Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Prog Histochem Cytochem ; 50(1-2): 1-10, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26026732

RESUMO

Low voltage transmission electron microscopy (LVTEM) was employed to examine biological tissues with accelerating voltages as low as 5kV. Tissue preparation was modified to take advantage of the low-voltage techniques. Treatments with heavy metals, such as post-fixation with osmium tetroxide, on block and counterstaining were omitted. Sections (40nm) were thinner than usual and generated highly contrasted images. General appearance of the cells remains similar to that of conventional TEM. New features were however revealed. The matrix of the pancreatic granules displays heterogeneity with partitions that may correspond to the inner-segregation of their secretory proteins. Mitochondria revealed the presence of the ATP synthase granules along their cristea. The nuclear dense chromatin displayed a honeycomb organization while distinct beads, nucleosomes, aligned along thin threads were seen in the dispersed chromatin. Nuclear pore protein complexes revealed their globular nature. The intercalated disks in cardiac muscle displayed their fine structural organization. These features correlate well with data described or predicted by cell and molecular biology. These new aspects are not revealed when thicker and conventionally osmicated tissue sections were examined by LVTEM, indicating that major masking effects are associated with standard TEM techniques. Immunogold was adapted to LVTEM further enhancing its potential in cell biology.


Assuntos
Microscopia Eletrônica de Transmissão/métodos , Microtomia/métodos , Miocárdio/ultraestrutura , Pâncreas/ultraestrutura , Amilases/análise , Amilases/metabolismo , Animais , Imuno-Histoquímica/métodos , Lamina Tipo A/análise , Lamina Tipo A/metabolismo , Microscopia Eletrônica de Transmissão/instrumentação , Mitocôndrias/ultraestrutura , Miocárdio/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/ultraestrutura , Nucleossomos/ultraestrutura , Pâncreas/metabolismo , Ratos , Ratos Sprague-Dawley , Vesículas Secretórias/ultraestrutura , Inclusão do Tecido/métodos , Fixação de Tecidos/métodos
2.
Microsc Res Tech ; 77(12): 999-1004, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25164611

RESUMO

Low voltage transmission electron microscopy (LVTEM) with accelerating voltages as low as 5 kV was applied to cell biology. To take advantage of the increased contrast given by LVTEM, tissue preparation was modified omitting all heavy metals such as osmium, uranium, and lead from the fixation, on block staining and counterstaining. Nonstained ultra-thin tissue sections (40 nm thick) generated highly contrasted images. While the aspect of the cells remains similar to that obtained by conventional TEM, some new substructures were revealed. The pancreatic acinar cells granules present a heterogeneous matrix with partitions corresponding to segregation of their different secretory proteins. Microvilli display their core of microfilaments anchored to the dense top membrane. Mitochondria revealed the presence of distinct particles along their cristea membranes that may correspond to the ATP synthase complexes or oxysomes. The dense nuclear chromatin displays a honey-comb appearance while distinct beads aligned along thin threads were seen in the dispersed chromatin. These new features revealed by LVTEM correlate with structures described or predicted through other approaches. Masking effects due to thickness of the tissue sections and to the presence of heavy metals must have prevented their observation by conventional TEM. Furthermore, the immunogold was adapted to LVTEM revealing nuclear lamin-A at the edge of the dense chromatin ribbons. Combining cytochemistry with LVTEM brings additional advantages to this new approach in cell biology.


Assuntos
Imuno-Histoquímica , Microscopia Eletrônica de Transmissão/métodos , Células Acinares/ultraestrutura , Biologia Celular , Células Cultivadas , Microtomia , Pâncreas/ultraestrutura
3.
Biochem Cell Biol ; 91(6): 476-86, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24219290

RESUMO

We have recently shown that a high glucose (HG) concentration raised intestinal cholesterol (CHOL) transport and metabolism in intestinal epithelial cells. The objective of the present work is to determine whether the stimulus for increased CHOL absorption by glucose originates from the apical site (corresponding to the intestinal lumen) or from the basolateral site (related to blood circulation). We tackled this issue by using differentiated Caco-2/15 cells. Only basolateral medium, supplemented with 25 mmol/L glucose, stimulated [(14)C]-CHOL uptake via the up-regulation of the critical CHOL transporter NPC1L1 protein, as confirmed by its specific ezetimibe inhibitor that abolished the rise in glucose-mediated CHOL capture. No significant changes were noted in SR-BI and CD36. Elevated CHOL uptake was associated with an increase in the transcription factors SREBP-2, LXR-ß, and ChREBP, along with a fall in RXR-α. Interestingly, although the HG concentration in the apical medium caused modest changes in CHOL processing, its impact was synergetic with that of the basolateral medium. Our results suggest that HG concentration influences positively intestinal CHOL uptake when present in the basolateral medium. In addition, excessive consumption of diets containing high levels of carbohydrates may strengthen intestinal CHOL uptake in metabolic syndrome, thereby contributing to elevated levels of circulating CHOL and, consequently, the risk of developing type 2 diabetes and cardiovascular disease.


Assuntos
Colesterol/metabolismo , Enterócitos/efeitos dos fármacos , Glucose/farmacologia , Proteínas de Membrana/genética , Anticolesterolemiantes/farmacologia , Azetidinas/farmacologia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Transporte Biológico , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células CACO-2 , Radioisótopos de Carbono , Diferenciação Celular , Polaridade Celular , Enterócitos/citologia , Enterócitos/metabolismo , Ezetimiba , Regulação da Expressão Gênica , Glucose/metabolismo , Humanos , Receptores X do Fígado , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Receptores Depuradores Classe B/genética , Receptores Depuradores Classe B/metabolismo , Transdução de Sinais , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo
4.
Autophagy ; 8(6): 927-37, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22692030

RESUMO

The endothelium plays a central role in the regulation of vascular wall cellularity and tone by secreting an array of mediators of importance in intercellular communication. Nutrient deprivation of human endothelial cells (EC) evokes unconventional forms of secretion leading to the release of nanovesicles distinct from apoptotic bodies and bearing markers of multivesicular bodies (MVB). Nutrient deficiency is also a potent inducer of autophagy and vesicular transport pathways can be assisted by autophagy. Nutrient deficiency induced a significant and rapid increase in autophagic features, as imaged by electron microscopy and immunoblotting analysis of LC3-II/LC3-I ratios. Increased autophagic flux was confirmed by exposing serum-starved cells to bafilomycin A 1. Induction of autophagy was followed by indices of an apoptotic response, as assessed by microscopy and poly (ADP-ribose) polymerase cleavage in absence of cell membrane permeabilization indicative of necrosis. Pan-caspase inhibition with ZVAD-FMK did not prevent the development of autophagy but negatively impacted autophagic vacuole (AV) maturation. Adopting a multidimensional proteomics approach with validation by immunoblotting, we determined that nutrient-deprived EC released AV components (LC3I, LC3-II, ATG16L1 and LAMP2) whereas pan-caspase inhibition with ZVAD-FMK blocked AV release. Similarly, nutrient deprivation in aortic murine EC isolated from CASP3/caspase 3-deficient mice induced an autophagic response in absence of apoptosis and failed to prompt LC3 release. Collectively, the present results demonstrate the release of autophagic components by nutrient-deprived apoptotic human cells in absence of cell membrane permeabilization. These results also identify caspase-3 as a novel regulator of AV release.


Assuntos
Autofagia , Caspases/metabolismo , Espaço Extracelular/metabolismo , Vacúolos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Caspase 3/deficiência , Caspase 3/metabolismo , Inibidores de Caspase/farmacologia , Cromatografia Líquida , Meios de Cultura Livres de Soro , Ativação Enzimática/efeitos dos fármacos , Espaço Extracelular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Humanos , Espectrometria de Massas , Camundongos , Vacúolos/efeitos dos fármacos , Vacúolos/ultraestrutura
5.
Anat Cell Biol ; 45(1): 1-16, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22536547

RESUMO

A major advance in the understanding of the regulation of food intake has been the discovery of the adipokine leptin a hormone secreted by the adipose tissue. After crossing the blood-brain barrier, leptin reaches its main site of action at the level of the hypothalamic cells where it plays fundamental roles in the control of appetite and in the regulation of energy expenditure. At first considered as a hormone specific to the white adipose tissue, it was rapidly found to be expressed by other tissues. Among these, the gastric mucosa has been demonstrated to secrete large amounts of leptin. Secretion of leptin by the gastric chief cells was found to be an exocrine secretion. Leptin is secreted towards the gastric lumen into the gastric juice. We found that while secretion of leptin by the white adipose tissue is constitutive, secretion by the gastric cells is a regulated one responding very rapidly to secretory stimuli such as food intake. Exocrine-secreted leptin survives the hydrolytic conditions of the gastric juice by forming a complex with its soluble receptor. This soluble receptor is synthesized by the gastric cells and the leptin-leptin receptor complex gets formed at the level of the gastric chief cell secretory granules before being released into the gastric lumen. The leptin-leptin receptor upon resisting the hydrolytic conditions of the gastric juice is channelled, to the duodenum. Transmembrane leptin receptors expressed at the luminal membrane of the duodenal enterocytes interact with the luminal leptin. Leptin is actively transcytosed by the duodenal enterocytes. From the apical membrane it is transferred to the Golgi apparatus where it binds again its soluble receptor. The newly formed leptin-leptin receptor complex is then secreted baso-laterally into the intestinal mucosa to reach the blood capillaries and circulation thus reaching the hypothalamus where its action regulates food intake. Exocrine-secreted gastric leptin participates in the short term regulation of food intake independently from that secreted by the adipose tissue. Adipose tissue leptin on the other hand, regulates in the long term energy storage. Both tissues work in tandem to ensure management of food intake and energy expenditure.

6.
Artigo em Inglês | MEDLINE | ID: mdl-22214336

RESUMO

BACKGROUND: Leptin receptors (LEPR) are expressed in intestinal epithelial cells from the duodenum to the colon. Since their role is fundamental for the proper control of nutrient absorption, mucus secretion and mucosa renewal, the regulation of LEPR expression is for the first time investigated as a function of various potential effectors. METHODOLOGY/PRINCIPAL FINDINGS: Fully differentiated Caco-2/15 cells were incubated for 24 hours with nutrients [carbohydrates, fatty acids (FA), amino acids and sterols], hormones (leptin, insulin, hydrocortisone and epithelial growth factor), inflammatory agents (Interferon-γ, LPS, TNF-α), and PPAR agonists (rosiglitazone and WY14643). Levels of LEPR mRNA and protein expressions were measured by RT-PCR and Western blots, respectively. RESULTS: Long (219.1) and short (219.3) isoforms of the LEPR were detected in Caco-2/15 cells, while absence of the isoform 219.2 was noted. Their gene expression was modulated by carbohydrates, FA, PPAR agonists, biliary salts, insulin and leptin itself. On the other hand, LEPR protein expression was modulated by FA, cholesterol, biliary salts, PPAR agonists and insulin. Interestingly, the same effectors may have opposite effects on the short and the long LEPR isoforms, as well as on mRNA and protein levels. Finally, Caco-2/15 cells were found to be sensitive to the effector location, i.e. apical or basolateral compartment. CONCLUSIONS/SIGNIFICANCE: Our results suggest that (i) the expression of LEPR in Caco-2/15 cell line is not constitutive; (ii) the agents present in the apical or basolateral medium have different effects on LEPR mRNA and/or protein levels; and (iii) short and long isoforms of LEPR follow different patterns of regulation.


Assuntos
Polaridade Celular/genética , Regulação Neoplásica da Expressão Gênica , Receptores para Leptina/genética , Aminoácidos/farmacologia , Células CACO-2 , Carboidratos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Polaridade Celular/efeitos dos fármacos , Colesterol/farmacologia , Ácidos Graxos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Isoformas de Proteínas , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores para Leptina/metabolismo
7.
Anat Cell Biol ; 44(3): 176-85, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22025969

RESUMO

The Psammomys obesus lives in natural desert habitat on low energy (LE) diet, however when maintained in laboratory conditions with high energy (HE) diet it exhibits pathological metabolic changes resembling those of type 2 diabetes. We have evaluated and correlated the histopathology, metabolic and functional renal alterations occurring in the diabetic Psammomys. Renal function determined by measuring glomerular filtration rate (GFR), protein excretion, protein/creatinine ratio and morpho-immunocytochemical evaluations were performed on HE diet diabetic animals and compared to LE diet control animals. The diabetic animals present a 54% increase in GFR after one month of hyperglycemic condition and a decrease of 47% from baseline values after 4 months. Protein excretion in diabetic animals was 5 folds increased after 4 months. Light microscopy showed an increase in glomeruli size in the diabetic Psammomys, and electron microscopy and immunocytochemical quantitative evaluations revealed accumulation of basement membrane material as well as frequent splitting of the glomerular basement membrane. In addition, glycogen-filled Armanni-Ebstein clear cells were found in the distal tubules including the thick ascending limbs of the diabetic animals. These renal complications in the Psammomys, including changes in GFR with massive proteinuria sustained by physiological and histopathological changes, are very similar to the diabetic nephropathy in human. The Psamommys obesus represents therefore a reliable animal model of diabetic nephropathy.

8.
J Histochem Cytochem ; 59(10): 899-907, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21832147

RESUMO

Low-voltage (5-kV) transmission electron microscopy revealed a novel aspect of the pancreatic acinar cell secretory granules not previously detected by conventional (80-kV) transmission electron microscopy. Examination of ultra-thin (30-nm) sections of non-osmicated, stain-free pancreatic tissue sections by low-voltage electron microscopy revealed the existence of granules with non-homogeneous matrix and sub-compartments having circular or oval profiles of different electron densities and sizes. Such partition is completely masked when observing tissues after postfixation with osmium tetroxide by low-voltage transmission electron microscopy at 5 kV and/or when thicker sections (70 nm) are examined at 80 kV. This morphological partition reflects an internal compartmentalization of the granule content that was previously predicted by morphological, physiological, and biochemical means. It corresponds to the segregation of the different secretory proteins inside the granule as demonstrated by high-resolution immunocytochemistry and reflects a well-organized aggregation of the secretory proteins at the time of granule formation in the trans-Golgi. Such partition of the granule matrix undergoes changes under experimental conditions known to alter the secretory process such as stimulation of secretion or diabetes.


Assuntos
Microscopia Eletrônica de Transmissão/métodos , Pâncreas/ultraestrutura , Vesículas Secretórias/ultraestrutura , Animais , Microscopia Eletrônica de Transmissão/instrumentação , Tetróxido de Ósmio/química , Pâncreas/enzimologia , Ratos , Ratos Sprague-Dawley
9.
Prog Histochem Cytochem ; 45(3): 143-200, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20621336

RESUMO

The understanding of the regulation of food intake has become increasingly complex. More than 20 hormones, both orexigenic and anorexigenic, have been identified. After crossing the blood-brain barrier, they reach their main site of action located in several hypothalamic areas and interact to balance satiety and hunger. One of the most significant advances in this matter has been the discovery of leptin. This hormone plays fundamental roles in the control of appetite and in regulating energy expenditure. In accordance with the lipostatic theory stated by Kennedy in 1953, leptin was originally discovered in white adipose tissue. Its expression by other tissues was later established. Among them, the gastric mucosa has been shown to secrete large amounts of leptin. Both the adipose and the gastric tissues share similar characteristics in the synthesis and storage of leptin in granules, in the formation of a complex with the soluble receptor and a secretion modulated by hormones and energy substrates. However while adipose tissue secretes leptin in a slow constitutive endocrine way, the gastric mucosa releases leptin in a rapid regulated exocrine fashion into the gastric juice. Exocrine-secreted leptin survives the extreme hydrolytic conditions of the gastric juice and reach the duodenal lumen in an intact active form. Scrutiny into transport mechanisms revealed that a significant amount of the exocrine leptin crosses the intestinal wall by active transcytosis. Leptin receptors, expressed on the luminal and basal membrane of intestinal epithelial cells, are involved in the control of nutrient absorption by enterocytes, mucus secretion by goblet cells and motility, among other processes, and this control is indeed different depending upon luminal or basal stimulus. Gastric leptin after transcytosis reaches the central nervous system, to control food intake. Studies using the Caco-2, the human intestinal cell line, in vitro allowed analysis of the mechanisms of leptin actions on the intestinal mucosa, identification of the mechanisms of leptin transcytosis and understanding the modulation of leptin receptors by nutrients and hormones. Exocrine-secreted gastric leptin thus participates in a physiological axis independent in terms of time and regulation from that of adipose tissue to rapidly control food intake and nutrient absorption. Adipocytes and gastric epithelial cells are two cell types the metabolism of which is closely linked to food intake and energy storage. The coordinated secretion of adipose and gastric leptins ensures proper management of food processing and energy storage.


Assuntos
Tecido Adiposo/metabolismo , Regulação do Apetite , Metabolismo Energético/fisiologia , Mucosa Gástrica/metabolismo , Leptina/fisiologia , Ingestão de Alimentos , Humanos , Hipotálamo/fisiologia , Leptina/metabolismo , Obesidade/metabolismo , Receptores para Leptina/fisiologia
10.
Int J Cell Biol ; 2010: 928169, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20454702

RESUMO

Gastric Leptin is absorbed by duodenal enterocytes and released on the basolateral side towards the bloodstream. We investigated in vitro some of the mechanisms of this transport. Caco-2/15 cells internalize leptin from the apical medium and release it through transcytosis in the basal medium in a time- temperature-dependent and saturable fashion. Leptin receptors are revealed on the apical brush-border membrane of the Caco-2 cells. RNA-mediated silencing of the receptor led to decreases in the uptake and basolateral release. Leptin in the basal medium was found bound to the soluble form of its receptor. An inhibitor of clathrin-dependent endocytosis (chlorpromazine) decreased leptin uptake. Confocal immunocytochemistry and the use of brefeldin A and okadaic acid revealed the passage of leptin through the Golgi apparatus. We propose that leptin transcytosis by intestinal cells depends on its receptor, on clathrin-coated vesicles and transits through the Golgi apparatus.

11.
J Histochem Cytochem ; 58(6): 567-76, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20197491

RESUMO

Nestin, which was initially identified as a marker of neural stem cells, has been reported in regenerating pancreas as well as in early embryonic stem (ES) cell derivatives. However, little is known about its specific roles in stem cells as a functional regulator. We investigated the source of the action of nestin in ES and adult pancreatic ductal stem (PDS) cells in regard to the neogenesis of insulin-secreting beta-cells. In ES cells, suppression of nestin by gene silencing led to an increased expression of the pluripotency-associated genes, including Oct 4, Nanog, and SSEA-1, before embryoid body (EB) formation, whereas it reduced endodermal and pancreatic transcription factors in EBs. Inhibition of nestin expression in adult PDS cells caused a low expression of pancreatic transcription factors and islet hormones, leading to poor beta-cell development and insulin secretion. These data may indicate not only that nestin is a simple stem cell marker, but also that it constitutes a functional factor at the time of stem cell differentiation. We suggest that nestin plays pivotal roles as an intermediate regulator governing both stemness and differentiation of stem cells in the process of their differentiation into insulin-secreting cells.


Assuntos
Diferenciação Celular/fisiologia , Células Secretoras de Insulina/citologia , Proteínas de Filamentos Intermediários/genética , Proteínas do Tecido Nervoso/genética , Ductos Pancreáticos/citologia , Animais , Primers do DNA , Regulação da Expressão Gênica , Gliceraldeído 3-Fosfato Desidrogenase (NADP+)/genética , Células Secretoras de Insulina/metabolismo , Proteínas de Filamentos Intermediários/fisiologia , Masculino , Camundongos , Proteínas do Tecido Nervoso/fisiologia , Nestina , Ductos Pancreáticos/metabolismo , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Células-Tronco/citologia , Células-Tronco/fisiologia
12.
J Histochem Cytochem ; 57(10): 963-71, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19581628

RESUMO

Immunogold cytochemistry was applied to reveal the intracellular location of AMP-activated protein kinase (AMPK) subunits in liver tissue of normal rats fed ad libitum. AMPK alpha and beta subunits were located both in the cytosol and in close association with rosettes of glycogen particles (alpha particles). To reveal their true in situ association with glycogen, particular tissue processing conditions that retain glycogen in the cells were required. These included fixation with a combination of glutaraldehyde and paraformaldehyde, followed by postfixation with osmium tetroxide and lead citrate and embedding in Epon. Processing by less-stringent fixation conditions and embedding in Lowicryl led to the extraction of the glycogen deposits, which in turn resulted in the absence of any labeling. This indicates that the loss of glycogen deposits leads to the loss of closely associated proteins. Labeling for the alpha(1) and alpha(2) subunits of AMPK was found to be about 2-fold greater over glycogen than over cytosol, whereas labeling for beta(1) was 8-fold higher over the glycogen particles than over the cytosol. Immunogold combined with morphometric analysis demonstrated that the beta(1) subunits are located at the periphery of the glycogen rosettes, consistent with a recent hypothesis developed via biochemical approaches.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Glicogênio Hepático/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/imunologia , Animais , Especificidade de Anticorpos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Microscopia Imunoeletrônica , Subunidades Proteicas/genética , Subunidades Proteicas/imunologia , Subunidades Proteicas/metabolismo , Ratos , Ratos Sprague-Dawley
13.
Histochem Cell Biol ; 132(3): 351-67, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19499240

RESUMO

Although intestinal (I) and liver (L) fatty acid binding proteins (FABP) have been widely studied, the physiological significance of the presence of the two FABP forms (I- and L-FABP) in absorptive cells remains unknown as do the differences related to their distribution along the crypt-villus axis, regional expression, ontogeny and regulation in the human intestine. Our morphological experiments supported the expression of I- and L-FABP as early as 13 weeks of gestation. Whereas cytoplasmic immunofluorescence staining of L-FABP was barely detectable in the lower half of the villus and in the crypt epithelial cells, I-FABP was visualized in epithelial cells of the crypt-villus axis in all intestinal segments until the adult period in which the staining was maximized in the upper part of the villus. Immunoelectron microscopy revealed more intense labeling of L-FABP compared with I-FABP, accompanied with a heterogeneous distribution in the cytoplasm, microvilli and basolateral membranes. By western blot analysis, I- and L-FABP at 15 weeks of gestation appeared predominant in jejunum compared with duodenum, ileum, proximal and distal colon. Exploration of the maturation aspect documented a rise in L-FABP in adult tissues. Permanent transfections of Caco-2 cells with I-FABP cDNA resulted in decreased lipid export, apolipoprotein (apo) biogenesis and chylomicron secretion. Additionally, supplementation of Caco-2 with insulin, hydrocortisone and epidermal growth factor differentially modulated the expression of I- and L-FABP, apo B-48 and microsomal triglyceride transfer protein (MTP), emphasizing that these key proteins do not exhibit a parallel modulation. Overall, our findings indicate that the two FABPs display differences in localization, regulation and developmental pattern.


Assuntos
Colo/metabolismo , Proteínas de Ligação a Ácido Graxo/metabolismo , Jejuno/metabolismo , Lipoproteínas/metabolismo , Células CACO-2 , Colo/embriologia , Colo/crescimento & desenvolvimento , Humanos , Lactente , Jejuno/embriologia , Jejuno/crescimento & desenvolvimento , Especificidade de Órgãos
14.
J Neurosci Res ; 87(4): 1023-36, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18855943

RESUMO

A major concern regarding the chronic administration of antiretroviral drugs is the potential for induction of drug efflux transporter expression (i.e., P-glycoprotein, P-gp) at tissue sites that can significantly affect drug distribution and treatment efficacy. Previous data have shown that the inductive effect of human immunodeficiency virus protease inhibitors (PIs) is mediated through the human orphan nuclear receptor, steroid xenobiotic receptor (SXR or hPXR). The objectives of this study were to investigate transport and inductive properties on efflux drug transporters of two PIs, atazanavir and ritonavir, at the blood-brain barrier by using a human brain microvessel endothelial cell line, hCMEC/D3. Transport properties of PIs by the drug efflux transporters P-gp and multidrug resistance protein 1 (MRP1) were assessed by measuring the cellular uptake of (3)H-atazanavir or (3)H-ritonavir in P-gp and MRP1 overexpressing cells as well as hCMEC/D3. Whereas the P-gp inhibitor, PSC833, increased atazanavir and ritonavir accumulation in hCMEC/D3 cells by 2-fold, the MRP inhibitor MK571 had no effect. P-gp, MRP1, and hPXR expression and localization were examined by Western blot analysis and immunogold cytochemistry at the electron microscope level. Treatment of hCMEC/D3 cells for 72 hr with rifampin or SR12813 (two well-established hPXR ligands) or PIs (atazanavir or ritonavir) resulted in an increase in P-gp expression by 1.8-, 6-, and 2-fold, respectively, with no effect observed for MRP1 expression. In hCMEC/D3 cells, cellular accumulation of these PIs appears to be primarily limited by P-gp efflux activity. Long-term exposure of atazanavir or ritonavir to brain microvessel endothelium may result in further limitations in brain drug permeability as a result of the up-regulation of P-gp expression and function.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Endotélio Vascular/metabolismo , Inibidores da Protease de HIV/farmacologia , Regulação para Cima , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Sulfato de Atazanavir , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Ciclosporinas/farmacologia , Difosfonatos/farmacologia , Endotélio Vascular/efeitos dos fármacos , Humanos , Microvasos/efeitos dos fármacos , Microvasos/metabolismo , Oligopeptídeos/farmacologia , Propionatos/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Rifampina/farmacologia , Ritonavir/farmacologia , Trítio/metabolismo
15.
J Mol Histol ; 39(6): 579-84, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18941912

RESUMO

Adiponectin receptor ADIPOR1 activates the intracellular second messenger AMP-activated protein kinase (AMPK) that participates in the control of the oxidative stress and apoptosis. This study reveals the presence of a functional ADIPOR1 receptor in all the cells of the renal glomeruli. Isolated glomeruli were incubated in vitro with adiponectin and proteins analysed by western blot. Electron microscopy using immunogold labeling was carried out on kidney sections. ADIPOR1 and catalytic AMPK sub-units alpha1 and alpha2 were revealed in normal rat glomeruli and incubation of freshly isolated rat glomeruli with either adiponectin or AICAR led to the activation by phosphorylation of catalytic AMPK. Electron microscopy localized with high resolution these proteins at the plasma membrane of the three glomerular cells, namely the endothelial, the mesangial and the podocyte cells, as well as on Bowman's capsule epithelial cells. It is concluded that glomerular cells express a functional adiponectin receptor ADIPOR1 which, through activation of AMPK, may play important roles in the control of oxidative stress and cell survival within the glomerulus.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adiponectina/metabolismo , Glomérulos Renais/enzimologia , Receptores de Adiponectina/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Animais , Humanos , Imuno-Histoquímica , Glomérulos Renais/ultraestrutura , Masculino , Fosforilação , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Ribonucleotídeos/metabolismo
16.
Microsc Res Tech ; 71(9): 659-62, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18512738

RESUMO

Novel approach in low voltage transmission electron microscopy (TEM) has revealed the presence of SV40 viral like particles in the secretory zymogen granules and in spherical membrane-bound dense bodies of SV40 infected pancreatic cells. The presence of SV40 antigen in these cellular compartments was confirmed by immunocytochemistry of the VP1 antigen. Visualization of the viral particles was only possible by examining ultrathin tissue sections with low-voltage TEM that significantly enhances imaging contrast. Results indicate that following infection of the cell entry and trafficking of the viral particles are present in unique cellular compartments such as ER, dense bodies, and secretory granules.


Assuntos
Grânulos Citoplasmáticos/virologia , Microscopia Eletrônica de Transmissão/métodos , Pâncreas/citologia , Vírus 40 dos Símios/ultraestrutura , Animais , Compartimento Celular , Grânulos Citoplasmáticos/metabolismo , Pâncreas/ultraestrutura
17.
Pancreas ; 36(4): 411-6, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18437088

RESUMO

OBJECTIVES: Viral vector uptake into the pancreas is rare. The few viral vectors reported to transduce in vivo pancreatic islets after systemic injection required additional physical measures, such as direct pancreatic injection or hepatic vessel clamping. Because pancreatic islet uptake of the human polyomavirus family member BK virus was previously reported in hamsters after systemic administration, we hypothesized that SV40, a polyomavirus member remarkably similar to BK virus, may also infect the pancreas. METHODS: We injected intravenously a low dose of SV40, unaided by any other physical or chemical means, and evaluated viral uptake by pancreatic islets and pancreatic exocrine tissue via polymerase chain reaction, Western blot, electron microscopy, immunofluorescent microscopy, and protein A-gold immunocytochemistry. RESULTS: Pancreatic uptake of SV40 was comparable to other major organs (ie, liver and spleen). SV40 viral particles were detected in both pancreatic islets and acini. In pancreatic islets, all islet cell types were infected by SV40, albeit the infection rate of glucagon-producing alpha cells surpassed beta- and delta-islet cells. Low-dose SV40 administration was not sufficient to induce heterologous gene expression in the pancreas. CONCLUSIONS: Our study shows that pancreatic islet and acinar cell uptake of SV40 is feasible with a single, low-dose intravenous injection. However, this dose did not result in gene delivery into the murine pancreas.


Assuntos
Ilhotas Pancreáticas/virologia , Pancreatopatias/virologia , Vírus 40 dos Símios/patogenicidade , Animais , Diabetes Mellitus Tipo 1/virologia , Feminino , Regulação da Expressão Gênica , Ilhotas Pancreáticas/ultraestrutura , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Pancreatopatias/patologia , Reação em Cadeia da Polimerase , Vírus 40 dos Símios/isolamento & purificação , Vírus 40 dos Símios/ultraestrutura
18.
J Histochem Cytochem ; 56(6): 605-14, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18319274

RESUMO

Aging and diabetes are associated with exacerbated expression of adhesion molecules. Given their importance in endothelial dysfunction and their possible involvement in the alteration of glomerular permeability occurring in diabetes, we have evaluated expression of the sialomucin-type adhesion molecule CD34 in renal glomerular cells of normal and diabetic animals at two different ages by colloidal gold immunocytochemistry and immunoblotting. CD34 labeling was mostly assigned to the plasma membranes of glomerular endothelium and mesangial processes. Podocyte membranes were also labeled, but to a lesser degree. Short- and long-term diabetes triggers a substantial increase in immunogold labeling for CD34 in renal tissues compared with young normoglycemic animals. However, the level of labeling in old diabetic and healthy control rats is similar, suggesting that the effect of diabetes and aging on CD34 expression is similar but not synergistic. Western blotting of isolated glomerular fractions corroborated immunocytochemical results. Increased expression of CD34 may reflect its involvement in the pathogenesis of glomerular alterations related to age and diabetes. Alterations present in early diabetes, resembling those occurring with age, strengthen the concept that diabetes is an accelerated form of aging.


Assuntos
Antígenos CD34/biossíntese , Diabetes Mellitus Experimental/metabolismo , Glomérulos Renais/metabolismo , Envelhecimento , Sequência de Aminoácidos , Animais , Western Blotting , Hiperglicemia/metabolismo , Imuno-Histoquímica , Glomérulos Renais/irrigação sanguínea , Masculino , Dados de Sequência Molecular , Ratos , Ratos Sprague-Dawley
19.
Am J Physiol Renal Physiol ; 294(4): F881-9, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18256313

RESUMO

Diabetic nephropathies are characterized by glycogen accumulation in distal tubular cells, which eventually leads to their apoptosis. The present study aims to determine whether adiponectin and AMPK are involved in the regulation of glycogen synthase (GS) in these structures. Western blots of isolated distal tubules revealed the presence of adiponectin receptor ADIPOR1, catalytic AMPK subunits alpha(1) and alpha(2), their phosphorylated active forms, and the glycogen-binding AMPK subunit beta(2). ADIPOR2 was not detected. Expression levels of ADIPOR1, AMPKalpha(1), AMPKalpha(2), and AMPKbeta(2) were increased in streptozotocin-treated diabetic rats, whereas phosphorylated active AMPK levels were strongly decreased. Immunohistochemistry revealed the presence of ADIPOR1 on the luminal portion of distal tubules and thick ascending limb cells. Catalytic subunits alpha(1) and alpha(2), their phosphorylated active forms, and the glycogen-binding subunit beta(2) were also found in the same cells, confirming immunoblot results. In vitro, 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR; 2 mM) and globular adiponectin (10 mug/ml) activated catalytic AMPK in distal tubules isolated from kidneys of normal rats but much more weakly in those from diabetic rats. GS inhibition paralleled AMPK activation in both groups of animals: active GS levels were low in control animals and elevated in diabetic ones. Finally, glucose-6-phosphate, an allosteric activator of GS, was also increased in diabetic rats. These results demonstrate that in distal tubular cells, adiponectin through luminal ADIPOR1 activates AMPK, leading to the inhibition of GS. During hyperglycemia, this regulation is altered, which may explain, at least in part, the accumulation of large glycogen deposits.


Assuntos
Adenilato Quinase/metabolismo , Adiponectina/farmacologia , Aminoimidazol Carboxamida/análogos & derivados , Diabetes Mellitus Experimental/fisiopatologia , Glicogênio Sintase/metabolismo , Túbulos Renais Distais/fisiopatologia , Receptores de Adiponectina/metabolismo , Ribonucleotídeos/farmacologia , Adenilato Quinase/efeitos dos fármacos , Aminoimidazol Carboxamida/farmacologia , Animais , Diabetes Mellitus Experimental/enzimologia , Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase/efeitos dos fármacos , Imuno-Histoquímica , Rim/efeitos dos fármacos , Rim/patologia , Túbulos Renais Distais/efeitos dos fármacos , Túbulos Renais Distais/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Adiponectina/efeitos dos fármacos
20.
Nat Med ; 14(1): 55-63, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18084301

RESUMO

Podocyte dysfunction, represented by foot process effacement and proteinuria, is often the starting point for progressive kidney disease. Therapies aimed at the cellular level of the disease are currently not available. Here we show that induction of urokinase receptor (uPAR) signaling in podocytes leads to foot process effacement and urinary protein loss via a mechanism that includes lipid-dependent activation of alphavbeta3 integrin. Mice lacking uPAR (Plaur-/-) are protected from lipopolysaccharide (LPS)-mediated proteinuria but develop disease after expression of a constitutively active beta3 integrin. Gene transfer studies reveal a prerequisite for uPAR expression in podocytes, but not in endothelial cells, for the development of LPS-mediated proteinuria. Mechanistically, uPAR is required to activate alphavbeta3 integrin in podocytes, promoting cell motility and activation of the small GTPases Cdc42 and Rac1. Blockade of alphavbeta3 integrin reduces podocyte motility in vitro and lowers proteinuria in mice. Our findings show a physiological role for uPAR signaling in the regulation of kidney permeability.


Assuntos
Regulação da Expressão Gênica , Rim/metabolismo , Podócitos/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Movimento Celular , Técnicas de Transferência de Genes , Humanos , Integrina alfaVbeta3/metabolismo , Rim/patologia , Lipopolissacarídeos/metabolismo , Microdomínios da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...