are part of milk. During suckling period, fasting promotes the withdrawal of milk-born molecules while it stimulates gastric epithelial cell proliferation. Such response can be reversed by exogenous TGFß1, as it directly affects cell cycle through the regulation of p27 levels. We used fasting condition to induce the hyperproliferation of gastric epithelial cells in 14-day-old Wistar rats, and evaluated the effects of TGFß1 gavage on p27 expression, phosphorylation at threonine 187 (phospho-p27Thr187) and degradation. p27 protein level was reduced during fasting when compared to suckling counterparts, while phospho-p27Thr187/p27 ratio was increased. TGFß1 gavage reversed this response, which was confirmed through immunostaining. By using a neutralizing antibody against TGFß1, we found that it restored the p27 and phosphorylation levels detected during fasting, indicating the specific role of the growth factor. We noted that neither fasting nor TGFß1 changed p27 expression, but after cycloheximide administration, we observed that protein synthesis was influenced by TGFß1. Next, we evaluated the capacity of the gastric mucosa to degrade p27 and we recorded a higher concentration of the remaining protein in pups treated with TGFß1, suggesting augmented stability under this condition. Thus, we showed for the first time that luminal TGFß1 increased p27 levels in the rat gastric mucosa by up- regulating translation and reducing protein degradation. We concluded that such mechanisms might be used by rapidly proliferating cells to respond to milk-born TGFß1 and food restriction.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Blotting, Western , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/genetics , Male , Phosphorylation , Proteolysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/genetics
15.
Eur J Endocrinol ; 171(3): 335-42, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24920291

ABSTRACT

OBJECTIVE: To date, no evidence of robust genotype-phenotype correlation or disease modifiers for multiple endocrine neoplasia type 1 (MEN1) syndrome has been described, leaving the highly variable clinical presentation of patients unaccounted for. DESIGN: As the CDKN1B (p27) gene causes MEN4 syndrome and it is transcriptionally regulated by the product of the MEN1 gene (menin), we sought to analyze whether p27 influences the phenotype of MEN1-mutated patients. The cohort consisted of 100 patients carrying germline MEN1 gene mutations and 855 population-matched control individuals. METHODS: Genotyping of the coding p27 c.326T>G (V109G) variant was performed by sequencing and restriction site digestion, and the genotypes were associated with clinical parameters by calculating odds ratios (ORs) and their 95% CIs using logistic regression. RESULTS: There were significant differences in p27 V109G allele frequencies between controls and MEN1-mutated patients (OR=2.55, P=0.019, CI=1.013-5.76). Among patients who are ≥30 years old carrying truncating MEN1 mutations, the T allele was strongly associated with susceptibility to tumors in multiple glands (three to four glands affected vs one to two glands affected; OR=18.33; P=0.002, CI=2.88-16.41). This finding remained significant after the Bonferroni's multiple testing correction, indicating a robust association. No correlations were observed with the development of MEN1-related tumors such as hyperparathyroidism, pituitary adenomas, and enteropancreatic and adrenocortical tumors. CONCLUSIONS: Our study suggests that the p27 tumor suppressor gene acts as a disease modifier for the MEN1 syndrome associated with MEN1 germline mutations. If confirmed in independent patient cohorts, this finding could facilitate the management of this clinically complex disease.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/genetics , Genetic Association Studies/methods , Genetic Variation/genetics , Germ-Line Mutation/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Aged , Cohort Studies , Female , Humans , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/diagnosis , Young Adult
16.
Endocr Relat Cancer ; 21(3): 395-404, 2014 06.
Article in English | MEDLINE | ID: mdl-24532476

ABSTRACT

Germline mutations in p27(kip1) are associated with increased susceptibility to multiple endocrine neoplasias (MEN) both in rats and humans; however, the potential role of common polymorphisms of this gene in endocrine tumor susceptibility and tumorigenesis remains mostly unrecognized. To assess the risk associated with polymorphism rs2066827 (p27-V109G), we genotyped a large cohort of Brazilian patients with sporadic endocrine tumors (pituitary adenomas, n=252; pheochromocytomas, n=125; medullary thyroid carcinoma, n=51; and parathyroid adenomas, n=19) and 885 population-matched healthy controls and determined the odds ratios and 95% CIs. Significant associations were found for the group of patients with pituitary adenomas (P=0.01), particularly for those with ACTH-secreting pituitary adenomas (P=0.005). In contrast, no association was found with GH-secreting pituitary tumors alone or with the sporadic counterpart of MEN2-component neoplasias. Our in vitro analyses revealed increased colony formation and cell growth rate for an AtT20 corticotropin mouse cell line overexpressing the p27-V109G variant compared with cells transfected with the WT p27. However, the genotypic effects in genetic and in vitro approaches were divergent. In accordance with our genetic data showing specificity for ACTH-secreting pituitary tissues, the overexpression of p27-V109G in a GH3 somatotropin rat cell line resulted in no difference compared with the WT. Pituitary tumors are one of the major clinical components of syndromes associated with the p27 pathogenic mutations MENX and MEN4. Our genetic and in vitro data indicate that the common polymorphism rs2066827 may play a role in corticotropinoma susceptibility and tumorigenesis through a molecular mechanism not fully understood thus far.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/genetics , Mutation/genetics , Parathyroid Neoplasms/genetics , Pheochromocytoma/genetics , Pituitary Neoplasms/genetics , Thyroid Neoplasms/genetics , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Adult , Aged , Animals , Apoptosis , Blotting, Western , Carcinoma, Neuroendocrine , Case-Control Studies , Cohort Studies , Female , Fluorescent Antibody Technique , Humans , In Vitro Techniques , Male , Mice , Middle Aged , Multiple Endocrine Neoplasia/genetics , Multiple Endocrine Neoplasia/metabolism , Multiple Endocrine Neoplasia/pathology , Parathyroid Neoplasms/metabolism , Parathyroid Neoplasms/pathology , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Rats , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Cells, Cultured , Tumor Stem Cell Assay
17.
Nutrition ; 30(3): 343-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24484684

ABSTRACT

OBJECTIVES: The development of the gastrointestinal tract depends on many elements, including glucocorticoids. In the current study, we evaluated the effects of early weaning on corticosterone function and the growth of rat gastric mucosa. METHODS: By using Wistar rats submitted to early weaning at 15 d, we analyzed plasma corticosterone, corticosteroid-binding globulin (CBG), and glucocorticoid receptor (GR) distribution in the gastric epithelium. RESULTS: With the use of radioimmunoassay, we found that early weaning increased corticosterone concentration at day 16 and 17 in test subjects as compared with controls, whereas it was equivalent between groups at day 18. CBG binding capacity decreased during treatment, and it was significantly lower at day 18. At this age, GR levels and distribution in the gastric mucosa were also reduced as compared with suckling counterparts. To reduce corticosterone activity during early weaning and to explore cell proliferation responses, we administered RU486 to 15-d-old pups. We found that cytoplasmic GR reached a peak after 48 h, whereas nuclear levels remained constant, thereby confirming the inhibition of receptor function. Next, by checking gastric proliferative responses, we observed that RU486 induced higher DNA synthesis and mitotic indices in test subjects as compared with control groups. CONCLUSIONS: We demonstrated that early weaning changed corticosterone activity by increasing hormone levels, reducing CBG binding capacity, and decreasing GR distribution in the gastric epithelium. These modifications seem to be important to the reorganization of gastric growth after the abrupt interruption of suckling.


Subject(s)
Cell Proliferation , Corticosterone/physiology , Gastric Mucosa/metabolism , Animals , Female , Gastric Mucosa/cytology , Hormone Antagonists/pharmacology , Male , Mifepristone/pharmacology , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Transcortin/metabolism , Weaning
18.
PLoS One ; 8(6): e66651, 2013.
Article in English | MEDLINE | ID: mdl-23762493

ABSTRACT

During rat postnatal development, gastric cell proliferation and differentiation depend on many elements, which include dietary pattern, hormones, growth factors and their signaling pathways. Among them, EGFR activity is increased through MAPK and Src cascades in response to early weaning that represents the abrupt transition from milk to solid food. We herein investigated the direct involvement of ERK pathway in the control of cell cycle progression during early weaning, and studied the specific role of p27. At 15 days, Wistar rats were separated from dams, fed with powdered chow and daily injected with PD98059 (MEK inhibitor, 300 µg/kg) or 0.5% DMSO (control). By using HE staining and immunohistochemistry for PCNA, we respectively detected mitotic (MI) and proliferative (PI) indices in 18-day-old pups, and observed that both were reduced by PD98059. As cell cycle-related proteins (cyclin E, CDK2, cyclin D1, CDK4, p21 and p27) are involved in proliferative regulation, we compared samples obtained at 17 days in the morning (17 d) and evening (17.5 d). We found that they were not altered after ERK inhibition, but cyclin D1, p21 and p27 levels changed throughout the day in the control group. As p27 activity depends on its integrity, we studied p27 phosphorylation (threonin 187), and observed that ERK inhibition reduced this process. We suggest that MAPK pathway interferes in the regulation of p27 function in the gastric mucosa during early weaning, possibly by controlling its degradation, and altogether this mechanism might contribute to the increase of epithelial proliferation at this condition.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gastric Mucosa/cytology , Gastric Mucosa/enzymology , MAP Kinase Signaling System , Phosphothreonine/metabolism , Weaning , Animals , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flavonoids/pharmacology , Gastric Mucosa/drug effects , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects , Rats , Rats, Wistar
19.
Int J Mol Sci ; 14(5): 10143-61, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23670595

ABSTRACT

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.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells/cytology , Gastrointestinal Tract/cytology , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Animals , Epithelial Cells/metabolism , Gastrointestinal Tract/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism
20.
Nutrition ; 28(6): 707-12, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22209463

ABSTRACT

OBJECTIVE: Gastric development depends directly on the proliferation and differentiation of epithelial cells, and these processes are controlled by multiple elements, such as diet, hormones, and growth factors. Protein restriction affects gastrointestinal functions, but its effects on gastric growth are not fully understood. METHODS: The present study evaluated cell proliferation in the gastric epithelia of rats subjected to protein restriction since gestation. Because ghrelin is increasingly expressed from the fetal to the weaning stages and might be part of growth regulation, its distribution in the stomach of rats was investigated at 14, 30, and 50 d old. RESULTS: Although the protein restriction at 8% increased the intake of food and body weight, the body mass was lower (P < 0.05). The stomach and intestine were also smaller but increased proportionately throughout treatment. Cell proliferation was estimated through DNA synthesis and metaphase indices, and lower rates (P < 0.05) were detected at the different ages. The inhibition was concomitant with a larger number of ghrelin-immunolabeled cells at 30 and 50 d postnatally. CONCLUSION: Protein restriction impairs cell proliferation in the gastric epithelium, and a ghrelin upsurge under this condition is parallel to lower gastric and body growth rates.


Subject(s)
Diet, Protein-Restricted/adverse effects , Dietary Proteins/administration & dosage , Gastric Mucosa , Ghrelin/metabolism , Growth/physiology , Protein Deficiency/metabolism , Stomach , Animal Nutritional Physiological Phenomena/genetics , Animals , Animals, Newborn , Body Weight , Cell Proliferation , DNA/biosynthesis , Energy Intake , Gastric Mucosa/cytology , Gastric Mucosa/growth & development , Gastric Mucosa/metabolism , Intestinal Mucosa/metabolism , Intestines/growth & development , Metaphase , Organ Size , Protein Deficiency/etiology , Protein Deficiency/genetics , Rats , Stomach/cytology , Stomach/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...