Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 110(49): E4733-42, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248383

ABSTRACT

Melanocortin-4 receptor (MC4R) is a G protein-coupled receptor expressed in neurons of the hypothalamus where it regulates food intake. MC4R responds to an agonist, α-melanocyte-stimulating hormone (α-MSH) and to an antagonist/inverse agonist, agouti-related peptide (AgRP), which are released by upstream neurons. Binding to α-MSH leads to stimulation of receptor activity and suppression of food intake, whereas AgRP has opposite effects. MC4R cycles constantly between the plasma membrane and endosomes and undergoes agonist-mediated desensitization by being routed to lysosomes. MC4R desensitization and increased AgRP expression are thought to decrease the effectiveness of MC4R agonists as an antiobesity treatment. In this study, α-MSH, instead of being delivered extracellularly, is targeted to the endoplasmic reticulum (ER) of neuronal cells and cultured hypothalamic neurons. We find that the ER-targeted agonist associates with MC4R at this location, is transported to the cell surface, induces constant cAMP and AMP kinase signaling at maximal amplitude, abolishes desensitization of the receptor, and promotes both cell-surface expression and constant signaling by an obesity-linked MC4R variant, I316S, that otherwise is retained in the ER. Formation of the MC4R/agonist complex in the ER stabilizes the receptor in an active conformation that at the cell surface is insensitive to antagonism by AgRP and at the endosomes is refractory to routing to the lysosomes. The data indicate that targeting agonists to the ER can stabilize an active conformation of a G protein-coupled receptor that does not become desensitized, suggesting a target for therapy.


Subject(s)
Appetite Regulation/physiology , Endoplasmic Reticulum/metabolism , Hypothalamus/cytology , Neurons/metabolism , Protein Conformation , Receptor, Melanocortin, Type 4/chemistry , alpha-MSH/pharmacology , Agouti-Related Protein/pharmacology , Analysis of Variance , Animals , Appetite Regulation/genetics , Cyclic AMP/metabolism , Immunoenzyme Techniques , Lysosomes/metabolism , Mice , Microscopy, Fluorescence , Mutation, Missense/genetics , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/genetics
2.
J Biol Chem ; 288(4): 2157-66, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23233669

ABSTRACT

A spectrin-based cytoskeleton is associated with endomembranes, including the Golgi complex and cytoplasmic vesicles, but its role remains poorly understood. Using new generated antibodies to specific peptide sequences of the human ßIII spectrin, we here show its distribution in the Golgi complex, where it is enriched in the trans-Golgi and trans-Golgi network. The use of a drug-inducible enzymatic assay that depletes the Golgi-associated pool of PI4P as well as the expression of PH domains of Golgi proteins that specifically recognize this phosphoinositide both displaced ßIII spectrin from the Golgi. However, the interference with actin dynamics using actin toxins did not affect the localization of ßIII spectrin to Golgi membranes. Depletion of ßIII spectrin using siRNA technology and the microinjection of anti-ßIII spectrin antibodies into the cytoplasm lead to the fragmentation of the Golgi. At ultrastructural level, Golgi fragments showed swollen distal Golgi cisternae and vesicular structures. Using a variety of protein transport assays, we show that the endoplasmic reticulum-to-Golgi and post-Golgi protein transports were impaired in ßIII spectrin-depleted cells. However, the internalization of the Shiga toxin subunit B to the endoplasmic reticulum was unaffected. We state that ßIII spectrin constitutes a major skeletal component of distal Golgi compartments, where it is necessary to maintain its structural integrity and secretory activity, and unlike actin, PI4P appears to be highly relevant for the association of ßIII spectrin the Golgi complex.


Subject(s)
Golgi Apparatus/metabolism , Spectrin/genetics , Spectrin/physiology , Animals , Biological Transport , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Cytoskeleton/metabolism , Epithelial Cells/cytology , HeLa Cells , Humans , Protein Transport , RNA, Small Interfering/metabolism , Rats , Subcellular Fractions/metabolism
3.
Physiol Rep ; 1(6): e00125, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24400134

ABSTRACT

Liver myofibroblasts derived from hepatic stellate cells (HSC) are critical mediators of liver fibrosis. Release of tissue inhibitor of metalloproteinase-1 (TIMP-1) advances liver fibrosis by blocking fibrinolysis. The mechanisms responsible for the posttranslational regulation of TIMP-1 by myofibroblastic HSC are unknown. Here, we demonstrate that TIMP-1 release by HSC is regulated in a posttranslational fashion via calcium-sensitive vesicular exocytosis. To our knowledge, this is the first article to directly examine vesicular trafficking in myofibroblastic HSC, potentially providing a new target to treat and or prevent liver fibrosis.

4.
PLoS One ; 7(12): e50894, 2012.
Article in English | MEDLINE | ID: mdl-23251400

ABSTRACT

Heterozygous mutations in the melanocortin-4 receptor (MC4R) gene represent the most frequent cause of monogenic obesity in humans. MC4R mutation analysis in a cohort of 77 children with morbid obesity identified previously unreported heterozygous mutations (P272L, N74I) in two patients inherited from their obese mothers. A rare polymorphism (I251L, allelic frequency: 1/100) reported to protect against obesity was found in another obese patient. When expressed in neuronal cells, the cell surface abundance of wild-type MC4R and of the N74I and I251L variants and the cAMP generated by these receptors in response to exposure to the agonist, α-MSH, were not different. Conversely, MC4R P272L was retained in the endoplasmic reticulum and had reduced cell surface expression and signaling (by ≈ 3-fold). The chemical chaperone PBA, which promotes protein folding of wild-type MC4R, had minimal effects on the distribution and signaling of the P272L variant. In contrast, incubation with UBE-41, a specific inhibitor of ubiquitin activating enzyme E1, inhibited ubiquitination of MC4R P272L and increased its cell surface expression and signaling to similar levels as wild-type MC4R. UBE41 had much less profound effects on MC4R I316S, another obesity-linked MC4R variant trapped in the ER. These data suggest that P272L is retained in the ER by a propensity to be ubiquitinated in the face of correct folding, which is only minimally shared by MC4R I316S. Thus, studies that combine clinical screening of obese patients and investigation of the functional defects of the obesity-linked MC4R variants can identify specific ways to correct these defects and are the first steps towards personalized medicine.


Subject(s)
Endoplasmic Reticulum/genetics , Obesity/genetics , Receptor, Melanocortin, Type 4/genetics , Ubiquitination/genetics , Alleles , Cell Line , Child , Child, Preschool , Cyclic AMP/metabolism , DNA Mutational Analysis , Endoplasmic Reticulum/metabolism , Female , Gene Frequency , Humans , Male , Mutation , Obesity/metabolism , Protein Folding , Receptor, Melanocortin, Type 4/metabolism , Transfection , Ubiquitin/genetics , Ubiquitin/metabolism
5.
J Biol Chem ; 287(26): 21873-90, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22544740

ABSTRACT

Melanocortin-4 receptor (MC4R) is a G-protein-coupled receptor expressed in the hypothalamus where it controls feeding behavior. MC4R cycles constitutively and is internalized at the same rate in the presence or absence of stimulation by the agonist, melanocyte-stimulating hormone (α-MSH). This is different from other G-protein-coupled receptors, such as ß(2)-adrenergic receptor (ß(2)AR), which internalizes more rapidly in response to agonist stimulation. Here, it is found that in immortalized neuronal Neuro2A cells expressing exogenous receptors, constitutive endocytosis of MC4R and agonist-dependent internalization of ß(2)AR were equally sensitive to clathrin depletion. Inhibition of MC4R endocytosis by clathrin depletion decreased the number of receptors at the cell surface that were responsive to the agonist, α-MSH, by 75%. Mild membrane cholesterol depletion also inhibited constitutive endocytosis of MC4R by ∼5-fold, while not affecting recycling of MC4R or agonist-dependent internalization of ß(2)AR. Reduced cholesterol did not change the MC4R dose-response curve to α-MSH, but it decreased the amount of cAMP generated per receptor number indicating that a population of MC4R at the cell surface becomes nonfunctional. The loss of MC4R function increased over time (25-50%) and was partially reversed by mutations at putative phosphorylation sites (T312A and S329A). This was reproduced in hypothalamic GT1-7 cells expressing endogenous MC4R. The data indicate that constitutive endocytosis of MC4R is clathrin- and cholesterol-dependent. MC4R endocytosis is required to maintain MC4R responsiveness to α-MSH by constantly eliminating from the plasma membrane a pool of receptors modified at Thr-312 and Ser-329 that have to be cycled to the endosomal compartment to regain function.


Subject(s)
Cholesterol/chemistry , Receptor, Melanocortin, Type 4/chemistry , alpha-MSH/metabolism , Cell Membrane/metabolism , Cholesterol/metabolism , Clathrin/chemistry , Clathrin/metabolism , Dose-Response Relationship, Drug , Endocytosis , Endosomes/metabolism , HEK293 Cells , Homeostasis , Humans , Mutation , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Serine/chemistry , Signal Transduction , Threonine/chemistry
6.
J Biol Chem ; 286(48): 41563-41577, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21976666

ABSTRACT

α(1)-Antitrypsin is a serine protease inhibitor secreted by hepatocytes. A variant of α(1)-antitrypsin with an E342K (Z) mutation (ATZ) has propensity to form polymers, is retained in the endoplasmic reticulum (ER), is degraded by both ER-associated degradation and autophagy, and causes hepatocyte loss. Constant features in hepatocytes of PiZZ individuals and in PiZ transgenic mice expressing ATZ are the formation of membrane-limited globular inclusions containing ATZ and mitochondrial damage. Expression of ATZ in the liver does not induce the unfolded protein response (UPR), a protective mechanism aimed to maintain ER homeostasis in the face of an increased load of proteins. Here we found that in hepatoma cells the ER E3 ligase HRD1 functioned to degrade most of the ATZ before globular inclusions are formed. Activation of the activating transcription factor 6 (ATF6) branch of the UPR by expression of spliced ATF6(1-373) decreased intracellular accumulation of ATZ and the formation of globular inclusions by a pathway that required HRD1 and the proteasome. Expression of ATF6(1-373) in ATZ-expressing hepatoma cells did not induce autophagy and increased the level of the proapoptotic factor CCAAT/enhancer-binding protein (C/EBP) homologous protein (CHOP) but did not lead to apoptotic DNA fragmentation. Expression of ATF6(1-373) did not cause inhibition of protein synthesis and prevented mitochondrial damage induced by ATZ expression. It was concluded that activation of the ATF6 pathway of the UPR limits ATZ-dependent cell toxicity by selectively promoting ER-associated degradation of ATZ and is thereby a potential target to prevent hepatocyte loss in addition to autophagy-enhancing drugs.


Subject(s)
Activating Transcription Factor 6/metabolism , Carcinoma, Hepatocellular/metabolism , Mitochondria, Liver/metabolism , Mutation, Missense , Neoplasm Proteins/metabolism , alpha 1-Antitrypsin/metabolism , Activating Transcription Factor 6/genetics , Amino Acid Substitution , Animals , Apoptosis/genetics , Autophagy/genetics , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA Fragmentation , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Mice , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Neoplasm Proteins/genetics , Proteolysis , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Unfolded Protein Response/genetics , alpha 1-Antitrypsin/genetics
7.
Mol Endocrinol ; 24(9): 1805-21, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20631012

ABSTRACT

Melanocortin-4 receptor (MC4R) is a G protein-coupled receptor expressed in the brain where it controls food intake. Many obesity-linked MC4R variants are poorly expressed at the plasma membrane and are retained intracellularly. We have studied the intracellular localization of four obesity-linked MC4R variants, P78L, R165W, I316S, and I317T, in immortalized neurons. We find that these variants are all retained in the endoplasmic reticulum (ER), are ubiquitinated to a greater extent than the wild-type (wt) receptor, and induce ER stress with increased levels of ER chaperones as compared with wt-MC4R and appearance of CCAAT/enhancer-binding protein homologous protein (CHOP). Expression of the X-box-binding-protein-1 (XBP-1) with selective activation of a protective branch of the unfolded protein response did not have any effect on the cell surface expression of MC4R-I316S. Conversely, the pharmacological chaperone 4-phenyl butyric acid (PBA) increased the cell surface expression of wt-MC4R, MC4R-I316S, and I317T by more than 40%. PBA decreased ubiquitination of MC4R-I316S and prevented ER stress induced by expression of the mutant, suggesting that the drug functions to promote MC4R folding. MC4R-I316S rescued to the cell surface is functional, with a 52% increase in agonist-induced cAMP production, as compared with untreated cells. Also direct inhibition of wt-MC4R and MC4R-I316S ubiquitination by a specific inhibitor of the ubiquitin-activating enzyme 1 increased by approximately 40% the expression of the receptors at the cell surface, and the effects of PBA and ubiquitin-activating enzyme 1 were additive. These data offer a cell-based rationale that drugs that improve MC4R folding or decrease ER-associated degradation of the receptor may function to treat some forms of hereditary obesity.


Subject(s)
Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Mutation/genetics , Obesity/genetics , Phenylbutyrates/pharmacology , Protein Folding , Receptor, Melanocortin, Type 4/genetics , Cell Line, Tumor , Cell Membrane/drug effects , Cyclic AMP/biosynthesis , Endoplasmic Reticulum/drug effects , Humans , Mutant Proteins/metabolism , Obesity/pathology , Protein Folding/drug effects , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/metabolism , Recombinant Fusion Proteins/metabolism , Ubiquitination/drug effects , Unfolded Protein Response/drug effects
8.
J Biol Chem ; 283(51): 35517-25, 2008 Dec 19.
Article in English | MEDLINE | ID: mdl-18974096

ABSTRACT

Vinblastine and other microtubule inhibitors used as antimitotic cancer drugs characteristically promote the phosphorylation of the key anti-apoptotic protein, Bcl-xL. However, putative sites of phosphorylation have been inferred based on potential recognition by JNK, and no direct biochemical analysis has been performed. In this study we used protein purification and mass spectrometry to identify Ser-62 as a single major site in vivo. Site-directed mutagenesis confirmed Ser-62 to be the site of Bcl-xL phosphorylation induced by several microtubule inhibitors tested. Vinblastine-treated cells overexpressing a Ser-62 --> Ala mutant showed highly significantly reduced apoptosis compared with cells expressing wild-type Bcl-xL. Co-immunoprecipitation revealed that phosphorylation caused wild-type Bcl-xL to release bound Bax, whereas phospho-defective Bcl-xL retained the ability to bind Bax. In contrast, phospho-mimic (Ser-62 --> Asp) Bcl-xL exhibited a reduced capacity to bind Bax. Functional tests were performed by transiently co-transfecting Bax in the context of different Bcl-xL mutants. Co-expression of wild-type or phospho-defective Bcl-xL counteracted the adverse effects of Bax expression on cell viability, whereas phospho-mimic Bcl-xL failed to provide the same level of protection against Bax. These studies suggest that Bcl-xL phosphorylation induced by microtubule inhibitors plays a key pro-apoptotic role at least in part by disabling the ability of Bcl-xL to bind Bax.


Subject(s)
Apoptosis/physiology , Microtubules/metabolism , Peptide Mapping , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Amino Acid Substitution , Cell Line , Cell Survival/physiology , Humans , Microtubules/genetics , Mutagenesis, Site-Directed/methods , Phosphorylation/drug effects , Protein Binding/physiology , Tubulin Modulators/pharmacology , Vincristine/pharmacology , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
9.
Autophagy ; 4(3): 375-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18212527

ABSTRACT

A hallmark of some endoplasmic reticulum (ER)-storage diseases is the formation of inclusion bodies (IBs) that are membrane-limited. The nature and function of the IBs has started to be investigated. We have recently found that sequestration of mutated alpha1-antitrypsin (ATZ) into IBs is a cell protective mechanism that maintains ER function. We also found that IBs are ER-derived and yet separate from the main ER and do not have markers of autophagosomes and lysosomes. We propose that formation of the IBs is a quality control mechanism that leads to storage of unwanted proteins outside the secretory pathway by a mechanism different than direct autophagosome formation from the ER.


Subject(s)
Autophagy/physiology , Endoplasmic Reticulum/physiology , Inclusion Bodies/physiology , Phagosomes/physiology , Animals , Humans , alpha 1-Antitrypsin/metabolism
10.
Mol Biol Cell ; 19(2): 572-86, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18045994

ABSTRACT

A variant alpha1-antitrypsin with E342K mutation has a high tendency to form intracellular polymers, and it is associated with liver disease. In the hepatocytes of individuals carrying the mutation, alpha1-antitrypsin localizes both to the endoplasmic reticulum (ER) and to membrane-surrounded inclusion bodies (IBs). It is unclear whether the IBs contribute to cell toxicity or whether they are protective to the cell. We found that in hepatoma cells, mutated alpha1-antitrypsin exited the ER and accumulated in IBs that were negative for autophagosomal and lysosomal markers, and contained several ER components, but not calnexin. Mutated alpha1-antitrypsin induced IBs also in neuroendocrine cells, showing that formation of these organelles is not cell type specific. In the presence of IBs, ER function was largely maintained. Increased levels of calnexin, but not of protein disulfide isomerase, inhibited formation of IBs and lead to retention of mutated alpha1-antitrypsin in the ER. In hepatoma cells, shift of mutated alpha1-antitrypsin localization to the ER by calnexin overexpression lead to cell shrinkage, ER stress, and impairment of the secretory pathway at the ER level. We conclude that segregation of mutated alpha1-antitrypsin from the ER to the IBs is a protective cell response to maintain a functional secretory pathway.


Subject(s)
Cytoprotection , Endoplasmic Reticulum/metabolism , Inclusion Bodies/metabolism , Mutant Proteins/metabolism , alpha 1-Antitrypsin/metabolism , Animals , Autophagy , Biomarkers/metabolism , Calnexin/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/ultrastructure , Cell Line, Tumor , Cell Size , Endoplasmic Reticulum/ultrastructure , Humans , Inclusion Bodies/enzymology , Inclusion Bodies/ultrastructure , Lysosomes/metabolism , Lysosomes/ultrastructure , Mice , Neurites/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuropeptides/metabolism , Phagosomes/metabolism , Phagosomes/ultrastructure , Protein Disulfide-Isomerases/metabolism , Protein Folding , Protein Transport , Serpins/metabolism , Time Factors , Neuroserpin
11.
J Biol Chem ; 282(7): 4963-4974, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17166828

ABSTRACT

Melanocortin-4 receptor (MC4R) is a G protein-coupled receptor (GPCR) that binds alpha-melanocyte-stimulating hormone (alpha-MSH) and has a central role in the regulation of appetite and energy expenditure. Most GPCRs are endocytosed following binding to the agonist and receptor desensitization. Other GPCRs are internalized and recycled back to the plasma membrane constitutively, in the absence of the agonist. In unstimulated neuroblastoma cells and immortalized hypothalamic neurons, epitopetagged MC4R was localized both at the plasma membrane and in an intracellular compartment. These two pools of receptors were in dynamic equilibrium, with MC4R being rapidly internalized and exocytosed. In the absence of alpha-MSH, a fraction of cell surface MC4R localized together with transferrin receptor and to clathrin-coated pits. Constitutive MC4R internalization was impaired by expression of a dominant negative dynamin mutant. Thus, MC4R is internalized together with transferrin receptor by clathrin-dependent endocytosis. Cell exposure toalpha-MSH reduced the amount of MC4R at the plasma membrane by blocking recycling of a fraction of internalized receptor, rather than by increasing its rate of endocytosis. The data indicate that, in neuronal cells, MC4R recycles constitutively and that alpha-MSH modulates MC4R residency at the plasma membrane by acting at an intracellular sorting step.


Subject(s)
Hypothalamus/metabolism , Neurons/metabolism , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/pharmacology , Animals , Appetite/drug effects , Appetite/genetics , Cell Line, Transformed , Cell Line, Tumor , Cell Membrane/metabolism , Clathrin-Coated Vesicles/metabolism , Endocytosis/drug effects , Endocytosis/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Exocytosis/drug effects , Exocytosis/genetics , Gene Expression , Humans , Mice , Mutation , Protein Transport/drug effects , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/genetics , Receptors, Transferrin/metabolism , Retinoblastoma/metabolism , alpha-MSH/metabolism
12.
World J Gastroenterol ; 12(40): 6458-63, 2006 Oct 28.
Article in English | MEDLINE | ID: mdl-17072978

ABSTRACT

AIM: To determine the effects of prophylactic peroxi-some proliferator-activated receptor (PPARgamma) agonist administration in an experimental model of post-endoscopic retrograde cholangiopancreatography (post-ERCP) acute pancreatitis. METHODS: Post-ERCP pancreatitis was induced in male Wistar rats by infusion of contrast medium into the pancreatic duct. In additional group, rosiglitazone, a PPARgamma agonist, was administered 1 h before infusion of contrast medium. Plasma and pancreas samples were obtained 6 h after the infusion. RESULTS: Infusion of contrast medium into the pan-creatic duct resulted in an inflammatory process characterized by increased lipase levels in plasma, and edema and myeloperoxidase activity (MPO) in pancreas. This result correlated with the activation of nuclear factor kappaB (NFkappaB) and the inducible NO synthase (iNOS) expression in pancreatic cells. Rosiglitazone reduced the increase in lipase and the level of edema and the increase in myeloperoxidase as well as the activation of NFkappaB and iNOS expression. CONCLUSION: A single oral dose of rosiglitazone, given 1 h before post-ERCP pancreatitis induction is effective in reducing the severity of the subsequent inflammatory process. The protective effect of rosiglitazone was associated with NFkappaB inhibition and the blockage of leukocyte infiltration in pancreas.


Subject(s)
Cholangiopancreatography, Endoscopic Retrograde/adverse effects , PPAR gamma/agonists , Pancreatitis/drug therapy , Thiazolidinediones/therapeutic use , Acute Disease , Animals , Contrast Media/adverse effects , Gene Expression Regulation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Lipase/blood , Liver/enzymology , Male , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Pancreatitis/chemically induced , Peroxidase/genetics , Peroxidase/metabolism , Rats , Rats, Wistar , Rosiglitazone , Severity of Illness Index , Thiazolidinediones/pharmacology
13.
J Immunol ; 176(6): 3774-9, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16517747

ABSTRACT

Pancreatitis-associated protein I (PAP I), also known as HIP, p23, or Reg2 protein, has recently been implicated in the endogenous regulation of inflammation. Although it was initially characterized as a protein that is overexpressed in acute pancreatitis, PAP I has also been associated with a number of inflammatory diseases, such as Crohn's disease. Knowing that PAP I and IL-10 responses share several features, we have used a pancreatic acinar cell line (AR42J) to assess the extent to which their expression is reciprocally regulated, and whether the JAK/STAT and NF-kappaB signaling pathways are involved in the suppression of inflammation mediated by PAP I. We observed that PAP I is induced in epithelial cells by IL-10 and by PAP I itself. In contrast, we found phosphorylation and nuclear translocation of STAT3 and induction of suppressor of cytokine signaling 3 in response to PAP I exposure. Finally, a JAK-specific inhibitor, tyrphostin AG490, markedly prevented PAP I-induced NF-kappaB inhibition, pointing to a cross-talk between JAK/STAT3 and NF-kappaB signaling pathways. Together, these findings indicate that PAP I inhibits the inflammatory response by blocking NF-kappaB activation through a STAT3-dependent mechanism. Important functional similarities to the anti-inflammatory cytokine IL-10 suggest that PAP I could play a role similar to that of IL-10 in epithelial cells.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Epithelial Cells/metabolism , Lectins, C-Type/metabolism , NF-kappa B/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Active Transport, Cell Nucleus , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/pharmacology , Cell Line , Gene Expression Regulation , Interleukin-10/genetics , Interleukin-10/metabolism , Lectins, C-Type/genetics , NF-kappa B/metabolism , Pancreatitis-Associated Proteins , Phosphotyrosine/metabolism , Protein Biosynthesis , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/genetics , Rats , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Tyrphostins/pharmacology
14.
Intensive Care Med ; 31(3): 487-90, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15668763

ABSTRACT

OBJECTIVE: To design, implement, and test a selective lung ventilator for setting a rat model of unilateral ventilator-induced lung injury (VILI). DESIGN AND SETTING: Interventional animal study in a university laboratory for animal research. SUBJECTS: Anesthetized and paralyzed male Wistar rats. INTERVENTIONS: A selective ventilator designed to apply varying tidal volume, PEEP, and breathing gas to each lung of the rat was implemented and evaluated. Five control animals were ventilated at 7 ml/kg (3.5 ml/kg each lung). Unilateral VILI was induced in six animals subjected to selective ventilation (3.5 ml/kg in one lung and 15 ml/kg in the other lung). After 3 h of ventilation the animals were killed and the lungs excised. MEASUREMENTS AND RESULTS: Lung edema was assessed by means of the ratio between wet and dry lung weights. No significant differences were found in lungs of control animals (5.16+/-0.22 and 4.96+/-0.25), but the W/D ratio in the over ventilated lung (8.98+/-3.80) was significantly greater than that in the normally ventilated lung (4.76+/-0.15), indicating selective induction of lung edema by over stretch. CONCLUSIONS: This selective ventilator can be implemented into a rat model of unilateral VILI to gain further insight into the mechanisms of pulmonary injury induced by different ventilatory strategies.


Subject(s)
Disease Models, Animal , Respiration, Artificial/adverse effects , Respiratory Distress Syndrome/etiology , Animals , Male , Positive-Pressure Respiration , Pressure , Rats , Rats, Wistar , Respiratory Distress Syndrome/diagnosis , Tidal Volume
15.
Surgery ; 137(2): 186-91, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15674200

ABSTRACT

BACKGROUND: The liver is a source of systemic proinflammatory mediators in acute pancreatitis. We have investigated the effects of blood from the pancreas and intestine in liver activation and lung inflammation during early stages of experimental acute pancreatitis in a rat model. METHODS: A portosystemic shunt and a mesosystemic shunt were created to prevent the passage of blood coming from the pancreas and the intestine, respectively, to the liver. Pancreatitis was induced by retrograde injection of 5% sodium taurocholate into the biliopancreatic duct. After 3 hours, the inflammatory process in the lung and intestine, plasma levels of tumor necrosis factor (TNF)-alpha and their soluble receptor, and mRNA expression of inflammatory mediators in the lung were evaluated. RESULTS: Portocaval shunting of blood prevented the inflammatory process in the lung, an increase in plasma TNF-alpha concentration, and the expression of TNF-alpha, interleukin (IL)-1beta, and heat-shock protein (HSP)-72 in the lung, but had no effect on plasma levels of soluble TNF-alpha receptor or on expression of inducible nitric oxide synthase (iNOS) and macrophage inflammatory protein (MIP)-2 in the lung. In contrast, mesocaval shunting of blood did not modify any of the parameters evaluated. CONCLUSIONS: Pancreatic blood, but not intestinal blood, plays a key role in liver activation during experimental acute pancreatitis.


Subject(s)
Pancreatitis/blood , Pancreatitis/etiology , Acute Disease , Animals , Base Sequence , Disease Models, Animal , Inflammation Mediators/metabolism , Intestines/blood supply , Liver/blood supply , Lung/immunology , Male , Pancreas/blood supply , Pancreatitis/genetics , Pancreatitis/immunology , Portal System , Portasystemic Shunt, Surgical , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor, Type I/blood , Tumor Necrosis Factor-alpha/metabolism
16.
Planta Med ; 70(9): 866-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15503356

ABSTRACT

A new beta-hydroxy-gamma-methyl-gamma-lactone bistetrahydrofuranic acetogenin, tucumanin, with the infrequent symmetrical threo/trans/threo/trans/threo relative configuration at the tetrahydrofuran rings was isolated from Annona cherimolia (Annonaceae) seeds. The inhibitory potency on the mitochondrial complex I of acetogenins with this relative configuration (tucumanin and asimicin)was compared with that shown by the corresponding pairs with an asymmetrical threo/trans/threo/trans/erythro relative configuration (laherradurin/rolliniastatin-2, and itrabin/molvizarin). All these compounds act as selective inhibitors of mitochondrial complex I in the 0.18 - 1.55 nM range.


Subject(s)
Annona , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Furans/pharmacology , Lactones/pharmacology , Phytotherapy , Plant Extracts/pharmacology , Electron Transport Complex I/metabolism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Furans/administration & dosage , Furans/therapeutic use , Humans , Inhibitory Concentration 50 , Lactones/administration & dosage , Lactones/therapeutic use , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Seeds
17.
Free Radic Biol Med ; 37(10): 1640-7, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15477015

ABSTRACT

During the early stages of acute pancreatitis, acute respiratory distress syndrome often occurs. This is associated with the release of proinflammatory mediators into the blood, but it remains unclear why these mediators induce inflammation especially in the lung. One of the first events occurring during the progression of acute pancreatitis is the induction of P-selectin expression in the endothelial cells of the lung. This expression has been associated with the generation of superoxide radicals by circulating xanthine oxidase. Because this enzyme needs molecular oxygen to perform the reaction, we have hypothesized that oxygen present in the alveolar space favors the generation of free radicals by xanthine oxidase and explains why P-selectin is expressed only in the lung. For this purpose, we evaluated the progression of the inflammatory process in rats with induced acute pancreatitis and one lung breathing nitrogen while the other lung continued breathing air. Acute pancreatitis was induced by intraductal administration of taurocholate and myeloperoxidase; P-selectin expression was measured 3 h after induction. Results indicated that, in the absence of oxygen in the alveolar space, the xanthine oxidase-dependent P-selectin expression did not occur and lung inflammation was significantly reduced.


Subject(s)
Oxygen/metabolism , P-Selectin/metabolism , Pancreatitis/metabolism , Pulmonary Alveoli/metabolism , Xanthine Oxidase/metabolism , Animals , Lung/metabolism , Lung/pathology , Male , Pancreatitis/chemically induced , Peroxidase/toxicity , Pulmonary Alveoli/pathology , Rats , Rats, Wistar , Superoxides/metabolism , Taurocholic Acid/toxicity
18.
J Leukoc Biol ; 76(3): 537-44, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15197232

ABSTRACT

Oxidant stress has been implicated in the pathogenesis of inflammatory bowel disease. Antioxidant enzymes, such as superoxide dismutase (SOD), are candidate drugs for modulating this pathogenic factor. This study was designed to determine the therapeutic value of SOD in an experimental model of colitis and to study the mechanisms underlying its effects on intestinal inflammation. For that purpose, colitic (trinitrobenzene sulfonic acid-induced) and control rats were studied. Groups of colitic animals were treated with different doses of SOD (1, 4, or 13 mg/kg/day) or vehicle, starting after induction of colitis and during 7 days. Clinical and pathological markers of colitis severity and lipid peroxidation in colonic tissue were measured. Leukocyte-endothelial cell interactions in colonic venules and expression of vascular cell adhesion molecule 1 (VCAM-1) were determined. Development of colitis was associated with a significant loss in body weight, an increase in macroscopic and microscopic damage scores, and colonic myeloperoxidase activity. Administration of SOD significantly attenuated these changes in a dose-dependent manner and reduced lipid peroxidation in colonic tissue. The increase in leukocyte rolling and adhesion in colonic venules of colitic rats were significantly reduced by administration of SOD, 13 mg/kg/day. Development of colitis was associated with a marked increase in endothelial VCAM-1 expression, which was significantly reduced by treatment with SOD. In conclusion, treatment with SOD significantly reduces peroxidation reactions in the inflamed colon and affords significant amelioration of colonic inflammatory changes in experimental colitis. This effect is related to a reduction in VCAM-1 expression and leukocyte recruitment into the inflamed intestine.


Subject(s)
Cell Adhesion Molecules/antagonists & inhibitors , Chemotaxis, Leukocyte/drug effects , Colitis/drug therapy , Oxidative Stress/drug effects , Superoxide Dismutase/pharmacology , Animals , Body Weight/drug effects , Body Weight/immunology , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/immunology , Chemotaxis, Leukocyte/immunology , Colitis/chemically induced , Colitis/immunology , Colon/drug effects , Colon/metabolism , Colon/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Down-Regulation/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lipid Peroxidation/drug effects , Lipid Peroxidation/immunology , Male , Oxidative Stress/immunology , Peroxidase/drug effects , Peroxidase/immunology , Rats , Rats, Sprague-Dawley , Superoxide Dismutase/therapeutic use , Trinitrobenzenesulfonic Acid , Vascular Cell Adhesion Molecule-1/drug effects , Vascular Cell Adhesion Molecule-1/metabolism , Venules/drug effects , Venules/immunology , Venules/physiopathology
19.
Cytokine ; 25(4): 187-91, 2004 Feb 21.
Article in English | MEDLINE | ID: mdl-15164724

ABSTRACT

Clinical and experimental studies have shown increased concentrations of TNF-alpha and its soluble receptors in serum of patients with acute pancreatitis. In this work, we have investigated the time-course of TNF-alpha and its soluble receptors during taurocholate-induced acute pancreatitis. In addition, since TNF-alpha itself could mediate the shedding of its receptors, we have assessed the effect of inhibiting TNF-alpha production on the release of soluble TNF-alpha receptors in experimental acute pancreatitis. Our results indicate that soluble receptors are released in the early stages of the disease and this increase is concomitant with the release of TNF-alpha, which is mainly bound to specific proteins. The increased concentrations of its receptors strongly suggest that they could be these binding proteins. Inhibition of TNF-alpha generation with pentoxifylline abrogated the shedding of sTNF-alphaR1, but had no effect on sTNF-alphaR2. This finding suggests that the shedding of sTNF-alphaR1 is induced by TNF-alpha itself, but in the case of sTNF-alphaR2, the shedding appears to be induced by another mechanism.


Subject(s)
Pancreatitis/blood , Pancreatitis/pathology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/metabolism , Acute Disease , Animals , Disease Models, Animal , Male , Pentoxifylline/pharmacology , Rats , Rats, Wistar , Solubility
20.
Planta Med ; 70(3): 266-8, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15114508

ABSTRACT

Four bisbenzyltetrahydroisoquinoline alkaloids (-)-medelline, (+)-antioquine, (+)-aromoline, and (+)-obamegine were isolated from the fruits of Xylopia columbiana. These compounds, the previously isolated alkaloids (+)-thaligrisine and (+)-isotetrandrine, as well as their O-acetylated derivatives were assayed on submitochondrial particles from beef heart as inhibitors of the mammalian respiratory chain. The results revealed that these alkaloids act as selective inhibitors of mitochondrial complex I in a 0.15 - 4.71 microM range. O-Acetylation, which increases their lipophilicity, considerably increased the inhibitory potency.


Subject(s)
Annonaceae , Benzylisoquinolines/pharmacology , Enzyme Inhibitors/pharmacology , NADH, NADPH Oxidoreductases/drug effects , Phytotherapy , Plant Extracts/pharmacology , Animals , Benzylisoquinolines/administration & dosage , Benzylisoquinolines/therapeutic use , Cattle , Electron Transport/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/therapeutic use , Inhibitory Concentration 50 , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/biosynthesis , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...