Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 414
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 316(2): G263-G277, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30520694

RESUMO

Rat pancreatic acinar cells possess only the p21-activated kinase (PAKs), PAK4 of the group II PAK, and it is activated by gastrointestinal hormones/neurotransmitters stimulating PLC and by a number of growth factors. However, little is known generally of cAMP agents causing PAK4 activation, and there are no studies with gastrointestinal hormones/neurotransmitters activating cAMP cascades. In the present study, we examined the ability of VIP and secretin, which stimulate cAMP generation in pancreatic acini, to stimulate PAK4 activation, the signaling cascades involved, and their possible role in activating sodium-potassium adenosine triphosphatase (Na+,K+-ATPase). PAK4 activation was compared with activation of the well-established cAMP target, cyclic AMP response element binding protein (CREB). Secretin-stimulated PAK4 activation was inhibited by KT-5720 and PKA Type II inhibitor (PKI), protein kinase A (PKA) inhibitors, whereas VIP activation was inhibited by ESI-09 and HJC0197, exchange protein directly activated by cAMP (EPAC) inhibitors. In contrast, both VIP/secretin-stimulated phosphorylation of CREB (pCREB) via EPAC activation; however, it was inhibited by the p44/42 inhibitor PD98059 and the p38 inhibitor SB202190. The specific EPAC agonist 8-CPT-2- O-Me-cAMP as well 8-Br-cAMP and forskolin stimulated PAK4 activation. Secretin/VIP activation of Na+,K+-ATPase, was inhibited by PAK4 inhibitors (PF-3758309, LCH-7749944). These results demonstrate PAK4 is activated in pancreatic acini by stimulation of both VIP-/secretin-preferring receptors, as is CREB. However, they differ in their signaling cascades. Furthermore, PAK4 activation is needed for Na+,K+ATPase activation, which mediates pancreatic fluid secretion. These results, coupled with recent studies reporting PAKs are involved in both pancreatitis/pancreatic cancer growth/enzyme secretion, show that PAK4, similar to PAK2, likely plays an important role in both pancreatic physiological/pathological responses. NEW & NOTEWORTHY Pancreatic acini possess only the group II p21-activated kinase, PAK4, which is activated by PLC-stimulating agents/growth factors and is important in enzyme-secretion/growth/pancreatitis. Little information exists on cAMP-activating agents stimulating group II PAKs. We studied ability/effect of cyclic AMP-stimulating agents [vasoactive intestinal polypeptide (VIP), secretin] on PAK4 activity in rat pancreatic-acini. Both VIP/secretin activated PAK4/CREB, but the cAMP signaling cascades differed for EPAC, MAPK, and PKA pathways. Both hormones require PAK4 activation to stimulate sodium-potassium adenosine triphosphatase activity. This study shows PAK4 plays an important role in VIP-/secretin-stimulated pancreatic fluid secretion and suggests it plays important roles in pancreatic acinar physiological/pathophysiological responses mediated by cAMP-activating agents.


Assuntos
Células Acinares/efeitos dos fármacos , Antineoplásicos/farmacologia , AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/efeitos dos fármacos , Células Acinares/metabolismo , Animais , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Masculino , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Pirazóis/farmacologia , Pirróis/farmacologia , Quinazolinas/farmacologia , Ratos Sprague-Dawley , Secretina/efeitos dos fármacos
2.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G302-G317, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29672153

RESUMO

p21-activated kinases (PAKs) are highly conserved serine/threonine protein kinases, which are divided into two groups: group-I (PAKs1-3) and group-II (PAKs4-6). In various tissues, Group-II PAKs play important roles in cytoskeletal dynamics and cell growth as well as neoplastic development/progression. However, little is known about Group-II PAK's role in a number of physiological events, including their ability to be activated by gastrointestinal (GI) hormones/neurotransmitters/growth factors (GFs). We used rat pancreatic acini to explore the ability of GI hormones/neurotransmitters/GFs to activate Group-II-PAKs and the signaling cascades involved. Only PAK4 was detected in pancreatic acini. PAK4 was activated by endothelin, secretagogues-stimulating phospholipase C (bombesin, CCK-8, and carbachol), by pancreatic GFs (insulin, insulin-like growth factor 1, hepatocyte growth factor, epidermal growth factor, basic fibroblast growth factor, and platelet-derived growth factor), and by postreceptor stimulants (12-O-tetradecanoylphobol-13-acetate and A23187 ). CCK-8 activation of PAK4 required both high- and low-affinity CCK1-receptor state activation. It was reduced by PKC-, Src-, p44/42-, or p38-inhibition but not with phosphatidylinositol 3-kinase-inhibitors and only minimally by thapsigargin. A protein kinase D (PKD)-inhibitor completely inhibited CCK-8-stimulated PKD-activation; however, stimulated PAK4 phosphorylation was only inhibited by 60%, demonstrating that it is both PKD-dependent and PKD-independent. PF-3758309 and LCH-7749944, inhibitors of PAK4, decreased CCK-8-stimulated PAK4 activation but not PAK2 activation. Each inhibited ERK1/2 activation and amylase release induced by CCK-8 or bombesin. These results show that PAK4 has an important role in modulating signal cascades activated by a number of GI hormones/neurotransmitters/GFs that have been shown to mediate both physiological/pathological responses in acinar cells. Therefore, in addition to the extensive studies on PAK4 in pancreatic cancer, PAK4 should also be considered an important signaling molecule for pancreatic acinar physiological responses and, in the future, should be investigated for a possible role in pancreatic acinar pathophysiological responses, such as in pancreatitis. NEW & NOTEWORTHY This study demonstrates that the only Group-II p21-activated kinase (PAK) in rat pancreatic acinar cells is PAK4, and thus differs from islets/pancreatic cancer. Both gastrointestinal hormones/neurotransmitters stimulating PLC and pancreatic growth factors activate PAK4. With cholecystokinin (CCK), activation is PKC-dependent/-independent, requires both CCK1-R affinity states, Src, p42/44, and p38 activation. PAK4 activation is required for CCK-mediated p42/44 activation/amylase release. These results show PAK4 plays an important role in mediating CCK physiological signal cascades and suggest it may be a target in pancreatic acinar diseases besides cancer.


Assuntos
Células Acinares/metabolismo , Bombesina , Colecistocinina/metabolismo , Hormônios Gastrointestinais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurotransmissores/metabolismo , Pâncreas , Quinases Ativadas por p21 , Animais , Bombesina/metabolismo , Bombesina/farmacologia , Trato Gastrointestinal/metabolismo , Neurotransmissores/farmacologia , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatopatias/metabolismo , Ratos , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/classificação , Quinases Ativadas por p21/metabolismo
3.
Biochem Pharmacol ; 115: 64-76, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27346274

RESUMO

Bombesin-receptor-subtype-3 (BB3 receptor) is a G-protein-coupled-orphan-receptor classified in the mammalian Bombesin-family because of high homology to gastrin-releasing peptide (BB2 receptor)/neuromedin-B receptors (BB1 receptor). There is increased interest in BB3 receptor because studies primarily from knockout-mice suggest it plays roles in energy/glucose metabolism, insulin-secretion, as well as motility and tumor-growth. Investigations into its roles in physiological/pathophysiological processes are limited because of lack of selective ligands. Recently, a selective, peptide-antagonist, Bantag-1, was described. However, because BB3 receptor has low-affinity for all natural, Bn-related peptides, little is known of the molecular basis of its high-affinity/selectivity. This was systematically investigated in this study for Bantag-1 using a chimeric-approach making both Bantag-1 loss-/gain-of-affinity-chimeras, by exchanging extracellular (EC) domains of BB3/BB2 receptor, and using site-directed-mutagenesis. Receptors were transiently expressed and affinities determined by binding studies. Bantag-1 had >5000-fold selectivity for BB3 receptor over BB2/BB1 receptors and substitution of the first EC-domain (EC1) in loss-/gain-of affinity-chimeras greatly affected affinity. Mutagenesis of each amino acid difference in EC1 between BB3 receptor/BB2 receptor showed replacement of His(107) in BB3 receptor by Lys(107) (H107K-BB3 receptor-mutant) from BB2 receptor, decreased affinity 60-fold, and three replacements [H107K, E11D, G112R] decreased affinity 500-fold. Mutagenesis in EC1's surrounding transmembrane-regions (TMs) demonstrated TM2 differences were not important, but R127Q in TM3 alone decreased affinity 400-fold. Additional mutants in EC1/TM3 explored the molecular basis for these changes demonstrated in EC1, particularly important is the presence of aromatic-interactions by His(107), rather than hydrogen-bonding or charge-charge interactions, for determining Bantag-1 high affinity/selectivity. In regard to Arg(127) in TM3, both hydrogen-bonding and charge-charge interactions contribute to the high-affinity/selectivity for Bantag-1.


Assuntos
Peptídeos/antagonistas & inibidores , Receptores da Bombesina/metabolismo , Animais , Células CHO , Cricetulus , Humanos , Camundongos , Mutagênese , Peptídeos/metabolismo , Ligação Proteica , Receptores da Bombesina/genética , Proteínas Recombinantes de Fusão/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 310(11): G1015-27, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27033118

RESUMO

In pancreatic acinar cells, the Src family of kinases (SFK) is involved in the activation of several signaling cascades that are implicated in mediating cellular processes (growth, cytoskeletal changes, apoptosis). However, the role of SFKs in various physiological responses such as enzyme secretion or in pathophysiological processes such as acute pancreatitis is either controversial, unknown, or incompletely understood. To address this, in this study, we investigated the role/mechanisms of SFKs in acute pancreatitis and enzyme release. Enzyme secretion was studied in rat dispersed pancreatic acini, in vitro acute-pancreatitis-like changes induced by supramaximal COOH-terminal octapeptide of cholecystokinin (CCK). SFK involvement assessed using the chemical SFK inhibitor (PP2) with its inactive control, 4-amino-7-phenylpyrazol[3,4-d]pyrimidine (PP3), under experimental conditions, markedly inhibiting SFK activation. In CCK-stimulated pancreatic acinar cells, activation occurred of trypsinogen, various MAP kinases (p42/44, JNK), transcription factors (signal transducer and activator of transcription-3, nuclear factor-κB, activator protein-1), caspases (3, 8, and 9) inducing apoptosis, LDH release reflective of necrosis, and various chemokines secreted (monocyte chemotactic protein-1, macrophage inflammatory protein-1α, regulated on activation, normal T cell expressed and secreted). All were inhibited by PP2, not by PP3, except caspase activation leading to apoptosis, which was increased, and trypsin activation, which was unaffected, as was CCK-induced amylase release. These results demonstrate SFK activation is playing a dual role in acute pancreatitis, inhibiting apoptosis and promoting necrosis as well as chemokine/cytokine release inducing inflammation, leading to more severe disease, as well as not affecting secretion. Thus, our studies indicate that SFK is a key mediator of inflammation and pancreatic acinar cell death in acute pancreatitis, suggesting it could be a potential therapeutic target in acute pancreatitis.


Assuntos
Células Acinares/metabolismo , Pancreatite Necrosante Aguda/metabolismo , Quinases da Família src/metabolismo , Células Acinares/efeitos dos fármacos , Animais , Apoptose , Caspases/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Colecistocinina/farmacologia , Sistema de Sinalização das MAP Quinases , Masculino , Necrose , Pancreatite Necrosante Aguda/patologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Fatores de Transcrição/metabolismo , Tripsinogênio/metabolismo , Quinases da Família src/antagonistas & inibidores
5.
Biochim Biophys Acta ; 1862(6): 1122-36, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26912410

RESUMO

In a recent study we explored Group-1-p21-activated kinases (GP.1-PAKs) in rat pancreatic acini. Only PAK2 was present; it was activated by gastrointestinal-hormones/neurotransmitters and growth factors in a PKC-, Src- and small-GTPase-mediated manner. PAK2 was required for enzyme-secretion and ERK/1-2-activation. In the present study we examined PAK2's role in CCK and TPA-activation of important distal signaling cascades mediating their physiological/pathophysiological effects and analyzed its role in pathophysiological processes important in early pancreatitis. In rat pancreatic acini, PAK2-inhibition by the specific, GP.1.PAK-inhibitor, IPA-3-suppressed cholecystokinin (CCK)/TPA-stimulated activation of focal-adhesion kinases and mitogen-activated protein-kinases. PAK2-inhibition reversed the dual stimulatory/inhibitory effect of CCK/TPA on the PI3K/Akt/GSK-3ß pathway. However, its inhibition did not affect PKC activation. PAK2-inhibition protected acini from CCK-induced ROS-generation; caspase/trypsin-activation, important in early pancreatitis; as well as from cell-necrosis. Furthermore, PAK2-inhibition reduced proteolytic-activation of PAK-2p34, which is involved in programmed-cell-death. To ensure that the study did not only rely in the specificity of IPA-3 as a PAK inhibitor, we used two other approaches for PAK inhibition, FRAX597 a ATP-competitive-GP.1-PAKs-inhibitor and infection with a PAK2-dominant negative(DN)-Advirus. Those two approaches confirmed the results obtained with IPA-3. This study demonstrates that PAK2 is important in mediating CCK's effect on the activation of signaling-pathways known to mediate its physiological/pathophysiological responses including several cellular processes linked to the onset of pancreatitis. Our results suggest that PAK2 could be a new, important therapeutic target to consider for the treatment of diseases involving deregulation of pancreatic acinar cells.


Assuntos
Células Acinares/patologia , Pâncreas/patologia , Pancreatite/patologia , Transdução de Sinais , Quinases Ativadas por p21/metabolismo , Células Acinares/enzimologia , Células Acinares/metabolismo , Animais , Morte Celular , Ativação Enzimática , Masculino , Pâncreas/enzimologia , Pâncreas/metabolismo , Pancreatite/enzimologia , Pancreatite/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Biochim Biophys Acta ; 1853(10 Pt A): 2371-82, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25979836

RESUMO

P-21-activated kinases (PAKs) are serine/threonine kinases comprising six isoforms divided in two groups, group-I (PAK1-3)/group-II (PAK4-6) which play important roles in cell cytoskeletal dynamics, survival, secretion and proliferation and are activated by diverse stimuli. However, little is known about PAKs ability to be activated by gastrointestinal (GI) hormones/neurotransmitters/growth-factors. We used rat pancreatic acini to explore the ability of GI-hormones/neurotransmitters/growth-factors to activate Group-I-PAKs and the signaling cascades involved. Only PAK2 was present in acini. PAK2 was activated by some pancreatic growth-factors [EGF, PDGF, bFGF], by secretagogues activating phospholipase-C (PLC) [CCK, carbachol, bombesin] and by post-receptor stimulants activating PKC [TPA], but not agents only mobilizing cellular calcium or increasing cyclic AMP. CCK-activation of PAK2 required both high- and low-affinity-CCK1-receptor-state activation. It was partially reduced by PKC- or Src-inhibition, but not with PI3K-inhibitors (wortmannin, LY294002) or thapsigargin. IPA-3, which prevents PAK2 binding to small-GTPases partially inhibited PAK2-activation, as well as reduced CCK-induced ERK1/2 activation and amylase release induced by CCK or bombesin. This study demonstrates pancreatic acini, possess only one Group-I-PAK, PAK2. CCK and other GI-hormones/neurotransmitters/growth-factors activate PAK2 via small GTPases (CDC42/Rac1), PKC and SFK but not cytosolic calcium or PI3K. CCK-activation of PAK2 showed several novel features being dependent on both receptor-activation states, having PLC- and PKC-dependent/independent components and small-GTPase-dependent/independent components. These results show that PAK2 is important in signaling cascades activated by numerous pancreatic stimuli which mediate their various physiological/pathophysiological responses and thus could be a promising target for the development of therapies in some pancreatic disorders such as pancreatitis.


Assuntos
Hormônios Gastrointestinais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurotransmissores/metabolismo , Pâncreas Exócrino/enzimologia , Transdução de Sinais/fisiologia , Quinases Ativadas por p21/metabolismo , Animais , Ativação Enzimática/fisiologia , Masculino , Pâncreas Exócrino/citologia , Ratos , Ratos Sprague-Dawley
8.
J Cell Biochem ; 116(1): 22-36, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25079913

RESUMO

Recent studies report the Src-family kinases (SFK's) are important in a number of physiological and pathophysiological responses of pancreatic acinar cells (pancreatitis, growth, apoptosis); however, the role of SFKs in various signaling cascades important in mediating these cell functions is either not investigated or unclear. To address this we investigated the action of SFKs in these signaling cascades in rat pancreatic acini by modulating SFK activity using three methods: adenovirus-induced expression of an inactive dominant-negative CSK (Dn-CSK-Advirus) or wild-type CSK (Wt-CSK-Advirus), which activate or inhibit SFK, respectively, or using the chemical inhibitor, PP2, with its inactive control, PP3. CCK (0.3, 100 nM) and TPA (1 µM) activated SFK and altered the activation of FAK proteins (PYK2, p125(FAK)), adaptor proteins (p130(CAS), paxillin), MAPK (p42/44, JNK, p38), Shc, PKC (PKD, MARCKS), Akt but not GSK3-ß. Changes in SFK activity by using the three methods of altering SFK activity affected CCK/TPAs activation of SFK, PYK2, p125(FAK), p130(CAS), Shc, paxillin, Akt but not p42/44, JNK, p38, PKC (PKD, MARCKS) or GSK3-ß. With chemical inhibition the active SFK inhibitor, PP2, but not the inactive control analogue, PP3, showed these effects. For all stimulated changes pre-incubation with both adenoviruses showed similar effects to chemical inhibition of SFK activity. In conclusion, using three different approaches to altering Src activity allowed us to define fully for the first time the roles of SFKs in acinar cell signaling. Our results show that in pancreatic acinar cells, SFKs play a much wider role than previously reported in activating a number of important cellular signaling cascades shown to be important in mediating both acinar cell physiological and pathophysiological responses.


Assuntos
Pâncreas/citologia , Pâncreas/metabolismo , Quinases da Família src/metabolismo , Animais , Western Blotting , Colecistocinina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Masculino , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia
9.
Peptides ; 51: 91-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24220502

RESUMO

Several studies showed that the orphan Bombesin Receptor Subtype-3 (BRS-3) - member of the bombesin receptor family - has an important role in glucose homeostasis (v.g.: BRS-3-KO mice developed mild obesity, and decreased levels of BRS-3 mRNA/protein have been described in muscle from obese (OB) and type 2 diabetic (T2D) patients). In this work, to gain insight into BRS-3 receptor cell signaling pathways, and its implication on glucose metabolism, primary cultured myocytes from normal subjects, OB or T2D patients were tested using high affinity ligand - [d-Tyr(6),ß-Ala(11),Phe(13),Nle(14)]bombesin6-14. In muscle cells from all metabolic conditions, the compound significantly increased not only MAPKs, p90RSK1, PKB and p70s6K phosphorylation levels, but also PI3K activity; moreover, it produced a dose-response stimulation of glycogen synthase a activity and glycogen synthesis. Myocytes from OB and T2D patients were more sensitive to the ligand than normal, and T2D cells even more than obese myocytes. These results widen the knowledge of human BRS-3 cell signaling pathways induced by a BRS-3 agonist, described its insulin-mimetic effects on glucose metabolism, showed the role of BRS-3 receptor in glucose homeostasis, and also propose the employing of BRS-3/ligand system, as participant in the obese and diabetic therapies.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Obesidade/metabolismo , Receptores da Bombesina/fisiologia , Adulto , Idoso , Bombesina/farmacologia , Células Cultivadas , Diabetes Mellitus Tipo 2/patologia , Feminino , Glicogênio/biossíntese , Glicogênio Sintase/metabolismo , Homeostase , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Obesidade/patologia , Fragmentos de Peptídeos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores da Bombesina/agonistas , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Transdução de Sinais
10.
Biochim Biophys Acta ; 1823(8): 1285-94, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22617836

RESUMO

For growth factors, cytokines, G-protein-coupled receptors and numerous other stimuli, the Src Family of kinases (SFK) play a central signaling role. SFKs also play an important role in pancreatic acinar cell function including metabolism, secretion, endocytosis, growth and cytoskeletal integrity, although the specific SFKs involved are not fully known. In the present study we used specific antibodies for the SFK, Yes, to determine its presence, activation by pancreatic secretagogues or growth factors, and interaction with cellular signaling cascades mediated by CCK in which Yes participates in to cause acinar cell responses. Yes was identified in acini and secretagogues known to activate phospholipase C (PLC) [CCK, carbachol, bombesin] as well as post-receptor stimulants activating PKC [TPA] or mobilizing cellular calcium [thapsigargin/calcium ionophore (A23187)] each activated Yes. Secretin, which activates adenylate cyclase did not stimulate Yes, nor did pancreatic growth factors. CCK activation of Yes required both high- and low-affinity CCK(1)-receptor states. TPA-/CCK-stimulated Yes activation was completely inhibited by thapsigargin and the PKC inhibitor, GF109203X. CCK/TPA stimulated the association of Yes with focal adhesion kinases (Pyk2, FAK) and its autophosphorylated forms (pY397FAK, pY402Pyk2). Moreover, CCK/TPA stimulated Yes interacted with a number of other signaling proteins, including Shc, PKD, p130(Cas), PI3K and PTEN. This study demonstrates that in rat pancreatic acini, the SFK member Yes is expressed and activated by CCK and other gastrointestinal hormones/neurotransmitters. Because its activation results in the direct activation of many cellular signaling cascades that have been shown to mediate CCK's effect in acinar cell function our results suggest that it is one of the important pancreatic SFKs mediating these effects.


Assuntos
Células Acinares/enzimologia , Hormônios Gastrointestinais/fisiologia , Neurotransmissores/fisiologia , Pâncreas/citologia , Proteínas Proto-Oncogênicas c-yes/metabolismo , Animais , Bombesina/farmacologia , Calcimicina/farmacologia , Ionóforos de Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Carbacol/farmacologia , Colecistocinina/farmacologia , Colecistocinina/fisiologia , Ativação Enzimática , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Hormônios Gastrointestinais/farmacologia , Indóis/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Masculino , Maleimidas/farmacologia , Neurotransmissores/farmacologia , Pâncreas/metabolismo , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/fisiologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Processamento de Proteína Pós-Traducional , Ratos , Ratos Sprague-Dawley , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Tapsigargina/farmacologia
11.
Biochim Biophys Acta ; 1813(12): 2145-56, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21810446

RESUMO

The novel PKCθ isoform is highly expressed in T-cells, brain and skeletal muscle and originally thought to have a restricted distribution. It has been extensively studied in T-cells and shown to be important for apoptosis, T-cell activation and proliferation. Recent studies showed its presence in other tissues and importance in insulin signaling, lung surfactant secretion, intestinal barrier permeability, platelet and mast-cell functions. However, little information is available for PKCθ activation by gastrointestinal (GI) hormones/neurotransmitters and growth factors. In the present study we used rat pancreatic acinar cells to explore their ability to activate PKCθ and the possible interactions with important cellular mediators of their actions. Particular attention was paid to cholecystokinin (CCK), a physiological regulator of pancreatic function and important in pathological processes affecting acinar function, like pancreatitis. PKCθ-protein/mRNA was present in the pancreatic acini, and T538-PKCθ phosphorylation/activation was stimulated only by hormones/neurotransmitters activating phospholipase C. PKCθ was activated in time- and dose-related manner by CCK, mediated 30% by high-affinity CCK(A)-receptor activation. CCK stimulated PKCθ translocation from cytosol to membrane. PKCθ inhibition (by pseudostrate-inhibitor or dominant negative) inhibited CCK- and TPA-stimulation of PKD, Src, RafC, PYK2, p125(FAK) and IKKα/ß, but not basal/stimulated enzyme secretion. Also CCK- and TPA-induced PKCθ activation produced an increment in PKCθ's direct association with AKT, RafA, RafC and Lyn. These results show for the first time the PKCθ presence in pancreatic acinar cells, its activation by some GI hormones/neurotransmitters and involvement in important cell signaling pathways mediating physiological responses (enzyme secretion, proliferation, apoptosis, cytokine expression, and pathological responses like pancreatitis and cancer growth).


Assuntos
Células Acinares/efeitos dos fármacos , Hormônios Gastrointestinais/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Isoenzimas/metabolismo , Neurotransmissores/farmacologia , Pâncreas Exócrino/efeitos dos fármacos , Proteína Quinase C/metabolismo , Células Acinares/citologia , Células Acinares/metabolismo , Animais , Western Blotting , Células Cultivadas , Ativação Enzimática , Imunoprecipitação , Isoenzimas/genética , Masculino , Pâncreas Exócrino/citologia , Pâncreas Exócrino/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C/genética , Proteína Quinase C-theta , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Colecistocinina A/genética , Receptor de Colecistocinina A/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Quinases raf/genética , Quinases raf/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
12.
Endoscopy ; 42(8): 664-71, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20669078

RESUMO

Well differentiated neuroendocrine tumors (NETs) of the stomach (gastric carcinoid tumors) are observed more often, with a tenfold increase in the US in the last 30 - 35 years, and the prognosis has improved greatly in that time. Nowadays most carcinoids of the stomach are diagnosed at an early stage. Four types of gastric NETs have been proposed and recognition of the type is important for defining the diagnostic approach and treatment. Often gastric NETs (especially type 1) are found incidentally during a gastroscopy performed for other reasons; most of these NETs are smaller than 20 mm in size. Conservative management and endoscopic surveillance is adequate for well differentiated, multifocal gastric carcinoids (type 1 or type 2 gastric NETs) that are less than 10 - 20 mm in diameter, unless they show angioinvasion, infiltrate the muscular wall, or have a proliferation rate above 2 %. Endoscopic ultrasound is the method of choice to determine tumor size and depth of infiltration. It is essential to distinguish between multifocal (types 1 and 2) and unifocal type 3 or type 4 gastric NETs, since surgery is indicated for type 3 gastric NETs larger than 10 mm in diameter and for poorly differentiated (localized) neuroendocrine gastric carcinomas (type 4 gastric NET). For optimal management, the type, biology, and stage of the tumor as well as the individual situation of the patient must be considered. Most patients with well differentiated gastric NETs can be treated conservatively and be followed up with endoscopic surveillance.


Assuntos
Tumor Carcinoide/diagnóstico , Tumor Carcinoide/patologia , Tumor Carcinoide/terapia , Carcinoma Neuroendócrino/patologia , Carcinoma Neuroendócrino/terapia , Mucosa Gástrica/patologia , Tumores Neuroendócrinos , Neoplasias Gástricas , Carcinoma Neuroendócrino/diagnóstico , Mucosa Gástrica/cirurgia , Gastroscopia , Humanos , Estadiamento de Neoplasias , Tumores Neuroendócrinos/classificação , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/epidemiologia , Tumores Neuroendócrinos/terapia , Neoplasias Gástricas/classificação , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/epidemiologia , Neoplasias Gástricas/terapia
13.
Peptides ; 31(2): 284-90, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19944725

RESUMO

We have previously demonstrated that pregnant ovine endometrium expresses the gastrin-releasing peptide (GRP) gene at a high level following conceptus implantation. Here we report the isolation, characterization and biological activity of ovine GRP 1-46, the primary product of this gene in the pregnant endometrium. Full thickness 125-140-day pregnant sheep uterus (term is 145 day) was homogenized in 80% acetonitrile/2% trifluoroacetic acid (1:7 ACN/TFA), concentrated on reverse-phase C18 cartridges and chromatographed successively on gel filtration (Sephadex G-50) and reverse-phase HPLC (C18 muBondapak). Purification was monitored by RIA. Purified GRP peptide was analysed by mass spectrometry giving a major mass ion at 4963 which corresponds exactly to GRP 1-46. Other mass ions from pro-GRP did not contain a biologically active N-terminus or antigenic determinant. Proteolytic cleavage of pro-GRP to give rise to GRP(1-46) would require preferential cleavage at the Glu-Glu bond by a Glu-C2-like enzyme, rather than the trypsin-like and C-terminal amidation enzymes (PAM) that produce GRP(18-27) and GRP(1-27) in other tissues. GRP 1-46 was synthesized and receptor binding and biological activity tested on a range of rodent and human cell lines that express GRP-related receptors GRPR, NMBR and BRS3. GRP 1-46 bound GRPR and NMBR with low affinity, and mobilized inositol phosphate in cell lines expressing the GRPR and NMBR, but not BRS-3. This study describes a new processed product of the GRP gene, GRP 1-46, which is highly expressed in the pregnant sheep endometrium and which acts as a weak agonist at the GRPR and NMBR.


Assuntos
Endométrio/química , Peptídeo Liberador de Gastrina/isolamento & purificação , Peptídeo Liberador de Gastrina/farmacologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Peptídeo Liberador de Gastrina/análise , Peptídeo Liberador de Gastrina/metabolismo , Humanos , Indóis/farmacologia , Fosfatos de Inositol/metabolismo , Camundongos , Dados de Sequência Molecular , Peso Molecular , Peptídeos/genética , Gravidez , Ligação Proteica/fisiologia , Precursores de Proteínas/genética , Piridinas/farmacologia , Ratos , Receptores da Bombesina/antagonistas & inibidores , Receptores da Bombesina/genética , Receptores da Bombesina/metabolismo , Ovinos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transfecção , Fosfolipases Tipo C/metabolismo
14.
Am J Med Genet B Neuropsychiatr Genet ; 147B(6): 807-13, 2008 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-18393381

RESUMO

The gastrin-releasing peptide receptor (GRPR) was implicated for the first time in the pathogenesis of Autism spectrum disorders (ASD) by Ishikawa-Brush et al. [Ishikawa-Brush et al. (1997): Hum Mol Genet 6: 1241-1250]. Since this original observation, only one association study [Marui et al. (2004): Brain Dev 26: 5-7] has further investigated, though unsuccessfully, the involvement of the GRPR gene in ASD. With the aim of contributing further information to this topic we have sequenced the entire coding region and the intron/exon junctions of the GRPR gene in 149 Italian autistic patients. The results of this study led to the identification of four novel point mutations, two of which, that is, C6S and L181F, involve amino acid changes identified in two patients with ASD and Rett syndrome, respectively. Both the leucine at position 181 and the cysteine at position 6 are strongly conserved in vertebrates. C6S and L181F mutant proteins were expressed in COS-7 and BALB/3T3 cells, but they did not affect either GRP's binding affinity or its potency for stimulating phospholipase C-mediated production of inositol 1,4,5-trisphosphate. In summary, our results do not provide support for a major role of the GRPR gene in ASD in the population of patients we have studied. However, there is a potential role of C6S and L181F mutations on GRPR function, and possibly in the pathogenesis of the autistic disorders in the two patients.


Assuntos
Transtorno Autístico/genética , Receptores da Bombesina/genética , Adolescente , Adulto , Idoso , Animais , Células 3T3 BALB , Células COS , Estudos de Casos e Controles , Criança , Chlorocebus aethiops , Análise Mutacional de DNA , Feminino , Humanos , Itália , Masculino , Camundongos , Pessoa de Meia-Idade , Linhagem , Mutação Puntual/fisiologia
15.
Pharmacol Rev ; 60(1): 1-42, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18055507

RESUMO

The mammalian bombesin receptor family comprises three G protein-coupled heptahelical receptors: the neuromedin B (NMB) receptor (BB(1)), the gastrin-releasing peptide (GRP) receptor (BB(2)), and the orphan receptor bombesin receptor subtype 3 (BRS-3) (BB(3)). Each receptor is widely distributed, especially in the gastrointestinal (GI) tract and central nervous system (CNS), and the receptors have a large range of effects in both normal physiology and pathophysiological conditions. The mammalian bombesin peptides, GRP and NMB, demonstrate a broad spectrum of pharmacological/biological responses. GRP stimulates smooth muscle contraction and GI motility, release of numerous GI hormones/neurotransmitters, and secretion and/or hormone release from the pancreas, stomach, colon, and numerous endocrine organs and has potent effects on immune cells, potent growth effects on both normal tissues and tumors, potent CNS effects, including regulation of circadian rhythm, thermoregulation; anxiety/fear responses, food intake, and numerous CNS effects on the GI tract as well as the spinal transmission of chronic pruritus. NMB causes contraction of smooth muscle, has growth effects in various tissues, has CNS effects, including effects on feeding and thermoregulation, regulates thyroid-stimulating hormone release, stimulates various CNS neurons, has behavioral effects, and has effects on spinal sensory transmission. GRP, and to a lesser extent NMB, affects growth and/or differentiation of various human tumors, including colon, prostate, lung, and some gynecologic cancers. Knockout studies show that BB(3) has important effects in energy balance, glucose homeostasis, control of body weight, lung development and response to injury, tumor growth, and perhaps GI motility. This review summarizes advances in our understanding of the biology/pharmacology of these receptors, including their classification, structure, pharmacology, physiology, and role in pathophysiological conditions.


Assuntos
Receptores da Bombesina , Animais , Doença , Humanos , Receptores da Bombesina/agonistas , Receptores da Bombesina/antagonistas & inibidores , Receptores da Bombesina/química , Receptores da Bombesina/fisiologia , Transdução de Sinais , Terminologia como Assunto
16.
Neurogastroenterol Motil ; 18(3): 217-25, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16487413

RESUMO

This study demonstrates the expression of functional somatostatin receptor (sstr) subtypes in human circular and longitudinal colonic smooth muscle cells (SMC). Native somatostatin (SS) and sstr subtype-specific analogues were used to characterize the sstr subtypes present in both cell types by contraction/relaxation studies. Qualitative and quantitative mRNA analysis and immunohistochemistry of sstr subtypes were also carried out. sstr subtype 2 mRNA was expressed in circular SMC, and various levels of subtypes 1, 2 and 3 mRNA were expressed in longitudinal colonic SMC. Native SS and each subtype-specific analogue exerted a modest, but significant, contraction, although inhibition of carbachol-induced contraction (relaxation) was the main effect on SMC from both layers. CH-288, a sstr subtype 1-specific analogue, and octreotide, a sstr subtype 2-specific analogue, were the most effective relaxant analogues on longitudinal and circular SMC, respectively. sstr subtypes display a distinct expression pattern on human colonic SMC; on circular SMC, subtype 2 is the only sstr, whereas sstr subtypes 1, 2 and 3 are expressed on human SMC isolated from the longitudinal layer. The contractile effects of SS are mediated through sstr subtype 2 and sstr subtype 1 on circular and longitudinal human colonic SMC, respectively.


Assuntos
Colo/fisiologia , Contração Muscular/fisiologia , Miócitos de Músculo Liso/metabolismo , Receptores de Somatostatina/biossíntese , Células Cultivadas , Colo/efeitos dos fármacos , Fármacos Gastrointestinais/farmacologia , Humanos , Imuno-Histoquímica , Contração Muscular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Octreotida/farmacologia , RNA Mensageiro/análise , Receptores de Somatostatina/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somatostatina/análogos & derivados , Somatostatina/farmacologia
17.
Biochim Biophys Acta ; 1763(1): 25-38, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16364465

RESUMO

Protein kinase C-delta (PKC-delta) becomes activated in pancreatic acini in response to cholecystokinin (CCK) and plays a pivotal role in the exocrine pancreatic secretion. Rottlerin, a polyphenolic compound, has been widely used as a potent and specific PKC-delta inhibitor. However, some recent studies showed that rottlerin was not effective in inhibiting PKCdelta activity in vitro and that may display unspecific effects. The aims of this work were to investigate the specificity of rottlerin as an inhibitor of PKC-delta activity in intact cells and to elucidate the biochemical causes of its unspecificity. Preincubation of pancreatic acini with rottlerin (6 microM) inhibited CCK-stimulated translocation, tyrosine phosphorylation (TyrP) and activation of PKC-delta in pancreatic acini in a time-dependent manner. Rottlerin inhibited amylase secretion stimulated by both PKC-dependent pathways (CCK, bombesin, carbachol, TPA) and also by PKC-independent pathways (secretin, VIP, cAMP analogue). CCK-stimulation of MAPK activation and p125(FAK) TyrP which are mediated by PKC-dependent and -independent pathways were also inhibited by rottlerin. Moreover, rottlerin rapidly depleted ATP content in pancreatic acini in a similar way as the mitochondrial uncouplers CCCP and FCCP. All studied inhibitory effects of rottlerin in pancreatic acini were mimicked by FCCP (agonists-stimulated amylase secretion, p125(FAK) TyrP, MAPK activation and PKC-delta TyrP and translocation). Finally, rottlerin as well as FCCP display a potent inhibitory effect on the activation of other PKC isoforms present in pancreatic acini. Our results suggest that rottlerin effects in pancreatic acini are not due to a specific PKC-delta blockade, but likely due to its negative effect on acini energy resulting in ATP depletion. Therefore, to study the role of PKC-delta in cellular processes using rottlerin it is essential to keep in mind that may deplete ATP levels and inhibit different PKC isoforms. Our results give reasons for a more careful choice of rottlerin for PKC-delta investigation.


Assuntos
Acetofenonas/farmacologia , Benzopiranos/farmacologia , Pâncreas Exócrino/enzimologia , Pâncreas Exócrino/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirosina/metabolismo , Trifosfato de Adenosina/metabolismo , Amilases/antagonistas & inibidores , Amilases/metabolismo , Animais , Carbonil Cianeto m-Clorofenil Hidrazona/farmacologia , Carbonil Cianeto p-Trifluormetoxifenil Hidrazona/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Colecistocinina/farmacologia , Quinase 1 de Adesão Focal/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pâncreas Exócrino/citologia , Pâncreas Exócrino/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Isoformas de Proteínas/metabolismo , Proteína Quinase C-delta/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Wistar , Receptor de Colecistocinina A/metabolismo
18.
Dig Liver Dis ; 36 Suppl 1: S106-20, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15077919

RESUMO

Numerous studies have established that gastroenteropancreatic endocrine tumours (carcinoids and pancreatic endocrine tumours) resemble a number of other tumours in overexpressing somatostatin receptors that can bind octreotide or lanreotide with high affinity (i.e. possess sst2/sst5 receptors). Recent studies report that radiolabelled somatostatin analogues can be used to image these tumours (somatostatin receptor scintigraphy) and may be useful for peptide-directed radionuclide therapy. In this paper the evidence is reviewed that has led to establishing somatostatin receptor scintigraphy as the initial imaging modality of choice in patients with gastroenteropancreatic tumours. This conclusion is based on an understanding of the results with conventional imaging modalities (ultrasound, computed tomographic scan, magnetic resonance imaging, angiography) available prior to somatostatin receptor scintigraphy and the results of studies demonstrating the sensitivity and specificity of somatostatin receptor scintigraphy. Most important in this regard are the results of studies that have assessed the use of somatostatin receptor scintigraphy on clinical management. Each of these areas is reviewed in this paper.


Assuntos
Neoplasias Gastrointestinais/diagnóstico por imagem , Tumores Neuroendócrinos/diagnóstico por imagem , Neoplasias Pancreáticas/diagnóstico por imagem , Receptores de Somatostatina , Neoplasias Gastrointestinais/diagnóstico , Neoplasias Gastrointestinais/terapia , Humanos , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/terapia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Cintilografia , Sensibilidade e Especificidade
19.
Biochim Biophys Acta ; 1643(1-3): 37-46, 2003 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-14654226

RESUMO

The receptor of hepatocyte growth factor (HGF), c-met induces different physiological responses in several cell types. Little is known about the role of HGF in exocrine pancreas. However, abnormal HGF signaling has been strongly implicated in pancreatic tumorigenesis and association of HGF with pancreatitis has been demonstrated. We have studied the presence of c-met and activation of their intracellular pathways associated in rat pancreatic acini in comparison with cholecystokinin (CCK) and epidermal growth factor (EGF). C-met expression in rat exocrine pancreas was identified by immunohistochemistry and immunoprecipitation followed by Western analysis. Tyrosine phosphorylation of c-met is strongly stimulated as well as kinase pathways leading to ERK1/2 cascade. HGF, but not CCK or EGF, selectively caused a consistent increase in the amount of p85 regulatory subunit of PI3-K present in anti-phosphotyrosine immunoprecipitates. Downstream of PI3-K, HGF increased Ser473 phosphorylation of Akt selectively, as CCK or EGF did not affect it. HGF selectively stimulated tyrosine phosphorylation of phosphatase PTP1D. HGF failed to promote the well-known CCK effects in pancreatic acini such as amylase secretion and intracellular calcium mobilization. Although HGF shares activation of ERK1/2 with CCK, we demonstrate that it promotes the selective activation of intracellular pathways not regulated by CCK or EGF. Our results suggest that HGF is an in vivo stimulus of pancreatic acini and provide novel insight into the transduction pathways and effects of c-met/HGF in normal pancreatic acinar cells.


Assuntos
Fator de Crescimento de Hepatócito/farmacologia , Pâncreas/citologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Sinalização do Cálcio/efeitos dos fármacos , Colecistocinina/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pâncreas/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Ratos Wistar
20.
Dig Liver Dis ; 35(8): 585-9, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-14567464

RESUMO

Until recently, the association of primary hyperparathyroidism and gastric carcinoid, with or without hypergastrinaemia, had been considered an incomplete form of multiple endocrine neoplasia type 1. This is because it seemed unlikely that the rare joint appearance of these diseases could occur only by chance. It is now possible to evaluate the pathogenetic involvement of the multiple endocrine neoplasia type 1 gene in many, apparently sporadic, clinical conditions. This is a case report of a female mimicking multiple endocrine neoplasia type 1 due to the presence of hyperparathyroidism, gastric carcinoid, and hypergastrinaemia. However, involvement of the MEN-1 gene (exons 2-10) was not detected, whereas hypergastrinaemia was attributed to a chronic atrophic gastritis.


Assuntos
Gastrite Atrófica/diagnóstico , Expressão Gênica , Neoplasia Endócrina Múltipla Tipo 1/genética , Idoso , Tumor Carcinoide/complicações , Diagnóstico Diferencial , Feminino , Gastrinas/sangue , Humanos , Hiperparatireoidismo/complicações , Neoplasia Endócrina Múltipla Tipo 1/diagnóstico , Neoplasias Gástricas/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...