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










Base de dados
Intervalo de ano de publicação
1.
Bone ; 123: 28-38, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30858147

RESUMO

The non-selective phosphodiesterase inhibitor pentoxifylline (PTX) is used for the treatment of intermittent claudication due to artery occlusion. Previous studies in rodents have reported salutary effects of the intraperitoneal administration of PTX in segmental bone defect and fracture healing, as well as stimulation of bone formation. We determined the effect of orally dosed PTX in skeletally mature ovariectomized (OVX) rabbits with osteopenia. The half-maximal effective concentration (EC50) of PTX in rabbit bone marrow stromal cells was 3.07 ±â€¯1.37 nM. The plasma PTX level was 2.05 ±â€¯0.522 nM after a single oral dose of 12.5mg/kg, which was one-sixth of the adult human dose of PTX. Four months of daily oral dosing of PTX at 12.5 mg/kg to osteopenic rabbits completely restored bone mineral density, bone mineral content (BMC), microarchitecture and bone strength to the level of the sham-operated (ovary intact) group. The bone strength to BMC relationship between PTX and sham was similar. The bone restorative effect of PTX was observed in both axial and appendicular bones. In osteopenic rabbits, PTX increased serum amino-terminal propeptide, mineralized nodule formation by stromal cells and osteogenic gene expression in bone. PTX reversed decreased calcium weight percentage and poor crystal packing found in osteopenic rabbits. Furthermore, similar to parathyroid hormone (PTH), PTX had no effect on bone resorption. Taken together, our data show that PTX completely restored bone mass, bone strength and bone mineral properties by an anabolic mechanism. PTX has the potential to become an oral osteogenic drug for the treatment of post-menopausal osteoporosis.


Assuntos
Doenças Ósseas Metabólicas/tratamento farmacológico , Pentoxifilina/administração & dosagem , Pentoxifilina/uso terapêutico , Inibidores de Fosfodiesterase/uso terapêutico , Administração Oral , Animais , Densidade Óssea/efeitos dos fármacos , Doenças Ósseas Metabólicas/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Células Cultivadas , Feminino , Humanos , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/administração & dosagem , Hormônio Paratireóideo/uso terapêutico , Coelhos , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo
2.
Cell Mol Gastroenterol Hepatol ; 5(3): 253-271, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29675451

RESUMO

BACKGROUND & AIMS: Cystic fibrosis (CF) patients and CF mouse models have increased risk for gastrointestinal tumors. CF mice show augmented intestinal proliferation of unknown etiology and an altered intestinal environment. We examined the role of the cystic fibrosis transmembrane conductance regulator (Cftr) in Wnt/ß-catenin signaling, stem cell proliferation, and its functional expression in the active intestinal stem cell (ISC) population. Dysregulation of intracellular pH (pHi) in CF ISCs was investigated for facilitation of Wnt/ß-catenin signaling. METHODS: Crypt epithelia from wild-type (WT) and CF mice were compared ex vivo and in intestinal organoids (enteroids) for proliferation and Wnt/ß-catenin signaling by standard assays. Cftr in ISCs was assessed by immunoblot of sorted Sox9 enhanced green fluorescent protein(EGFP) intestinal epithelia and pHi regulation by confocal microfluorimetry of leucine-rich G-protein-coupled receptor 5 ISCs. Plasma membrane association of the Wnt transducer Dishevelled 2 (Dvl2) was assessed by fluorescence imaging of live enteroids from WT and CF mice crossed with Dvl2-EGFP/ACTB-tdTomato,-EGFP)Luo/J (RosamT/mG) mice. RESULTS: Relative to WT, CF intestinal crypts showed an ∼30% increase in epithelial and Lgr5+ ISC proliferation and increased Wnt/ß-catenin signaling. Cftr was expressed in Sox9EGFPLo ISCs and loss of Cftr induced an alkaline pHi in ISCs. CF crypt-base columnar cells showed a generalized increase in plasma membrane Dvl2-EGFP association as compared with WT. Dvl2-EGFP membrane association was charge- and pH-dependent and increased in WT crypt-base columnar cells by Cftr inhibition. CONCLUSIONS: CF intestine shows increased ISC proliferation and Wnt/ß-catenin signaling. Loss of Cftr increases pHi in ISCs, which stabilizes the plasma membrane association of the Wnt transducer Dvl, likely facilitating Wnt/ß-catenin signaling. Absence of Cftr-dependent suppression of ISC proliferation in the CF intestine may contribute to increased risk for intestinal tumors.

3.
Best Pract Res Clin Endocrinol Metab ; 27(3): 385-402, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23856267

RESUMO

Expression and function of the CaSR have been shown in some mammalian taste buds and basal cells of the esophagus. Signaling cascades responsible for CaSR-mediated stimulation of H(+)-K(+)-ATPase on human parietal cells have been defined. Transgenic mice and reductionistic cell culture models have shown that the CaSR promotes gastrin secretion from G cells, cholecystokinin (CCK) secretion from duodenal I cells and BMP-2 secretion from sub-epithelial myofibroblasts. In addition, the CaSR mediates a novel paracrine relationship between myofibroblasts and overlying epithelial cells in the colon. Thus, CaSR activators stimulate secretion of Wnt5a from myofibroblasts and expression of the Wnt5a receptor Ror2 in epithelial cells. CaSR-mediated Wnt5a/Ror2 engagement stimulates epithelial differentiation and reduces expression of the receptor for tumor necrosis factor (TNFR1). CaSR activators also modulate intestinal motility, inhibit Cl(-) secretion and stimulate Na(+) absorption in both the small intestine and colon. Colonic epithelia from conditional and global CaSR knockout mice exhibit increased proliferation with increased Wnt/ß-catenin signaling, demonstrating that the CaSR negatively modulates colonic epithelial growth.


Assuntos
Colo/metabolismo , Intestino Delgado/metabolismo , Comunicação Parácrina/fisiologia , Receptores de Detecção de Cálcio/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Proliferação de Células , Humanos , Receptores de Detecção de Cálcio/genética , Equilíbrio Hidroeletrolítico/fisiologia
4.
Lab Invest ; 93(5): 520-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23545937

RESUMO

Epidemiological evidence suggests increased dietary calcium and dairy products reduce the onset of colon cancer. To understand a role of the colonic extracellular calcium-sensing receptor (CaSR) in calcium-mediated chemoprevention of colon cancer, we induced formation of aberrant crypt foci (ACF) caused by azoxymethane (AOM) injection in 'rescued' CaSR-/PTH- (C-/P-) double knockout colons compared with colons from control CaSR+/PTH+ (C+/P+) mice. C-/P- colonic epithelia had increased Wnt/ß-catenin signaling as evidenced by 3-8-fold increases in Wnt3a, CyclinD1, and MMP-7 proteins compared with C+/P+ colonic epithelia. The C-/P- colonic epithelia had reduced Wnt5a and Ror2, and a three-fold increase in TNFR1 compared with C+/P+ epithelia. The C-/P- colons and small intestine had extensive neutrophil infiltration with myeloperoxidase (MPO) levels 18-fold higher then C+/P+ small intestine and colon. Saline-injected C-/P- colons had the same number of ACF/cm(2) as C+/P+ colons, which were injected with AOM. However, there were eight times more ACF/cm(2) in the C-/P- injected with AOM compared with C+/P+ colons, which received AOM. Together our results suggest both inflammation and Wnt/ß-catenin signaling are increased in the epithelia of 'rescued' CaSR/PTH double knockout colons, and the capacity for non-canonical Wnt signaling through Wnt5a/Ror2 engagement is reduced. The loss of the colonic CaSR increased the number of ACF/cm(2) in response to AOM injection, suggesting colonic CaSR may mediate the chemoprotective effect of increased dietary calcium against colorectal cancer observed in humans.


Assuntos
Focos de Criptas Aberrantes/metabolismo , Hormônio Paratireóideo/deficiência , Receptores de Detecção de Cálcio/genética , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Focos de Criptas Aberrantes/induzido quimicamente , Focos de Criptas Aberrantes/genética , Animais , Azoximetano , Western Blotting , Colo/química , Colo/metabolismo , Colo/patologia , Ciclina D1/metabolismo , Predisposição Genética para Doença , Intestino Delgado/química , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Metaloproteinase 7 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Hormônio Paratireóideo/genética , Hormônio Paratireóideo/metabolismo , Peroxidase/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Proteína Wnt-5a
5.
Pediatr Res ; 73(4 Pt 1): 414-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23269116

RESUMO

BACKGROUND: Eosinophils reside in normal gastrointestinal tracts and increase during disease states. Receptors for eosinophil-derived granule proteins (EDGPs) have not been identified, but highly cationic molecules, similar to eosinophil proteins, bind extracellular calcium-sensing receptors (CaSRs). We hypothesized that stimulation of CaSRs by eosinophil proteins activates epithelial cells. METHODS: Caco2 intestinal epithelial cells, AML14.3D10 eosinophils, wild-type (WT) human embryonic kidney 293 (HEK293) cells not expressing CaSRs (HEK-WT), and CaSR-transfected HEK293 cells (HEK-CaSR) were stimulated with an eosinophil protein analog poly-L-arginine (PA) and phosphorylated extracellular signal-regulated kinase (pERK)1 and pERK2 were measured. Functional activation was measured with collagen lattice contraction assays. RESULTS: Coculture of Caco2 cells with AML14.3D10 eosinophils augmented lattice contraction as compared with lattices containing Caco2 cells alone. PA stimulation of Caco2 lattices augmented contraction. HEK-CaSR stimulation with PA or Ca(2+) resulted in greater pERK activation than that of stimulated HEK-WT cells. PA stimulated greater HEK-CaSR lattice contraction than unstimulated lattices. Contraction of PA-stimulated and PA-unstimulated HEK-WT lattices did not differ. CONCLUSION: Exposure of intestinal epithelia to the EDGP analog PA stimulates CaSR-dependent ERK phosphorylation and epithelial-mediated collagen lattice contraction. We speculate that EDGP release within the epithelial layers activates the CaSR receptor, leading to matrix contraction and tissue fibrosis.


Assuntos
Colágeno/metabolismo , Proteínas Granulares de Eosinófilos/farmacologia , Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Peptídeos/farmacologia , Receptores de Detecção de Cálcio/agonistas , Células CACO-2 , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibrose , Células HEK293 , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo , Fatores de Tempo , Transfecção
7.
Neuroreport ; 23(14): 846-50, 2012 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-22922600

RESUMO

Dopaminergic neurotransmission is thought to be involved in reward-related incentive learning and addictive behaviour. Amphetamine will alter glycogen synthase kinase-3ß (GSK-3ß) activity by increasing dopamine transporter efflux rates. We investigated the hypothesis that Wnt signalling will be altered in rat nucleus accumbens within 15 min of injection of amphetamine compared with saline. We isolated RNA from the nucleus accumbens and used reverse transcriptase-PCR to screen for altered Wnt expression. We found that amphetamine had no effect on Wnt5a or Wnt7a expression but increased Wnt3. We then measured protein expression of Wnt3, phosphorylated lipoprotein-related peptide 6, GSK-3ß phosphorylated at serine-9 and tyrosine-216 and total ß-catenin. We found that amphetamine increased Wnt3 protein expression, increased pLRP6 (threonine-1572) levels, increased ß-catenin levels, increased GSK-3ß phosphorylation at serine-9, consistent with inhibition of GSK-3ß activity, and diminished GSK-3ß phosphorylation at tyrosine-216. Our data support the hypothesis that proximate Wnt signalling is rapidly activated by amphetamine in the adult rat nucleus accumbens.


Assuntos
Anfetamina/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Proteína Wnt3/biossíntese , Transtornos Relacionados ao Uso de Anfetaminas/metabolismo , Animais , Western Blotting , Núcleo Accumbens/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos
8.
Am J Reprod Immunol ; 68(4): 282-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22626252

RESUMO

PROBLEM: Understanding of uterine natural killer (uNK) cell functions during normal pregnancy remains incomplete. METHOD OF STUDY: Cloud tag analysis of literature was used to document themes addressed experimentally for uNK cells. Immunohistochemistry, including whole-mount staining of early implantation sites, separation of uNK cells into molecularly distinct subsets, and physiologic measurements in normal and mutant mice, are further advancing understanding of uNK cell biology. RESULTS: Literature analyses revealed three key, current uNK cell research themes: angiogenesis, spiral arterial remodeling/pre-eclampsia/hypertension and infertility. UNK cells are being defined as cells potentially regulated by Wnt signaling that are heterogeneous in progenitor source and function and make unique contributions to implantation site development prior to spiral arterial remodeling. CONCLUSION: Future studies are poised to define uNK cell progenitor cells, identify the signaling pathways supporting established uNK cell functions and move current understanding of mouse uNK cells to clinical research questions.


Assuntos
Células Matadoras Naturais/imunologia , Útero/imunologia , Animais , Feminino , Humanos , Camundongos , Gravidez , Transdução de Sinais/imunologia
9.
Front Physiol ; 2: 58, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21949508

RESUMO

Regulation of expression of the intestinal epithelial actin-binding protein, villin, is poorly understood. The aim of this study was to determine whether Wnt5a stimulates Ror2 in intestinal epithelia caused transient increases in phospho-ERK1/2 (pERK1/2) and subsequently increased expression of villin transcript and protein. To demonstrate Wnt5a-Ror2 regulation of villin expression, we overexpressed wild-type, truncated, or mutant Ror2 constructs in HT29 adenocarcinoma cells and non-transformed fetally derived human intestinal epithelial cells, added conditioned media containing Wnt5a and measured changes in ERK1/2 phosphorylation, villin amplicons, and protein expression by RT-PCR and Western blot techniques. Wnt5a addition caused a transient increase in pERK1/2, which was maximal at 10 min but extinguished by 30 min. Transient transfection with a siRNA duplex against Ror2 diminished Ror2 amplicons and protein and reduced the extent of pERK1/2 activation. Structure-function analysis revealed that the deletion of the cysteine-rich, kringle, or tyrosine kinase domain or substitution mutations of tyrosine residues in the intracellular Ser/Thr-1 region of Ror2 prevented the Wnt5a stimulation of pERK1/2. Deletion of the intracellular proline and serine/threonine-rich regions of Ror2 had no effect on Wnt5a stimulation of pERK1/2. The increase in villin expression was blocked by pharmacological inhibition of MEK-1 and casein kinase 1, but not by PKC and p38 inhibitors. Neither Wnt3a nor epidermal growth factor addition caused increases in villin protein. Our findings suggest that Wnt5a/Ror2 signaling can regulate villin expression in the intestine.

10.
Front Physiol ; 2: 17, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21603229

RESUMO

Tumor necrosis factor alpha (TNFα) and its receptor TNFR1 play a central role in the development of colitis-associated colon cancer. To understand a role for the extracellular calcium-sensing receptor (CaSR) and its non-canonical Wnt mediators, Wnt5a/Ror2, we used reductionistic systems. We added lipopolysaccharide (LPS) to mouse peritoneal macrophages, RAW264.7 cells, a murine macrophage cell line, and 18Co colonic myofibroblasts, to stimulate TNFα secretion and then activated endogenous CaSR. CaSR activation inhibited TNFα secretion, which in RAW264.7 cells knockdown of CaSR by short-interfering RNA (siRNA) duplex reversed. LPS-stimulated NFκB promoter activity in RAW264.7 cells was inhibited by CaSR activation with Ca(2+) or other polyvalent CaSR agonists. Reducing CaSR expression with siRNA duplex prevented this inhibition. Following LPS addition to CaSR-HEK cells or RAW264.7 macrophages, CaSR stimulation deneddylated Cullin1. Wnt5a added to HT-29 cells which overexpressed Ror2 or T84 monolayers treated with 3 mM Ca(2+) reduced TNFR1 protein expression ∼70%. TNFα/INFγ addition to high resistance T84 monolayers reduced transepithelial resistance 50% within 4 h. CaSR activation (3 mM Ca(2+)) together with rhWnt5a (200 ng/ml) prevented this reduction while Wnt3a addition had no effect. LPS-stimulated TNFα secretion from RAW264.7 cells was not effected by rhWnt5a but increased 10-fold by Wnt3a. Together our results suggest that following LPS challenge, CaSR activation will inhibit NFκB activity and reduce TNFα secretion from macrophages and stroma while Wnt5a/Ror2 engagement on intestinal epithelia reduces TNFR1 expression, allowing TNFα signaling to be titrated. Our results also suggest that canonical Wnt signaling may enhance TLR4 stimulation of TNFα secretion from murine macrophages.

12.
Am J Physiol Gastrointest Liver Physiol ; 295(4): G748-59, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18703641

RESUMO

To understand whether extracellular calcium-sensing receptor (CaSR) expression on colonic myofibroblast cells (18Co) contributed to epithelial homeostasis, we activated the CaSR with 5 mM Ca(2+), screened by RT-PCR Wnt family members, and measured their secretion. Transcripts for Wnt 1, 2, 2b, 3a, 4, and 7a were either absent or unchanged whereas Wnt3 decreased and Wnt5a increased. We assessed Wnt5a secretion by Western blot. High Ca(2+) (5 mM) substantially increased Wnt5a secretion; small interfering RNA (siRNA) against the CaSR reduced this to constitutive amounts. Expression of Wnt5a plasmid but not Wnt1 or Wnt3a increased caudal homeodomain factor CDX2 transcripts and protein in HT-29 adenocarcinoma cells. Wnt5a increased activity of a sucrase-isomaltase (SI) promoter in Caco-2BBE cells. Wnt5a protein stimulation of CDX2 transcripts and protein and SI reporter were increased by overexpression of wild-type Ror2, a Wnt5a receptor, and reduced with siRNA against Ror2. CaSR activation of HT-29 cells increased Ror2 protein expression. Ror2 protein was expressed in mouse jejunum from crypt base to villus tip and in the colon on surface epithelia. Our results show that activation of a G protein-coupled receptor, the CaSR, stimulates secretion of Wnt5a from myofibroblasts. Stimulation of epithelia by the CaSR increased the expression of a receptor for Wnt5a, the tyrosine kinase Ror2, suggesting existence of a unique paracrine relationship for CDX2 homoeostasis in the intestine and revealing new contributions of CaSR-activated myofibroblasts to intestinal stem cell niche microenvironments.


Assuntos
Proteínas de Homeodomínio/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Detecção de Cálcio/fisiologia , Receptores de Superfície Celular/fisiologia , Complexo Sacarase-Isomaltase/fisiologia , Proteínas Wnt/metabolismo , Animais , Fator de Transcrição CDX2 , Células CACO-2 , Colo/citologia , Fibroblastos/fisiologia , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase , Regulação para Cima , Proteína Wnt-5a , beta Catenina/fisiologia
13.
J Mol Biol ; 380(2): 313-26, 2008 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-18534616

RESUMO

Intrinsically disordered proteins are emerging as substantial functional constituents of mammalian proteomes. Although the abundance of these proteins has been established by bioinformatics approaches, the vast majority have not been characterized structurally or functionally. The C/EBP homologous protein (CHOP) is a proto-oncogene, traditionally shown as a dominant-negative inhibitor of C/EBPs and a transcriptional activator of activating protein-1. We report here the in vitro characterization of CHOP, where our computational analyses and experimental evidences show for the first time that CHOP is an intrinsically disordered protein. Intrinsic fluorescence, NMR spectroscopy, and analytical size-exclusion chromatography studies indicate that CHOP contains extensive disordered regions and self-associate in solution. Interestingly, the disordered N-terminal region has a key role in the oligomerization of CHOP and is vital for its biological activity. We report a novel mechanistic role of CHOP in the inhibition of Wnt/TCF signaling and stimulation of c-Jun and sucrase-isomaltase reporter activity in intestinal colon cancer cells. These findings are discussed in the context of oligomerization of intrinsically disordered proteins as one of the mechanisms through which they exert their biological function.


Assuntos
Cálcio/metabolismo , Neoplasias do Colo/metabolismo , Fator de Transcrição CHOP/química , Fator de Transcrição CHOP/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Genes Reporter , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Regiões Promotoras Genéticas , Conformação Proteica , Proto-Oncogene Mas , Interferência de RNA , Receptores de Detecção de Cálcio/genética , Receptores de Detecção de Cálcio/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Complexo Sacarase-Isomaltase/genética , Complexo Sacarase-Isomaltase/metabolismo , Fator de Transcrição CHOP/genética , beta Catenina/genética , beta Catenina/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 294(1): G120-9, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17962359

RESUMO

Gastrointestinal reflux disease and eosinophilic esophagitis are characterized by basal cell hyperplasia. The extracellular calcium-sensing receptor (CaSR), a G protein-coupled receptor, which may be activated by divalent agonists, is expressed throughout the gastrointestinal system. The CaSR may regulate proliferation or differentiation, depending on cell type and tissue. The current experiments demonstrate the expression of the CaSR on a human esophageal epithelial cell line (HET-1A) and the location and expression of the CaSR in the human esophagus. CaSR immunoreactivity was seen in the basal layer of normal human esophagus. CaSR expression was confirmed in HET-1A cells by RT-PCR, immunocytochemistry, and Western blot analysis. CaSR stimulation by extracellular calcium or agonists, such as spermine or Mg(2+), caused ERK1 and 2 activation, intracellular calcium concentration ([Ca(2+)](i)) mobilization (as assessed by microspecfluorometry using Fluo-4), and secretion of the multifunctional cytokine IL-8 (CX-CL8). HET-1A cells transiently transfected with small interfering (si)RNA duplex against the CaSR manifested attenuated responses to Ca(2+) stimulation of phospho- (p)ERK1 and 2, [Ca(2+)](i) mobilization, and IL-8 secretion, whereas responses to acetylcholine (ACh) remained sustained. An inhibitor of phosphatidylinositol-specific phospholipase C (PI-PLC) (U73122) blocked CaSR-stimulated [Ca(2+)](i) release. We conclude that the CaSR is present on basal cells of the human esophagus and is present in a functional manner on the esophageal epithelial cell line, HET-1A.


Assuntos
Sinalização do Cálcio , Células Epiteliais/metabolismo , Esôfago/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Acetilcolina/farmacologia , Western Blotting , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Ativação Enzimática , Células Epiteliais/efeitos dos fármacos , Esôfago/citologia , Esôfago/efeitos dos fármacos , Estrenos/farmacologia , Humanos , Imuno-Histoquímica , Interleucina-8/metabolismo , Magnésio/metabolismo , Microespectrofotometria , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Fosforilação , Reação em Cadeia da Polimerase , Pirrolidinonas/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Receptores de Detecção de Cálcio/genética , Espermina/metabolismo , Transfecção , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
15.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G403-11, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17463182

RESUMO

To understand the role of the colonic extracellular calcium-sensing receptor (CaSR) in calcium chemoprotection against colon cancer, we activated the CaSR with 5 mM Ca(2+) on HT-29 cells, an adenocarcinoma cell line. High Ca(2+) stimulated the upregulation (as assessed by RT-PCR) and the secretion of Wnt5a (assessed by Western blot), a noncanonical Wnt family member. Inhibiting CaSR activity with a short interfering RNA (siRNA) duplex against the CaSR reduced CaSR protein and prevented the secretion of Wnt5a. Dominant negative CaSR (R185Q) or siRNA blocked the high Ca(2+)-mediated inhibition of the beta-catenin reporter TOPflash. The CaSR/Wnt5a inhibition of beta-catenin reporter was prevented by dominant negative ubiquitin ligase seven in absentia homolog 2 (Siah2). In low-calcium medium, overexpressing Wnt5a increased Siah2 amplicons and protein. Inducing the expression of full-length adenomatous polyposis coli (APC) prevented CaSRmediated increases of Siah2 and Wnt5a. Overexpressing the receptor tyrosine kinase-like orphan receptor 2 (Ror2) increased Wnt5a and CaSR-mediated inhibition of TOPflash. Conditioned medium from Wnt5a-transfected cells added to HT-29 cells in low-Ca(2+) medium inhibited the beta-catenin reporter. This inhibition was blocked dose responsively by Frizzled-8/Fc chimeric antibody. Overexpression of Ror2 in HT-29 cells in low-Ca(2+) medium increased the inhibition of beta-catenin reporter caused by recombinant Wnt5a protein compared with addition of Wnt5a protein alone. Our findings demonstrate that APC status plays a key role as a determinant of Wnt5a secretion and suggest that CaSR-mediated secretion of Wnt5a will inhibit defective Wnt signaling in APC-truncated cells in an autocrine manner.


Assuntos
Neoplasias do Colo/fisiopatologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Detecção de Cálcio/fisiologia , Proteínas Wnt/metabolismo , Adenocarcinoma , Proteína da Polipose Adenomatosa do Colo/biossíntese , Cálcio/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , Células Cultivadas , Humanos , Proteínas Nucleares/biossíntese , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase , Receptores de Superfície Celular/biossíntese , Ubiquitina-Proteína Ligases/biossíntese , Proteína Wnt-5a , beta Catenina/antagonistas & inibidores
16.
Am J Physiol Gastrointest Liver Physiol ; 292(3): G753-66, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17138967

RESUMO

To understand whether postprandial extracellular Ca(2+) (Ca(o)(2+)) changes were related to intestinal epithelial homeostasis, we performed array analysis on extracellular calcium-sensing receptor (CaSR)-expressing colonic myofibroblasts (18Co cells) and observed increases in bone morphogenetic protein (BMP)-2 transcripts. The present experiments demonstrated that regulated secretion of BMP-2 occurs in response to CaSR activation of these cells and revealed a new property of BMP-2 on the intestinal barrier. Activation by Ca(o)(2+), spermine, GdCl(3), or neomycin sulfate of 18Co cells or primary isolates of myofibroblasts from the normal human colon stimulated both the synthesis (RT-PCR) and secretion (ELISA) of BMP-2. Transient transfection with short interfering RNA against CaSR completely inhibited BMP-2 secretion. Transient transfection with dominant negative CaSR (R185Q) increased the EC(50) of Ca(o)(2+) (5.7 vs. 2.3 mM). Upregulation of BMP-2 transcript and secretion occurring within 3 h of CaSR activation was prevented by actinomycin D. CaSR-mediated BMP-2 synthesis and secretion required phosphatidylinositol 3-kinase activation (as assessed by phospho-Akt generation). Exogenous BMP-2 and conditioned medium from CaSR-stimulated 18Co cells accelerated restitution in wounded postconfluent Caco-2 cells. Exogenous BMP-2 and conditioned medium from CaSR-stimulated 18Co cells increased the transepithelial resistance of low- and high-resistance T-84 epithelial monolayers. CaSR stimulation of T-84 epithelia and colonic myofibroblasts downregulated the BMP family antagonist Noggin, as assessed by RT-PCR and Western blot analysis. Together, our data suggest that the CaSR mediates the effective concentration of BMP-2 in the intestine, which leads to enhanced repair and barrier development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Fibroblastos/metabolismo , Receptores de Detecção de Cálcio/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Células CACO-2 , Cálcio/agonistas , Cálcio/farmacologia , Proteínas de Transporte/genética , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Colo/citologia , Meios de Cultivo Condicionados/farmacologia , Impedância Elétrica , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/fisiologia , Cinética , Neomicina/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores de Detecção de Cálcio/agonistas , Fator de Crescimento Transformador beta/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
17.
Bone ; 35(3): 664-72, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15336602

RESUMO

We have previously reported that high extracellular Ca2+ stimulates parathyroid hormone-related protein (PTHrP) release from human prostate and breast cancer cell lines as well as from H-500 rat Leydig cancer cells, an action mediated by the calcium-sensing receptor (CaR). Activating the CaR leads to phosphorylation of mitogen-activated protein kinases (MAPKs) that participate in PTHrP synthesis and secretion. Because the CaR is a G protein-coupled receptor (GPCR), it is likely to transactivate the epidermal growth factor receptor (EGFR) or the platelet-derived growth factor receptor (PDGFR). In this study, we hypothesized that activation of the CaR transactivates the EGFR or PDGFR, and examined whether transactivation affects PTHrP secretion in PC-3 human prostate cancer cells. Using Western analysis, we observed that an increase in extracellular Ca2+ resulted in delayed activation of extracellular signal-regulated kinase (ERK) in PC-3 cells. Pre-incubation with AG1478 (an EGFR kinase inhibitor) or an EGFR neutralizing antibody inhibited the high Ca2+ -induced phosphorylation of ERK1/2. GM6001, a pan matrix metalloproteinase (MMP) inhibitor, also partially suppressed the ERK activation, but AG1296 (a PDGFR kinase inhibitor) did not. High extracellular Ca2+ stimulates PTHrP release during a 6-h incubation (1.5- to 2.5- and 3- to 4-fold increases in 3.0 and 7.5 mM Ca2+, respectively). When cells were preincubated with AG1478, GM6001, or an antihuman heparin-binding EGF (HB-EGF) antibody, PTHrP secretion was significantly inhibited under basal as well as high Ca2+ conditions, while AG1296 had no effect on PTHrP secretion. Taken together, these findings indicate that activation of the CaR transactivates the EGFR, but not the PDGFR, leading to phosphorylation of ERK1/2 and resultant PTHrP secretion, although CaR-EGFR-ERK might not be the only signaling pathway for PTHrP secretion. This transactivation is most likely mediated by activation of MMP and cleavage of proheparin-binding EGF (proHB-EGF) to HB-EGF.


Assuntos
Receptores ErbB/fisiologia , Proteína Relacionada ao Hormônio Paratireóideo/metabolismo , Neoplasias da Próstata/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Ativação Transcricional/fisiologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Masculino , Proteína Relacionada ao Hormônio Paratireóideo/genética , Neoplasias da Próstata/genética , Receptores de Detecção de Cálcio/genética
18.
Biochem Biophys Res Commun ; 320(2): 455-60, 2004 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-15219850

RESUMO

Activation of the extracellular calcium-sensing receptor (CaR) stimulates mitogen-activated protein kinases to upregulate the synthesis and secretion of parathyroid hormone related peptide (PTHrP) from cells expressing the CaR heterologously or endogenously. The current experiments demonstrate that this occurs because CaR activation "transactivates" the EGF receptor (EGFR). Time dependent increases in tyrosine phosphorylation of the EGFR after addition of extracellular calcium ([Ca2+]o, 3 mM) occurred in stably CaR-transfected HEK293 cells but not in non-transfected HEK293 cells. AG1478, an EGFR kinase inhibitor, prevented the CaR-mediated increases of pERK and PTHrP release, while AG1296, a PDGFR kinase inhibitor, had no effect. Inhibitors of matrix metalloproteinase and heparin bound-EGF prevented the CaR-mediated increases of pERK and PTHrP, consistent with a "triple-membrane-spanning signaling" requirement for transactivation of the EGFR by the CaR. Proximal and distal signal transduction cascades activated by the CaR may reflect transactivation of the EGFR by the extracellular calcium-sensing receptor.


Assuntos
Receptores ErbB/genética , Receptores de Detecção de Cálcio/fisiologia , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Humanos , Fosforilação , Quinazolinas , Tirosina/metabolismo , Tirfostinas/farmacologia
19.
J Neurosci Res ; 75(4): 491-8, 2004 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-14743432

RESUMO

By using pharmacological and molecular approaches, we previously showed that the G-protein-coupled, extracellular calcium (Ca2+(o))-sensing receptor (CaR) regulates a large-conductance (approximately 140 pS), Ca(2+)-activated K+ channel [IK(Ca); CAKC] in U87 astrocytoma cells. Here we show that elevated Ca2+(o) stimulates extracellular-signal-regulated kinase (ERK1/2) and p38 MAP kinase (MAPK). The effect of high Ca2+(o) on p38 MAPK but not ERK1/2 is CaR mediated, insofar as transduction with a dominant-negative CaR (R185Q) using recombinant adeno-associated virus (rAAV) attenuated the activation of p38 MAPK but not of ERK1/2. p38 MAPK activation by the CaR is likely to be protein kinase C (PKC) independent, in that the pan-PKC inhibitor GF109203X failed to abolish the high-Ca2+(o)-induced phosphorylation of p38 MAPK. Consistently with our data on the activation of this kinase, we observed that inhibiting p38 MAPK blocked the activation of the CAKC induced by the specific pharmacological CaR activator NPS R-467. In contrast, inhibiting MEK1 only transiently inhibited the activation of this K+ channel by NPS R-467, despite the continued presence of the antagonist. Similarly to the lack of any effect of the PKC inhibitor on the activation of ERK1/2 and p38 MAPK, inhibiting PKC had no effect on NPS R-467-induced activation of this channel. Therefore, our data show that the CaR, acting via p38 MAPK, regulates a large-conductance CAKC in U87 cells, a process that is PKC independent. Large-conductance CAKCs play an important role in the regulation of cellular volume, so our results have important implications for glioma cell volume regulation.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Canais de Potássio Cálcio-Ativados/fisiologia , Receptores de Detecção de Cálcio/metabolismo , Cálcio/farmacologia , Linhagem Celular Tumoral , Humanos , Canais de Potássio Cálcio-Ativados/agonistas , Receptores de Detecção de Cálcio/agonistas , Proteínas Quinases p38 Ativadas por Mitógeno
20.
Am J Physiol Endocrinol Metab ; 284(2): E435-42, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12388158

RESUMO

Increases in extracellular calcium concentration ([Ca(2+)](o)) stimulate from normal and malignant cells secretion of parathroid hormone-related protein (PTHrP), a major mediator of humoral hypercalcemia of malignancy. Because the calcium-sensing receptor (CaR) is a determinant of calcium-regulated hormone secretion, we examined whether HEK cells stably transfected with human CaR secreted PTHrP in response to CaR stimulation. Increases in [Ca(2+)](o) or neomycin and Gd(3+) all substantially increased PTHrP secretion in CaR-HEK cells but had no effect on nontransfected cells. CaR activation likewise increased PTHrP transcripts. PD-098059 and U-0126, inhibitors of the mitogen-activated protein kinase kinase MEK1/2, abolished CaR-stimulated secretion but had no effect on basal secretion. An inhibitor of p38 MAP kinase, SB-203580, also attenuated CaR-stimulated secretion. Western analysis revealed that CaR activation caused a robust increase in MEK1/2 and p38 MAP kinase phosphorylation. A Src family kinase inhibitor, PP2, blocked both basal and CaR-stimulated secretion. We conclude that CaR specifically mediates the effect of increasing [Ca(2+)](o) on PTHrP synthesis and secretion and that activated MEK1/2 and p38 MAP kinases are determinants of the CaR's stimulation of PTHrP secretion.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Hormônios Peptídicos/metabolismo , Receptores de Superfície Celular/metabolismo , Difosfato de Adenosina/farmacologia , Compostos de Anilina/farmacologia , Anisomicina/farmacologia , Cálcio/agonistas , Cálcio/farmacologia , Cátions/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Rim/citologia , MAP Quinase Quinase 1 , MAP Quinase Quinase 2 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteína Relacionada ao Hormônio Paratireóideo , Hormônios Peptídicos/genética , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno , Quinases da Família src/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...