Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 8(9)2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-37014710

RESUMO

RNA-binding protein 47 (RBM47) is required for embryonic endoderm development, but a role in adult intestine is unknown. We studied intestine-specific Rbm47-knockout mice (Rbm47-IKO) following intestinal injury and made crosses into ApcMin/+ mice to examine alterations in intestinal proliferation, response to injury, and tumorigenesis. We also interrogated human colorectal polyps and colon carcinoma tissue. Rbm47-IKO mice exhibited increased proliferation and abnormal villus morphology and cellularity, with corresponding changes in Rbm47-IKO organoids. Rbm47-IKO mice adapted to radiation injury and were protected against chemical-induced colitis, with Rbm47-IKO intestine showing upregulation of antioxidant and Wnt signaling pathways as well as stem cell and developmental genes. Furthermore, Rbm47-IKO mice were protected against colitis-associated cancer. By contrast, aged Rbm47-IKO mice developed spontaneous polyposis, and Rbm47-IKO ApcMin/+ mice manifested an increased intestinal polyp burden. RBM47 mRNA was decreased in human colorectal cancer versus paired normal tissue, along with alternative splicing of tight junction protein 1 mRNA. Public databases revealed stage-specific reduction in RBM47 expression in colorectal cancer associated independently with decreased overall survival. These findings implicate RBM47 as a cell-intrinsic modifier of intestinal growth, inflammatory, and tumorigenic pathways.


Assuntos
Colite , Neoplasias do Colo , Adulto , Camundongos , Humanos , Animais , Idoso , Camundongos Knockout , Colite/induzido quimicamente , Colite/genética , Neoplasias do Colo/genética , Carcinogênese/genética , Proliferação de Células , RNA Mensageiro/genética , Estresse Oxidativo , Proteínas de Ligação a RNA/genética
2.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G154-G168, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34816756

RESUMO

Loss of functional small bowel surface area causes short bowel syndrome (SBS), intestinal failure, and parenteral nutrition (PN) dependence. The gut adaptive response following resection may be difficult to predict, and it may take up to 2 yr to determine which patients will wean from PN. Here, we examined features of gut microbiota and bile acid (BA) metabolism in determining adaptation and ability to wean from PN. Stool and sera were collected from healthy controls and from patients with SBS (n = 52) with ileostomy, jejunostomy, ileocolonic, and jejunocolonic anastomoses fed with PN plus enteral nutrition or who were exclusively enterally fed. We undertook 16S rRNA gene sequencing, BA profiling, and 7α-hydroxy-4-cholesten-3-one (C4) quantitation with LC-MS/MS and serum amino acid analyses. Patients with SBS exhibited altered gut microbiota with reduced gut microbial diversity compared with healthy controls. We observed differences in the microbiomes of patients with SBS with ileostomy versus jejunostomy, jejunocolonic versus ileocolonic anastomoses, and PN dependence compared with those who weaned from PN. Stool and serum BA composition and C4 concentrations were also altered in patients with SBS, reflecting adaptive changes in enterohepatic BA cycling. Stools from patients who were weaned from PN were enriched in secondary BAs including deoxycholic acid and lithocholic aicd. Shifts in gut microbiota and BA metabolites may generate a favorable luminal environment in select patients with SBS, promoting the ability to wean from PN. Proadaptive microbial species and select BA may provide novel targets for patient-specific therapies for SBS.NEW & NOTEWORTHY Loss of intestinal surface area causes short bowel syndrome, intestinal failure, and parenteral nutrition dependence. We analyzed the gut microbiota and bile acid metabolome of a large cohort of short bowel syndrome adult patients with different postsurgical anatomies. We report a novel analysis of the microbiome of patients with ileostomy and jejunostomy. Enrichment of specific microbial and bile acid species may be associated with the ability to wean from parenteral nutrition.


Assuntos
Ácidos e Sais Biliares/metabolismo , Fezes/microbiologia , RNA Ribossômico 16S/metabolismo , Síndrome do Intestino Curto/metabolismo , Adaptação Fisiológica/fisiologia , Cromatografia Líquida , Microbioma Gastrointestinal/fisiologia , Humanos , Intestino Delgado/metabolismo , Metaboloma/fisiologia , Microbiota/fisiologia
3.
JCI Insight ; 5(23)2020 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-33141758

RESUMO

Loss of functional small bowel surface area following surgical resection for disorders such as Crohn's disease, intestinal ischemic injury, radiation enteritis, and in children, necrotizing enterocolitis, atresia, and gastroschisis, may result in short bowel syndrome, with attendant high morbidity, mortality, and health care costs in the United States. Following resection, the remaining small bowel epithelium mounts an adaptive response, resulting in increased crypt cell proliferation, increased villus height, increased crypt depth, and enhanced nutrient and electrolyte absorption. Although these morphologic and functional changes are well described in animal models, the adaptive response in humans is less well understood. Clinically the response is unpredictable and often inadequate. Here we address the hypotheses that human intestinal stem cell populations are expanded and that the stem cell niche is regulated following massive gut resection in short bowel syndrome (SBS). We use intestinal enteroid cultures from patients with SBS to show that the magnitude and phenotype of the adaptive stem cell response are both regulated by stromal niche cells, including intestinal subepithelial myofibroblasts, which are activated by intestinal resection to enhance epithelial stem and proliferative cell responses. Our data suggest that myofibroblast regulation of bone morphogenetic protein signaling pathways plays a role in the gut adaptive response after resection.


Assuntos
Células-Tronco Adultas/metabolismo , Mucosa Intestinal/metabolismo , Síndrome do Intestino Curto/fisiopatologia , Células-Tronco Adultas/fisiologia , Idoso , Doença de Crohn/metabolismo , Enterite/metabolismo , Feminino , Humanos , Mucosa Intestinal/crescimento & desenvolvimento , Intestinos , Masculino , Pessoa de Meia-Idade , Miofibroblastos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Síndrome do Intestino Curto/metabolismo , Transdução de Sinais
5.
Sci Rep ; 10(1): 3842, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32123209

RESUMO

The development and physiologic role of small intestine (SI) vasculature is poorly studied. This is partly due to a lack of targetable, organ-specific markers for in vivo studies of two critical tissue components: endothelium and stroma. This challenge is exacerbated by limitations of traditional cell culture techniques, which fail to recapitulate mechanobiologic stimuli known to affect vessel development. Here, we construct and characterize a 3D in vitro microfluidic model that supports the growth of patient-derived intestinal subepithelial myofibroblasts (ISEMFs) and endothelial cells (ECs) into perfused capillary networks. We report how ISEMF and EC-derived vasculature responds to physiologic parameters such as oxygen tension, cell density, growth factors, and pharmacotherapy with an antineoplastic agent (Erlotinib). Finally, we demonstrate effects of ISEMF and EC co-culture on patient-derived human intestinal epithelial cells (HIECs), and incorporate perfused vasculature into a gut-on-a-chip (GOC) model that includes HIECs. Overall, we demonstrate that ISEMFs possess angiogenic properties as evidenced by their ability to reliably, reproducibly, and quantifiably facilitate development of perfused vasculature in a microfluidic system. We furthermore demonstrate the feasibility of including perfused vasculature, including ISEMFs, as critical components of a novel, patient-derived, GOC system with translational relevance as a platform for precision and personalized medicine research.


Assuntos
Capilares/crescimento & desenvolvimento , Técnicas de Cocultura/instrumentação , Intestino Delgado/citologia , Dispositivos Lab-On-A-Chip , Miofibroblastos/citologia , Humanos , Miofibroblastos/metabolismo , Oxigênio/metabolismo , Perfusão
6.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G185-G194, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29631377

RESUMO

Stem cell therapy is a potential therapeutic approach for disorders characterized by intestinal injury or loss of functional surface area. Stem cell function and proliferation are mediated by the stem cell niche. Stromal cells such as intestinal subepithelial myofibroblasts (ISEMFs) are important but poorly studied components of the stem cell niche. To examine the role of ISEMFs, we have previously generated mice with deletion of epimorphin ( Epim), an ISEMF protein and member of the syntaxin family of intracellular vesicle docking proteins that regulate cell secretion. Herein we explore the mechanisms for previous observations that Epim deletion increases gut crypt cell proliferation, crypt fission, and small bowel length in vivo. Stem cell-derived crypt culture techniques were used to explore the interaction between enteroids and myofibroblasts from Epim-/- and WT mice. Enteroids cocultured with ISEMFS had increased growth and crypt-like budding compared with enteroids cultured without stromal support. Epim deletion in ISEMFs resulted in increased enteroid budding and surface area compared with cocultures with wild-type (WT) ISEMFs. In primary crypt cultures, Epim-/- enteroids had significantly increased surface area and budding compared with WTs. However, stem cell assays comparing the number of Epim-/- vs. WT colony-forming units after first passage showed no differences in the absence of ISEMF support. Epim-/- vs. WT ISEMFs had increased Wnt4 expression, and addition of Wnt4 to WT cocultures enhanced budding. We conclude that ISEMFs play an important role in the stem cell niche. Epim regulates stem cell proliferation and differentiation via stromal contributions to the niche microenvironment. NEW & NOTEWORTHY The role of subepithelial intestinal myofibroblasts (ISEMFs) in the gut stem cell niche is controversial. We provide novel evidence supporting ISEMFs as important niche contributors. We show that the in vivo intestinal effects of deletion of myofibroblast Epim can be recapitulated in crypt stem cell cultures in vitro. ISEMFs support cocultured stem cell proliferation and enteroid growth, and these effects are augmented by deletion of Epim, a syntaxin that regulates myofibroblast cell secretion.


Assuntos
Mucosa Intestinal/metabolismo , Intestino Delgado/citologia , Glicoproteínas de Membrana/metabolismo , Miofibroblastos/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Microambiente Celular/fisiologia , Camundongos
7.
Mol Cell Biochem ; 376(1-2): 163-73, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23361362

RESUMO

The two splice variants of human glucose transporter 9 (hGLUT9) are targeted to different polarized membranes. hGLUT9a traffics to the basolateral membrane, whereas hGLUT9b traffics to the apical region. This study examines the sorting mechanism of these variants, which differ only in their N-terminal domain. Mutating a di-leucine motif unique to GLUT9a did not affect targeting. Chimeric proteins were made using GLUT1, a basolaterally targeted transporter, and GLUT3, an apically targeted protein whose signal lies in the C-terminus. Overexpression of the chimeric proteins in polarized cells demonstrates that the N-terminus of hGLUT9b contains a signal capable of redirecting GLUT1 to the apical membrane. The N-terminus of hGLUT9a, however, does not contain a basolateral signal sufficient enough to redirect GLUT3. Portions of the GLUT9a N-terminus were substituted with corresponding portions of the GLUT9b N-terminus to determine the motif responsible for apical targeting. The first 16 amino acids were not found to be a sufficient apical signal. The last ten amino acids of the N-termini differ only in amino-acid class at one location. In the B-form, leucine, a hydrophobic residue, is substituted for lysine, a basic residue, found in the A-form. However, mutation of the leucine in hGLUT9b to a lysine resulted in retention of the apical signal. We therefore believe the apical signal exists as an interplay between the final ten amino acids of the N-terminus and another motif within the protein such as the intracellular loop or other motifs within the N-terminus.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Sinais Direcionadores de Proteínas , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Membrana Celular/metabolismo , Cães , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Leucina/metabolismo , Dados de Sequência Molecular , Mutação , Transporte Proteico
8.
Blood ; 121(9): 1651-62, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23297128

RESUMO

Energy metabolism in RBCs is characterized by O2-responsive variations in flux through the Embden Meyerhof pathway (EMP) or the hexose monophosphate pathway (HMP). Therefore, the generation of ATP, NADH, and 2,3-DPG (EMP) or NADPH (HMP) shift with RBC O2 content because of competition between deoxyhemoglobin and key EMP enzymes for binding to the cytoplasmic domain of the Band 3 membrane protein (cdB3). Enzyme inactivation by cdB3 sequestration in oxygenated RBCs favors HMP flux and NADPH generation (maximizing glutathione-based antioxidant systems). We tested the hypothesis that sickle hemoglobin disrupts cdB3-based regulatory protein complex assembly, creating vulnerability to oxidative stress. In RBCs from patients with sickle cell anemia, we demonstrate in the present study constrained HMP flux, NADPH, and glutathione recycling and reduced resilience to oxidative stress manifested by membrane protein oxidation and membrane fragility. Using a novel, inverted membrane-on-bead model, we illustrate abnormal (O2-dependent) association of sickle hemoglobin to RBC membrane that interferes with sequestration/inactivation of the EMP enzyme GAPDH. This finding was confirmed by immunofluorescent imaging during RBC O2 loading/unloading. Moreover, selective inhibition of inappropriately dispersed GAPDH rescues antioxidant capacity. Such disturbance of cdB3-based linkage between O2 gradients and RBC metabolism suggests a novel mechanism by which hypoxia may influence the sickle cell anemia phenotype.


Assuntos
Antioxidantes/metabolismo , Eritrócitos/metabolismo , Glicólise , Hemoglobina Falciforme/fisiologia , Oxigênio/metabolismo , Adolescente , Adulto , Estudos de Casos e Controles , Criança , Pré-Escolar , Eritrócitos/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Hemoglobina Falciforme/efeitos adversos , Hemoglobina Falciforme/farmacologia , Humanos , Modelos Biológicos , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Adulto Jovem
9.
Hepatology ; 56(4): 1489-98, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22707117

RESUMO

UNLABELLED: Mice subjected to partial hepatectomy (PH) develop hypoglycemia, followed by increased systemic lipolysis and hepatic fat accumulation, prior to onset of hepatocellular proliferation. Strategies that disrupt these metabolic events inhibit regeneration. These observations suggest that alterations in metabolism in response to hepatic insufficiency promote liver regeneration. Hepatic expression of the peroxisome proliferator-activated receptor gamma (PPARγ) influences fat accumulation in the liver. Therefore, the studies reported here were undertaken to assess the effects of disruption of hepatic PPARγ expression on hepatic fat accumulation and hepatocellular proliferation during liver regeneration. The results showed that liver regeneration was not suppressed, but rather modestly augmented in liver-specific PPARγ null mice maintained on a normal diet. These animals also exhibited accelerated hepatic cyclin D1 expression. Because hepatic PPARγ expression is increased in experimental models of fatty liver disease in which liver regeneration is impaired, regeneration in liver-specific PPARγ null mice with chronic hepatic steatosis was also examined. In contrast to the results described above, disruption of hepatic PPARγ expression in mice with diet-induced hepatic steatosis resulted in significant suppression of hepatic regeneration. CONCLUSION: The metabolic and hepatocellular proliferative responses to PH are modestly augmented in liver-specific PPARγ null mice, thus providing additional support for a metabolic model of liver regeneration. Furthermore, regeneration is significantly impaired in liver-specific PPARγ null mice in the setting of diet-induced chronic steatosis, suggesting that pharmacological strategies to augment hepatic PPARγ activity might improve regeneration of the fatty liver.


Assuntos
Fígado Gorduroso/etiologia , Hepatectomia/métodos , Regeneração Hepática/efeitos dos fármacos , Regeneração Hepática/fisiologia , PPAR gama/metabolismo , Tecido Adiposo/metabolismo , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Fígado Gorduroso/patologia , Regulação da Expressão Gênica , Glucose/farmacologia , Hepatectomia/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , RNA Mensageiro/metabolismo , Distribuição Aleatória , Sensibilidade e Especificidade , Triglicerídeos/análise , Triglicerídeos/metabolismo
10.
Am J Pathol ; 180(2): 575-82, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22155110

RESUMO

The relationship between liver and body mass is exemplified by the precision with which the liver:body mass ratio is restored after partial hepatic resection. Nevertheless, the compartments, against which liver mass is so exquisitely regulated, currently remain undefined. In the studies reported here, we investigated the role of skeletal muscle mass in the regulation of liver:body mass ratio and liver regeneration via the analysis of myostatin-null mice, in which skeletal muscle is hypertrophied. The results showed that liver mass is comparable and liver:body mass significantly diminished in the null animals compared to age-, sex-, and strain-matched controls. In association with these findings, basal hepatic Akt signaling is decreased, and the expression of the target genes of the constitutive androstane receptor and the integrin-linked kinase are dysregulated in the myostatin-null mice. In addition, the baseline expression levels of the regulators of the G1-S phase cell cycle progression in liver are suppressed in the null mice. The initiation of liver regeneration is not impaired in the null animals, although it progresses toward the lower liver:body mass set point. The data show that skeletal muscle is not the body component against which liver mass is positively regulated, and thus they demonstrate a previously unrecognized systemic compartmental specificity for the regulation of liver:body mass ratio.


Assuntos
Composição Corporal/fisiologia , Regeneração Hepática/fisiologia , Fígado/fisiologia , Músculo Esquelético/fisiologia , Animais , Ciclinas/metabolismo , Feminino , Hepatectomia , Fígado/anatomia & histologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos , Miostatina/deficiência , Tamanho do Órgão/fisiologia
11.
Hepatology ; 52(6): 2109-17, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20967828

RESUMO

UNLABELLED: We previously reported that mice subjected to partial hepatectomy exhibit rapid development of hypoglycemia followed by transient accumulation of fat in the early regenerating liver. We also showed that disrupting these metabolic alterations results in impaired liver regeneration. The studies reported here were undertaken to further characterize and investigate the functional importance of changes in systemic adipose metabolism during normal liver regeneration. The results showed that a systemic catabolic response is induced in each of two distinct, commonly used experimental models of liver regeneration (partial hepatectomy and carbon tetrachloride treatment), and that this response occurs in proportion to the degree of induced hepatic insufficiency. Together, these observations suggest that catabolism of systemic adipose stores may be essential for normal liver regeneration. To test this possibility, we investigated the hepatic regenerative response in fatty liver dystrophy (fld) mice, which exhibit partial lipodystrophy and have diminished peripheral adipose stores. The results showed that the development of hypoglycemia and hepatic accumulation of fat was attenuated and liver regeneration was impaired following partial hepatectomy in these animals. The fld mice also exhibited increased hepatic p21 expression and diminished plasma levels of the adipose-derived hormones adiponectin and leptin, which have each been implicated as regulators of liver regeneration. CONCLUSION: These data suggest that the hypoglycemia that develops after partial hepatectomy induces systemic lipolysis followed by accumulation of fat derived from peripheral stores in the early regenerating liver, and that these events may be essential for initiation of normal liver regeneration.


Assuntos
Fígado Gorduroso/fisiopatologia , Lipodistrofia/fisiopatologia , Regeneração Hepática/fisiologia , Adiponectina/sangue , Tecido Adiposo/fisiologia , Animais , Intoxicação por Tetracloreto de Carbono/fisiopatologia , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Hepatectomia , Hipoglicemia/fisiopatologia , Leptina/sangue , Camundongos
12.
Hepatology ; 50(1): 207-15, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19441104

RESUMO

UNLABELLED: The inhibitory effect of dextrose supplementation on liver regeneration was first described more than 4 decades ago. Nevertheless, the molecular mechanisms responsible for this observation have not been elucidated. We investigated these mechanisms using the partial hepatectomy model in mice given standard or 10% dextrose (D10)-supplemented drinking water. The results showed that D10-treated mice exhibited significantly reduced hepatic regeneration compared with controls, as assessed by hepatocellular bromodeoxyuridine (BrdU) incorporation and mitotic frequency. D10 supplementation did not suppress activation of hepatocyte growth factor (HGF), induction of transforming growth factor alpha (TGF-alpha) expression, or tumor necrosis factor alpha-interleukin-6 cytokine signaling, p42/44 extracellular signal-regulated kinase (ERK) activation, immediate early gene expression, or expression of CCAAT/enhancer binding protein beta (C/EBPbeta), but did augment expression of the mito-inhibitory factors C/EBPalpha, p21(Waf1/Cip1), and p27(Kip1). In addition, forkhead box M1 (FoxM1) expression, which is required for normal liver regeneration, was suppressed by D10 treatment. Finally, D10 did not suppress either FoxM1 expression or hepatocellular proliferation in p21 null mice subjected to partial hepatectomy, establishing the functional significance of these events in mediating the effects of D10 on liver regeneration. CONCLUSION: These data show that the inhibitory effect of dextrose supplementation on liver regeneration is associated with increased expression of C/EBPalpha, p21, and p27, and decreased expression of FoxM1, and that D10-mediated inhibition of liver regeneration is abrogated in p21-deficient animals. Our observations are consistent with a model in which hepatic sufficiency is defined by homeostasis between the energy-generating capacity of the liver and the energy demands of the body mass, with liver regeneration initiated when the functional liver mass is no longer sufficient to meet such demand.


Assuntos
Inibidor de Quinase Dependente de Ciclina p21/fisiologia , Glucose/farmacologia , Regeneração Hepática/efeitos dos fármacos , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
13.
Isr Med Assoc J ; 10(12): 858-61, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19160942

RESUMO

BACKGROUND: Insulin-dependent diabetes mellitus is dominated by a Th1 response whereas atopic diseases such as asthma, eczema and allergic rhinitis are characterized by a Th2 response. Because it is known that Th1 and Th2 cells reciprocally counteract each other, it can be speculated that the prevalence of Th2-mediated diseases is lower in patients with a Th1-mediated disease. OBJECTIVES: To compare the prevalence of atopic diseases among children with IDDM and age-matched controls. METHODS: The study group comprised 65 children with IDDM attending the pediatric endocrinology clinic at the Wolfson Medical Center. The control group consisted of 74 non-diabetic children who presented at the emergency room due to an acute illness (burns, abdominal pain, fever, head trauma). Patients were asked to complete a detailed questionnaire on their history of personal and familial atopic and autoimmune diseases. In addition, a total serum immunoglobulin E concentration and the presence of IgE antibodies to a panel of relevant inhalant allergens were analyzed. RESULTS: Children with IDDM and their first-degree relatives had a significantly higher prevalence of other autoimmune diseases such as thyroiditis and celiac as compared to controls. The two groups had a similar prevalence of atopic diseases with respect to history, total serum IgE, or the presence of IgE antibodies to a panel of relevant inhalant allergens. CONCLUSIONS: The prevalence of atopic diseases in IDDM patients was similar to that in the normal population. Our results suggest that the traditional Th1/Th2 theory to explain the complexity of the immune response is oversimplified.


Assuntos
Doenças Autoimunes/complicações , Doenças Autoimunes/imunologia , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/imunologia , Adolescente , Doenças Autoimunes/sangue , Autoimunidade/imunologia , Autoimunidade/fisiologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Feminino , Humanos , Imunoglobulina E/sangue , Lactente , Israel/epidemiologia , Masculino , Prevalência , Células Th2/imunologia , Células Th2/fisiologia , Adulto Jovem
14.
Neurobiol Dis ; 28(2): 184-96, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17714952

RESUMO

Mice with inactivation of the Tuberous sclerosis complex-1 (Tsc1) gene in glia (Tsc1 GFAP CKO mice) have deficient astrocyte glutamate transporters and develop seizures, suggesting that abnormal glutamate homeostasis contributes to neurological abnormalities in these mice. We examined the hypothesis that Tsc1 GFAP CKO mice have elevated extracellular brain glutamate levels that may cause neuronal death, abnormal glutamatergic synaptic function, and associated impairments in behavioral learning. In vivo microdialysis documented elevated glutamate levels in hippocampi of Tsc1 GFAP CKO mice and several cell death assays demonstrated neuronal death in hippocampus and neocortex. Impairment of long-term potentiation (LTP) with tetanic stimulation was observed in hippocampal slices from Tsc1 GFAP CKO mice and was reversed by low concentrations of NMDA antagonist, indicating that excessive synaptic glutamate directly inhibited LTP. Finally, Tsc1 GFAP CKO mice exhibited deficits in two hippocampal-dependent learning paradigms. These results suggest that abnormal glutamate homeostasis predisposes to excitotoxic cell death, impaired synaptic plasticity and learning deficits in Tsc1 GFAP CKO mice.


Assuntos
Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Deficiências da Aprendizagem/metabolismo , Plasticidade Neuronal/genética , Transmissão Sináptica/genética , Esclerose Tuberosa/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/fisiopatologia , Modelos Animais de Doenças , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Homeostase/genética , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/fisiopatologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/genética , Camundongos , Camundongos Knockout , Neocórtex/metabolismo , Neocórtex/fisiopatologia , Degeneração Neural/genética , Degeneração Neural/metabolismo , Degeneração Neural/fisiopatologia , Técnicas de Cultura de Órgãos , Esclerose Tuberosa/genética , Esclerose Tuberosa/fisiopatologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
15.
Brain Res ; 1130(1): 197-205, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17174280

RESUMO

Interpretation of the cellular and molecular pathogenic basis of post-minimal traumatic brain injury is a significant clinical and scientific problem, especially due to the high prevalence of motor vehicle--and other accidents. Pathogenetic brain mechanisms following traumatic impact are usually investigated by using models of severe or moderate trauma. Apoptotic neuronal degeneration after notable brain trauma is a well-known phenomenon, but the source of its activation is not clear, especially after mild, subclinical brain trauma. In the present study, we used a closed head weight-drop model to induce minimal brain injury in mice. Pellets of 5, 10, 15, 20, 25 and 30 g were dropped on the right side of mice's head kept under light ether anesthesia. No abnormal behavioral or neurophysiological changes were seen following the head trauma. Morphological assessment was done 72 h after the traumatic impact using TUNEL assay and silver staining. We found gradual increase of TUNEL-positive and silver-impregnated cells number in different cortical and hippocampal regions of both injured and contralateral hemispheres. The threshold of traumatic impact that caused a significant activation was 10-15 g pellets (evident by silver staining), and 15-20 g for apoptosis. The most sensitive zones for trauma were anterior cingulate cortex and CA3 area of hippocampus. No bilateral hemispheric differences were found. Our results demonstrate that even closed head minimal traumatic brain injury can cause diffused neuronal damage and apoptosis. This results correlate well with cognitive and behavioral deficits described for mice suffering similar mTBI and can also explain the wide variety of mental disturbances described for post-concussion syndrome in patients who suffered mild head injury.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/patologia , Córtex Cerebral/patologia , Hipocampo/patologia , Degeneração Neural/patologia , Animais , Córtex Cerebral/lesões , Modelos Animais de Doenças , Traumatismos Cranianos Fechados/patologia , Hipocampo/lesões , Marcação In Situ das Extremidades Cortadas , Masculino , Camundongos , Camundongos Endogâmicos ICR , Índice de Gravidade de Doença
16.
J Basic Clin Physiol Pharmacol ; 16(4): 231-43, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16438390

RESUMO

UNLABELLED: The activity of N-methyl-D-aspartate (NMDA) receptors is critical for neuronal survival in the immature brain. Studies have reported that chronic blockage of these receptors mediates apoptosis in neonatal animals. We investigated the apoptotic effect of a clinically relevant single dose of ketamine, an NMDA receptor antagonist, in the brain of neonatal mice. Seven-day-old ICR mice were injected with ketamine (1.25, 2.5, 5, 10, 20, and 40 mg/kg body weight, subcutaneously in 0.9% NaCl) or with 0.9% NaCl alone as control. Righting reflex testing was performed and mouse brains were examined at 24, 48, and 72 h and 7 days after injection. The number of degenerating neurons was measured using silver staining. Apoptosis was confirmed by DNA fragmentation (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling). We observed in the sensorimotor cortex and cerebellum of ketamine-treated mice extensive apoptosis, which was clearly dose-dependent and present even after a low dose of ketamine (5 mg/kg). The most prominent apoptotic damage was detected 72 h post-injection (P < 0.001 vs control), at doses ranging from 10 to 40 mg/kg. After 7 d the number of neurodegenerative neurons, at doses ranging from 5 to 40 mg/kg, remained significantly high. The brain weight was comparable to that of untreated control mice and no gross neurobehavioral effects in the righting reflex test or alteration in the pattern of behavior was observed. The results indicate that the administration of ketamine in a clinically relevant single dose triggers long-lasting neuronal apoptosis in certain brain areas of neonatal mice. IMPLICATIONS: The administration of ketamine in a clinically relevant single dose to 7-d-old mice induced apoptosis in the sensorimotor cortex and cerebellum. This effect was dose-dependent and long lasting.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/patologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ketamina/farmacologia , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Corantes , Amarelo de Eosina-(YS) , Hematoxilina , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos ICR , Degeneração Neural/patologia , Tamanho do Órgão/efeitos dos fármacos , Coloração pela Prata
17.
Pediatr Res ; 55(3): 372-9, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14681492

RESUMO

Mechanisms underlying cognitive dysfunction in young diabetic children are poorly understood, and may include synaptic dysfunction from insulin-induced hypoglycemia. We developed a model of repetitive insulin-induced hypoglycemia in young rats and examined hippocampal long-term potentiation, an electrophysiologic assay of synaptic plasticity, 3-5 d after the last hypoglycemic event. Three hypoglycemic events between postnatal d 21-25 produced modest cortical (17 +/- 2.9 dead neurons per section in parasagittal cortex), but not hippocampal, neuron death quantified by Fluoro-Jade B staining. There was no change in neurogenesis in the hippocampal dentate granule cell region by quantification of bromodeoxyuridine incorporation. Although normal baseline hippocampal synaptic responses were elicited from hippocampal slices from hypoglycemic animals, long-term synaptic potentiation could not be induced in hippocampal slices from rats subjected to hypoglycemia. These results suggest that repetitive hypoglycemia in the developing brain can cause selective impairment of synaptic plasticity in the absence of cell death, and without complete disruption of basal synaptic transmission. We speculate that impaired synaptic plasticity in the hippocampus caused by repetitive hypoglycemia could underlie memory and cognitive deficits observed in young diabetic children, and that cortical neuron death caused by repetitive hypoglycemia in the developing brain may contribute to other neurologic, cognitive, and psychological problems sometimes encountered in diabetic children.


Assuntos
Hipocampo/fisiopatologia , Hipoglicemia/fisiopatologia , Hipotálamo/fisiopatologia , Potenciação de Longa Duração , Animais , Potenciais Pós-Sinápticos Excitadores , Memória , Ratos , Ratos Sprague-Dawley
18.
J Cell Biochem ; 90(5): 957-63, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14624455

RESUMO

Previous investigation demonstrated the potential of beta-phenylpyruvate at high concentration to cause hypoglycemia in mice totally deprived of insulin. For further elucidation of the glucose-lowering mechanism, glucose uptake, and quantity of glucose transporters (GLUT1 and GLUT4) in mouse soleus muscle and C2C12 muscle cell lines were investigated following incubation with beta-phenylpyruvate in various concentrations. A marked enhancement of glucose uptake was demonstrated that peaked at 0.5 and 1.0 mM beta-phenylpyruvate in soleus muscle (P<0.01) and C2C12 cells (P<0.001), respectively. Kinetic analysis in C2C12 cells showed a twofold increase in Vmax compared with controls (P<0.001). In addition, both GLUT1 and GLUT4 levels were increased following exposure to beta-phenylpyruvate. Our findings point to a peripheral hypoglycemic effect of beta-phenylpyruvate.


Assuntos
Glucose/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Células Musculares/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Ácidos Fenilpirúvicos/metabolismo , Animais , Células Cultivadas , Transportador de Glucose Tipo 1 , Transportador de Glucose Tipo 4 , Cinética , Camundongos , Camundongos Endogâmicos ICR , Células Musculares/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos
19.
Metab Brain Dis ; 18(3): 221-31, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14567472

RESUMO

Previous investigation demonstrated the potential of L-cysteine (L-Cys) at high concentrations to cause hypoglycemia in mice totally deprived of insulin. For further elucidation of the glucose-lowering mechanism, glucose uptake and quantity of glucose transporters (GLUTs 3 and 4) in mouse soleus muscle and C2C12 muscle cells, as well as in human SH-SY5Y neuroblastoma cells, were investigated. A marked enhancement of glucose uptake was demonstrated, peaking at 5.0 mM L-Cys in soleus muscle (P < 0.05) and SH-SY5Y cells (P < 0.001), respectively. In contrast, glucose uptake was not affected in the C2C12 muscle cells. Kinetic analysis of the SH-SY5Y glucose uptake showed a 2.5-fold increase in maximum transport velocity compared with controls (P < 0.001). In addition, both GLUT3 and GLUT4 levels were increased following exposure to L-Cys. Our findings point to a possible hypoglycemic effect of L-Cys.


Assuntos
Cisteína/farmacologia , Glucose/metabolismo , Proteínas Musculares , Músculo Esquelético/metabolismo , Proteínas do Tecido Nervoso , Oxazinas , Xantenos , Animais , Western Blotting , Células Cultivadas , Corantes , Transportador de Glucose Tipo 3 , Transportador de Glucose Tipo 4 , Humanos , Cinética , Camundongos , Camundongos Endogâmicos ICR , Proteínas de Transporte de Monossacarídeos/metabolismo , Músculo Esquelético/efeitos dos fármacos , Neuroblastoma/metabolismo , Células Tumorais Cultivadas
20.
Pharmacol Biochem Behav ; 75(4): 795-9, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12957221

RESUMO

We investigated whether structural central neural damage and long-term neurobehavioral deficits after L-cysteine (L-Cys) administration in mice is caused by hypoglycemia. Neonatal ICR mice were injected subcutaneously with L-Cys (0.5-1.5 mg/g body weight [BW]) or saline (control). Blood glucose was measured. At 50 days of age, mice were introduced individually into an eight-arm maze for evaluation of spatial memory (hippocampal-related behavior). Times for visiting all eight arms and number of entries until completion of the eight-arm visits (maze criteria) were measured. The test was repeated once daily for 5 days. In situ terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay was used for detection of brain damage. As early as 20 min and up to 2 h postinjection, animals treated with L-Cys doses higher than 1.2 mg/g BW developed hypoglycemia and looked ill. Several animals convulsed. Long-term survivors required more time, in a dose-dependent manner, to assimilate the structure of the maze, and animals treated with L-Cys (1.5 mg/g BW) exhibited TUNEL-positive changes in the hippocampal regions. All these changes were reversible by coadministration of glucose. We conclude that L-Cys injection can cause pronounced hypoglycemia associated with long-term neurobehavioral changes and central neural damage in mice. Since L-Cys is chemically different from the other excitatory amino acids (glutamate and aspartate), the long-reported L-Cys-mediated neurotoxicity may be connected to its hypoglycemic effect.


Assuntos
Cisteína/toxicidade , Hipóxia Encefálica/induzido quimicamente , Hipóxia Encefálica/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Animais , Hipóxia Encefálica/psicologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos ICR , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...