Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
J Comp Neurol ; 527(18): 2931-2947, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31132148

RESUMO

Neural stem cells in the subventricular zone (SVZ) of the lateral ventricle generate new interneurons, which migrate tangentially through the rostral migratory stream (RMS) to the olfactory bulb (OB). The PROK2 (prokineticin 2) and PROKR2 (prokineticin receptor 2) signaling pathway has been identified to cause human Kallmann syndrome, a developmental disease that associates hypogonadism with anosmia (OB developmental defects). However, the identities and properties of PROK2+ and PROKR2+ cells in the SVZ-RMS-OB remain largely unknown. Here we examine the expression patterns of Prok2 and Prokr2 in the SVZ-RMS-OB using Prok2EGFP transgenic and Prokr2LacZ/+ knockin mice. Our results show that Prokr2 is expressed in postmitotic immature interneurons in the SVZ-RMS-OB. Prok2 is not expressed in the SVZ, but a few PROK2+ cells are found in the medial part of the RMS; they are not neural progenitors or migrating neuroblasts. In the OB, Prok2 is expressed in a subset of granule cells and tufted cells, but no coexpression of Prok2 and Prokr2 in the OB cells is observed. In Prok2 and Prokr2 mutant mice, severe tangential and radial migration defects of neuroblasts in the SVZ-RMS-OB result in loss of ~75% of GABAergic interneurons in the OB. These analyses demonstrate that PROK2/PROKR2 signaling is crucial for the tangential and radial migration of OB interneurons.


Assuntos
Movimento Celular/fisiologia , Hormônios Gastrointestinais/biossíntese , Interneurônios/metabolismo , Células-Tronco Neurais/metabolismo , Neuropeptídeos/biossíntese , Bulbo Olfatório/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Peptídeos/biossíntese , Animais , Hormônios Gastrointestinais/genética , Interneurônios/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais/química , Neuropeptídeos/genética , Bulbo Olfatório/química , Bulbo Olfatório/citologia , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Transdução de Sinais/fisiologia
2.
Medicine (Baltimore) ; 97(48): e13167, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30508896

RESUMO

This study aims to investigate the gastrointestinal functions of patients with liver failure (LF) based on gastrointestinal dysfunction (GD) scores and serum gastrointestinal hormone levels.The GD in LF patients was scored using the gastrointestinal dysfunction scoring criteria. Serum gastrin (GAS), cholecystokinin (CCK), and motilin (MTL) levels were determined in LF patients. In addition, liver function and prothrombin activity were detected, and ultrasonography was performed.The GD score was significantly higher in the LF groups than in the control group. Compared with the control group, serum GAS, CCK, and MTL levels significantly increased in the LF groups, and was positively correlated with the severity of LF. Furthermore, in the LF groups, GD was positively correlated with the severity of LF. However, the GD score and serum GAS, CCK, and MTL levels in the acute LF group were not statistically different, when compared with those in the subacute LF group, acute-on-chronic LF group and chronic LF group.LF plays a key role in the development of GD, and may be the main cause of obvious gastrointestinal symptoms, such as abdominal distension, nausea, vomiting and anorexia, in LF patients. The severity of GD is not associated with LF type, but is positively correlated with the severity of LF, suggesting that GD in LF patients may have complicated mechanisms.


Assuntos
Doença Hepática Terminal/epidemiologia , Gastroenteropatias/epidemiologia , Hormônios Gastrointestinais/sangue , Adulto , Idoso , Colecistocinina/sangue , Doença Hepática Terminal/sangue , Feminino , Gastrinas/sangue , Gastroenteropatias/sangue , Hormônios Gastrointestinais/biossíntese , Humanos , Testes de Função Hepática , Masculino , Pessoa de Meia-Idade , Motilina/sangue , Protrombina/biossíntese , Índice de Gravidade de Doença
3.
Peptides ; 101: 32-43, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29289697

RESUMO

AIM: To determine whether intestinal expression of guanylate cyclase activator 2A (GUCA2A) and guanylate cyclase activator 2B (GUCA2B) genes is regulated in obese humans following Roux-en-Y gastric bypass (RYGB), and to evaluate the corresponding guanylin (GN) and uroguanylin (UGN) peptides for potentially contributing to the beneficial metabolic effects of RYGB. METHODS: Enteroendocrine cells were harvested peri- and post-RYGB, and GUCA2A/GUCA2B mRNA expression was compared. GN, UGN and their prohormones (proGN, proUGN) were administered subcutaneously in normal-weight mice to evaluate effects on food intake and glucose regulation. The effect of pro-UGN or UGN overexpression, using adeno-associated virus (AAV) vectors, was assessed in diet-induced obese (DIO) mice. Intracerebroventricular administration of GN and UGN was performed in rats for assessment of putative centrally mediated effects on food intake. GN and UGN, as well as their prohormones, were evaluated for effects on glucose-stimulated insulin secretion (GSIS) in rat pancreatic islets and perfused rat pancreas. RESULTS: GUCA2A and GUCA2B mRNA expression was significantly upregulated in enteroendocrine cells after RYGB. Peripheral administration of guanylins or prohormones did not influence food intake, oral glucose tolerance, and GSIS. Central administration of GN and UGN did not affect food intake in rats. Chronic AVV-mediated overexpression of UGN and proUGN had no effect on body weight or glucose homeostasis in DIO mice. CONCLUSION: GN and UGN, as well as their prohormones, do not seem to play a significant role in body weight regulation and glycemic control, suggesting that guanylin-family peptides do not show promise as targets for the treatment of obesity or diabetes.


Assuntos
Manutenção do Peso Corporal , Células Enteroendócrinas/metabolismo , Derivação Gástrica , Hormônios Gastrointestinais/biossíntese , Regulação da Expressão Gênica , Peptídeos Natriuréticos/biossíntese , Adulto , Animais , Diabetes Mellitus/metabolismo , Diabetes Mellitus/cirurgia , Feminino , Proteínas Ativadoras de Guanilato Ciclase/biossíntese , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Obesidade/metabolismo , Obesidade/cirurgia
4.
Peptides ; 99: 14-19, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29097254

RESUMO

Recently we found that guanylin (Gn) and its receptor, guanylyl cyclase C (GC-C), are uniquely expressed in the mesenteric macrophages of some diet-resistant rats and that double-transgenic (dTg) rats overexpressing Gn and GC-C in macrophages demonstrate reduced fatty acid synthase and fat accumulation in fat tissue even when fed a high-fat diet (HFD). Lipid accumulation and fatty acid synthase mRNA levels in cocultured dTg rat adipocytes and macrophages were reduced compared with those in adipocytes cultured with WT rat macrophages. Here, we investigated whether Interleukin-15 (IL-15) derived from Gn-GC-C-expressing macrophages regulates lipid accumulation in adipocytes. IL-15 inhibited fatty acid synthase and lipid accumulation via STAT5 in cultured adipocytes. IL-15 mRNA and protein levels in the mesenteric fat of HFD-fed dTg rats were significantly higher than those of HFD-fed WT rats. Phosphorylated STAT5 levels in the mesenteric fat of HFD-fed dTg rats were increased compared with those of HFD-fed WT rats. In addition, the mRNA level of fatty acid synthase in the mesenteric fat was lower in HFD-fed dTg rats than in HFD-fed WT rats. These results support the hypothesis that IL-15 secreted from Gn-GC-C-expressing macrophages contributes to the inhibition of fatty acid synthase and lipid accumulation in adipocytes, leading to obesity resistance.


Assuntos
Adipócitos/metabolismo , Ácido Graxo Sintase Tipo I/metabolismo , Hormônios Gastrointestinais/biossíntese , Interleucina-15/biossíntese , Macrófagos/metabolismo , Peptídeos Natriuréticos/biossíntese , Receptores de Enterotoxina/biossíntese , Adipócitos/citologia , Animais , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Ácido Graxo Sintase Tipo I/genética , Hormônios Gastrointestinais/genética , Interleucina-15/genética , Macrófagos/citologia , Peptídeos Natriuréticos/genética , Ratos , Ratos Transgênicos , Receptores de Enterotoxina/genética
5.
Histochem Cell Biol ; 146(4): 445-55, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27246004

RESUMO

Guanylin (GN) and uroguanylin (UGN), through activation of guanylyl cyclase C (GCC), serve to control intestinal fluid homeostasis. Both peptides are produced in the intestinal epithelium, but their cellular origin has not been fully charted. Using quantitative PCR and an improved in situ hybridization technique (RNAscope), we have assessed the expression of GN (Guca2a), UGN (Guca2b), and GCC (Gucy2c) in mouse intestine. In the crypts of Lieberkühn, expression of Guca2a and Guca2b was restricted to cells of secretory lineage, at the crypt's base, and to a region above, previously identified as a common origin of cellular differentiation. In this compartment, comparatively uniform levels of Guca2a and Guca2b expression were observed throughout the length of the gut. In contrast, Guca2a and Guca2b expression in the villus-surface region was more variable, and reflected the distinct, but overlapping expression pattern observed previously. Accordingly, in jejunum and ileum, Guca2a and Guca2b were abundantly expressed by enterocytes, whereas in colon only Guca2a transcript was found in the surface region. In duodenum, only low levels of Guca2b transcript were observed in columnar cells, and Guca2a expression was restricted entirely to cells of the secretory lineage. Gucy2c was shown to be expressed relatively uniformly along the rostrocaudal and crypt-villus axes and was also found in the duodenal glands. Our study reveals novel aspects of the cellular localization of the GCC signaling axis that, apart from its role in the regulation of fluid balance, link it to pH regulation, cell cycle control, and host defense.


Assuntos
Linhagem da Célula , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Hormônios Gastrointestinais/biossíntese , Intestinos/citologia , Peptídeos Natriuréticos/biossíntese , Animais , Hormônios Gastrointestinais/análise , Hormônios Gastrointestinais/genética , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos , Peptídeos Natriuréticos/análise , Peptídeos Natriuréticos/genética , Transdução de Sinais
6.
Neuropharmacology ; 108: 39-48, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27140692

RESUMO

Bv8/prokineticin 2 (PK2) is a member of a bioactive family of peptides that regulate multiple functions in the CNS including hyperalgesia, neurogenesis, neuronal survival and inflammation. Recent studies have associated PK2 and prokineticin receptors (PKR) with human diseases, but because their role in neuropathology is still debated we examined whether prokineticins exert a protective or deleterious role in models of cerebral ischemia and ischemic tolerance in vitro. In order to mimic cerebral ischemia, we exposed primary murine cortical cell cultures or rat organotypic hippocampal slices to appropriate periods of oxygen-glucose deprivation (OGD), which leads to neuronal damage 24 h later. Ischemic tolerance was induced by exposing hippocampal slices to a preconditioning subtoxic pharmacological stimulus (3 µM NMDA for 1 h) 24 h before the exposure to OGD. Bv8 (10-100 nM) attenuated OGD injury in cortical cultures and hippocampal slices, and the effect was prevented by the PKR antagonist PC7. The development of OGD tolerance was associated with an increase in the expression of PK2, PKR1 and PKR2 mRNA and proteins and was prevented by addition of the antagonist PC7 into the medium during preconditioning. Both Bv8 at protective concentrations and the NMDA preconditioning stimulus promoted the phosphorylation of ERK1/2 and Akt. These findings indicate that the prokineticin system can be up-regulated by a defensive preconditioning subtoxic NMDA stimulus and that PK2 may act as an endogenous neuroprotective factor through the activation of the ERK1/2 and Akt transduction pathways.


Assuntos
Isquemia Encefálica/metabolismo , Córtex Cerebral/metabolismo , Hormônios Gastrointestinais/biossíntese , Hipocampo/metabolismo , Neuropeptídeos/biossíntese , Fármacos Neuroprotetores/metabolismo , Animais , Anuros , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Isquemia Encefálica/prevenção & controle , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hormônios Gastrointestinais/farmacologia , Hormônios Gastrointestinais/uso terapêutico , Hipocampo/efeitos dos fármacos , Camundongos , Neuropeptídeos/farmacologia , Neuropeptídeos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Técnicas de Cultura de Órgãos , Ratos
7.
PLoS One ; 10(8): e0136473, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26313571

RESUMO

Specific gene expression in oocytes and its surrounding cumulus (CC) and granulosa (GC) cells is needed for successful folliculogenesis and oocyte maturation. The aim of the present study was to compare genome-wide gene expression and biological functions of human GC and CC. Individual GC and CC were derived from 37 women undergoing IVF procedures. Gene expression analysis was performed using microarrays, followed by a meta-analysis. Results were validated using quantitative real-time PCR. There were 6029 differentially expressed genes (q < 10-4); of which 650 genes had a log2 FC ≥ 2. After the meta-analysis there were 3156 genes differentially expressed. Among these there were genes that have previously not been reported in human somatic follicular cells, like prokineticin 2 (PROK2), higher expressed in GC, and pregnancy up-regulated nonubiquitous CaM kinase (PNCK), higher expressed in CC. Pathways like inflammatory response and angiogenesis were enriched in GC, whereas in CC, cell differentiation and multicellular organismal development were among enriched pathways. In conclusion, transcriptomes of GC and CC as well as biological functions, are distinctive for each cell subpopulation. By describing novel genes like PROK2 and PNCK, expressed in GC and CC, we upgraded the existing data on human follicular biology.


Assuntos
Proteína Quinase Tipo 1 Dependente de Cálcio-Calmodulina/biossíntese , Células do Cúmulo/metabolismo , Hormônios Gastrointestinais/biossíntese , Neovascularização Fisiológica/fisiologia , Neuropeptídeos/biossíntese , Gravidez/metabolismo , Transcriptoma/fisiologia , Adulto , Diferenciação Celular/fisiologia , Células do Cúmulo/citologia , Feminino , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real
8.
Peptides ; 70: 23-31, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26022984

RESUMO

The impact of rapid weight gain on glucose metabolism during the early postnatal period remains unclear. We investigated the influence of rapid weight gain under different nutritional conditions on glucose metabolism, focusing on the production of pancreatic and gastric peptides. On postnatal day (PND) 2, C57BL/6N pups were divided into three groups: control (C) pups whose dams were fed a control diet (10%kcal fat) and nursed 10 pups each; maternal high-fat diet (HFD) pups whose dams were fed an HFD (45%kcal fat) and nursed 10 pups each; and overfeeding (OF) pups whose dams were fed the control diet and nursed 4 pups each. Data were collected on PND 7, 14 and 21. The body weight gains of the HFD and OF pups were 1.2 times higher than that of the C pups. On PND 14, the HFD pups had higher blood glucose levels, but there were no significant differences in serum insulin levels between the HFD and C pups. The OF pups had higher blood glucose and serum insulin levels than that of the C pups. Insulin resistance was found in the HFD and OF pups. On PND 14, the content of incretins in the jejunum was increased in the OF pups, and acyl ghrelin in the stomach was upregulated in the HFD and OF pups. These results suggest that neonatal weight gain induced by overfeeding pups and maternal high-fat diet during the early postnatal period modulates the insulin sensitivity and the production of pancreatic and gastrointestinal peptides.


Assuntos
Dieta Hiperlipídica , Hormônios Gastrointestinais/biossíntese , Glucose/metabolismo , Insulina/biossíntese , Lactação , Aumento de Peso/fisiologia , Animais , Glicemia , Feminino , Grelina/biossíntese , Resistência à Insulina , Camundongos , Camundongos Endogâmicos C57BL
9.
Oncotarget ; 6(8): 6053-61, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25788276

RESUMO

Hematogenous metastasis, mainly hepatic metastasis, is a frequent metastatic mode in colorectal cancer involving angiogenic growth factors. Two angiogenic growth factors, in particular, Vascular endothelial growth factor (VEGF) and Prokineticin1(PROK1), are considered to have an important role in hematogenous metastasis of colorectal cancer. Accordingly, we report our findings on the importance of the anti-tumor efffect by inhibiting these two factors in human colorectal cancer.When the culture fluid of Colorectal cancer cell lines(DLD-1, HCT116, and LoVo) with high levels of VEGF/PROK1 expression was injected subcutaneously into mice, the culture fluid increased subcutaneous angiogenesis. But when both anti-PROK1 and anti-VEGF antibodies were present in the culture fluid, the length and size of the blood vessels were reduced compared with those seen in the fluid-only, anti-PROK1, and anti-VEGF controls. Also, tumor masses were produced in mice by subcutaneously embedding colorectal cancer cells with high levels VEGF/PROK1 expression. When both anti-PROK1 and anti-VEGF antibodies were simultaneously applied, tumor formation and peritumoral angiogenesis were strongly suppressed, compared with when either anti-PROK1 antibody or anti-VEGF antibody was applied alone.Simultaneous targeting of both angiogenic growth factors (VEGF/PROK1) may prove more useful in colorectal cancer.


Assuntos
Anticorpos/farmacologia , Neoplasias Colorretais/terapia , Hormônios Gastrointestinais/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/imunologia , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/patologia , Feminino , Hormônios Gastrointestinais/biossíntese , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular/métodos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , Molécula-1 de Adesão Celular Endotelial a Plaquetas/imunologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Biomed Res Int ; 2015: 301292, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25685780

RESUMO

The new chemokine Prokineticin 2 (PROK2) and its receptors (PKR1 and PKR2) have a role in inflammatory pain and immunomodulation. Here we identified PROK2 as a critical mediator of neuropathic pain in the chronic constriction injury (CCI) of the sciatic nerve in mice and demonstrated that blocking the prokineticin receptors with two PKR1-preferring antagonists (PC1 and PC7) reduces pain and nerve damage. PROK2 mRNA expression was upregulated in the injured nerve since day 3 post injury (dpi) and in the ipsilateral DRG since 6 dpi. PROK2 protein overexpression was evident in Schwann Cells, infiltrating macrophages and axons in the peripheral nerve and in the neuronal bodies and some satellite cells in the DRG. Therapeutic treatment of neuropathic mice with the PKR-antagonist, PC1, impaired the PROK2 upregulation and signalling. This fact, besides alleviating pain, brought down the burden of proinflammatory cytokines in the damaged nerve and prompted an anti-inflammatory repair program. Such a treatment also reduced intraneural oedema and axon degeneration as demonstrated by the physiological skin innervation and thickness conserved in CCI-PC1 mice. These findings suggest that PROK2 plays a crucial role in neuropathic pain and might represent a novel target of treatment for this disease.


Assuntos
Hormônios Gastrointestinais/biossíntese , Neuralgia/metabolismo , Neuropeptídeos/biossíntese , Células de Schwann/metabolismo , Nervo Isquiático/metabolismo , Transdução de Sinais , Regulação para Cima , Animais , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Neuralgia/patologia , Receptores Acoplados a Proteínas G/metabolismo , Células de Schwann/patologia , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Subtilisinas/metabolismo
11.
PLoS One ; 9(9): e107531, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25216051

RESUMO

INTRODUCTION: Intestinal chemosensory receptors and transporters are able to detect food-derived molecules and are involved in the modulation of gut hormone release. Gut hormones play an important role in the regulation of food intake and the control of gastrointestinal functioning. This mechanism is often referred to as "nutrient sensing". Knowledge of the distribution of chemosensors along the intestinal tract is important to gain insight in nutrient detection and sensing, both pivotal processes for the regulation of food intake. However, most knowledge is derived from rodents, whereas studies in man and pig are limited, and cross-species comparisons are lacking. AIM: To characterize and compare intestinal expression patterns of genes related to nutrient sensing in mice, pigs and humans. METHODS: Mucosal biopsy samples taken at six locations in human intestine (n = 40) were analyzed by qPCR. Intestinal scrapings from 14 locations in pigs (n = 6) and from 10 locations in mice (n = 4) were analyzed by qPCR and microarray, respectively. The gene expression of glucagon, cholecystokinin, peptide YY, glucagon-like peptide-1 receptor, taste receptor T1R3, sodium/glucose cotransporter, peptide transporter-1, GPR120, taste receptor T1R1, GPR119 and GPR93 was investigated. Partial least squares (PLS) modeling was used to compare the intestinal expression pattern between the three species. RESULTS AND CONCLUSION: The studied genes were found to display specific expression patterns along the intestinal tract. PLS analysis showed a high similarity between human, pig and mouse in the expression of genes related to nutrient sensing in the distal ileum, and between human and pig in the colon. The gene expression pattern was most deviating between the species in the proximal intestine. Our results give new insights in interspecies similarities and provide new leads for translational research and models aiming to modulate food intake processes in man.


Assuntos
Ingestão de Alimentos/genética , Hormônios Gastrointestinais/biossíntese , Trato Gastrointestinal/metabolismo , Mucosa Intestinal/metabolismo , Animais , Alimentos , Hormônios Gastrointestinais/metabolismo , Expressão Gênica/genética , Humanos , Camundongos , Receptores Acoplados a Proteínas G/biossíntese , Receptores Acoplados a Proteínas G/metabolismo , Suínos
12.
Adv Exp Med Biol ; 817: 177-94, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24997034

RESUMO

Recent accumulating evidence indicates that the gut microbiome can affect the development and regulation of the hypothalamic-pituitary-adrenal axis and behavior, with central integrative systems being crucial in the successful physiological adaptation of the organism to external stressor. In contrast, host-derived hormones increase the bacterial proliferative capacity and pathogenicity. In the gut lumen, this type of cross-talk between microorganisms and the host is presumed to be performed continually through various kinds of luminal molecules, as numerous types of bacteria and host cells are in close proximity in the gastrointestinal tract of mammals.We herein focus on bidirectional signaling between the gut microbiome and the host in terms of commensal microbiota affecting the hypothalamic-pituitary-adrenal HPA axis response and behaviors and further discuss the role of gut luminal catecholamines and γ-aminobutyric acid, both of which are presumed to be involved in this signaling.


Assuntos
Hormônios Gastrointestinais/biossíntese , Sistema Hipotálamo-Hipofisário/fisiologia , Microbiota/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Animais , Catecolaminas/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Transdução de Sinais , Ácido gama-Aminobutírico/fisiologia
13.
J Biol Chem ; 287(23): 19574-84, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22528488

RESUMO

Bv8, also known as prokineticin 2, has been characterized as an important mediator of myeloid cell mobilization and myeloid cell-dependent tumor angiogenesis. Bv8 expression is dramatically enhanced by G-CSF, both in vitro and in vivo. The mechanisms involved in such up-regulation remain unknown. Using pharmacological inhibitors that interfere with multiple signaling pathways known to be activated by G-CSF, we show that signal transducer and activator of transcription 3 (Stat3) activation is required for Bv8 up-regulation in mouse bone marrow cells, whereas other Stat family members and extracellular signal-regulated kinase (ERK) activation are not involved. We further identified CD11b(+) Gr1(+) myeloid cells as the primary cell population in which Stat3 signaling is activated by G-CSF. Bv8 expression induced by G-CSF was also significantly reduced by siRNA-mediated Stat3 knockdown. Moreover, chromatin immunoprecipitation studies indicate that G-CSF significantly induces binding of phospho-Stat3 to the Bv8 promoter, which was abolished by pretreatment with the Stat3 inhibitor WP1066. Luciferase assay confirmed that the phospho-Stat3 binding site is a functional enhancer of the Bv8 promoter. The key role of Stat3 signaling in regulating G-CSF-induced Bv8 expression was further confirmed by in vivo studies. We show that the regulation of Bv8 expression in human bone marrow cells is also Stat3 signaling-dependent. Stat3 is recognized as a key regulator of inflammation-dependent tumorigenesis. We propose that such a role of Stat3 reflects at least in part its ability to regulate Bv8 expression.


Assuntos
Células da Medula Óssea/metabolismo , Antígeno CD11b , Hormônios Gastrointestinais/biossíntese , Regulação da Expressão Gênica/fisiologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Neuropeptídeos/biossíntese , Receptores de Superfície Celular , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hormônios Gastrointestinais/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Neuropeptídeos/genética , Regiões Promotoras Genéticas/fisiologia , Piridinas/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Transdução de Sinais/fisiologia , Tirfostinas/farmacologia
14.
Cancer Res ; 72(9): 2239-50, 2012 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-22406619

RESUMO

Bv8 (prokineticin 2) expressed by Gr1(+)CD11b(+) myeloid cells is critical for VEGF-independent tumor angiogenesis. Although granulocyte colony-stimulating factor (G-CSF) has been shown to be a key inducer of Bv8 expression, the basis for Bv8 production in driving tumor angiogenesis is undefined. Because the cell adhesion molecule CEACAM1, which is highly expressed on Gr1(+)CD11b(+) myeloid cells, is known to regulate G-CSF receptor (G-CSFR) signaling, we hypothesized that CEACAM1 would regulate Bv8 production in these cells. In support of this hypothesis, we found that Bv8 expression was elevated in Gr1(+)CD11b(+) cells from Ceacam1-deficient mice implanted with B16 melanoma, increasing the infiltration of Gr1(+)CD11b(+) myeloid cells in melanoma tumors and enhancing their growth and angiogenesis. Furthermore, treatment with anti-Gr1 or anti-Bv8 or anti-G-CSF monoclonal antibody reduced myeloid cell infiltration, tumor growth, and angiogenesis to levels observed in tumor-bearing wild-type (WT) mice. Reconstitution of CEACAM1-deficient mice with WT bone marrow cells restored tumor infiltration of Gr1(+)CD11b(+) cells along with tumor growth and angiogenesis to WT levels. Treatment of tumor-bearing WT mice with anti-CEACAM1 antibody limited tumor outgrowth and angiogenesis, albeit to a lesser extent. Tumor growth in Ceacam1-deficient mice was not affected significantly in Rag(-/-) background, indicating that CEACAM1 expression in T and B lymphocytes had a negligible role in this pathway. Together, our findings show that CEACAM1 negatively regulates Gr1(+)CD11b(+) myeloid cell-dependent tumor angiogenesis by inhibiting the G-CSF-Bv8 signaling pathway.


Assuntos
Antígeno Carcinoembrionário/metabolismo , Neoplasias do Colo/irrigação sanguínea , Melanoma Experimental/irrigação sanguínea , Células Mieloides/patologia , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Linfócitos B/patologia , Antígeno CD11b/biossíntese , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/imunologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Hormônios Gastrointestinais/biossíntese , Imuno-Histoquímica , Melanoma Experimental/genética , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Neovascularização Patológica/genética , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neuropeptídeos/biossíntese , Linfócitos T/patologia
15.
Histochem Cell Biol ; 137(5): 589-97, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22310983

RESUMO

Guanylin, a bioactive intestinal peptide, is involved in the cystic fibrosis transmembrane conductance (CFTR)-regulated electrolyte/water secretion in various epithelia. In the present work we report on the expression and cellular localization of guanylin and its affiliated signaling and effector proteins, including guanylate cyclase C (Gucy2c), Proteinkinase GII (Pkrg2), CFTR and the solute carrier family 4, anion exchanger, member 2 (Slc4a2) in the hepatobiliary system of rat and guinea pig. Localization studies in the liver and the gallbladder revealed that guanylin is located in the secretory epithelial cells of bile ducts of the liver and of the gallbladder, while Gucy2c, Pkrg2, CFTR, and Slc4a2 are confined exclusively to the apical membrane of the same epithelial cells. Based on these findings, we assume that guanylin is synthesized as an intrinsic peptide in epithelial cells of the hepatobiliary system and released luminally into the hepatic and cystic bile to regulate electrolyte secretion by a paracrine/luminocrine signaling pathway.


Assuntos
Vesícula Biliar/metabolismo , Hormônios Gastrointestinais/metabolismo , Fígado/metabolismo , Peptídeos Natriuréticos/metabolismo , Animais , Proteínas de Transporte de Ânions/análise , Proteínas de Transporte de Ânions/metabolismo , Antiporters/análise , Antiporters/metabolismo , Antiportadores de Cloreto-Bicarbonato , Regulador de Condutância Transmembrana em Fibrose Cística/análise , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/química , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Vesícula Biliar/química , Vesícula Biliar/citologia , Hormônios Gastrointestinais/análise , Hormônios Gastrointestinais/biossíntese , Guanilato Ciclase/análise , Guanilato Ciclase/metabolismo , Cobaias , Fígado/química , Fígado/citologia , Peptídeos Natriuréticos/análise , Peptídeos Natriuréticos/biossíntese , Ratos , Ratos Wistar , Proteínas SLC4A , Transdução de Sinais
16.
Med Oncol ; 29(3): 1748-57, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21972003

RESUMO

The molecular mechanisms leading to gastric carcinogenesis still remain unclear. Recently, several studies demonstrated that over-expression of guanylyl cyclase C (GCC) has been detected in intestinal-type gastric cancer (GC) and precursor lesions. Our objective was to explore the expression levels of GCC and endogenous ligands guanylin (GN) and uroguanylin (UGN) and the correlation between Helicobacter pylori (H. pylori) and GCC, GN, and UGN expressions in patients at different stages from normal mucosa to superficial gastritis, atrophic gastritis, intestinal metaplasia (IM), dysplasia, and finally adenocarcinoma. The expression of GCC and GN was absent in the distal normal gastric tissues and superficial gastritis in all cases, whereas they were measured in IM, dysplasia, and GC. The expression of GCC and GN was closely related to intestinal-type GC. From superficial gastritis to gastric carcinomas, the H. pylori positive rate was 19.7, 33.3, 69.6, 80.0, and 82.1%, respectively. The positive correlation was found between GCC and GN in IM, dysplasia, and GC. Also, the positive correlation was found between GCC, GN, and H. pylori infection in them. These results demonstrate that the detection of GCC and GN will be beneficial to diagnosis human gastric carcinoma and precancerous lesions. Ectopic expression of GCC and GN in human gastric mucosa and H. pylori infection may play an important role in the carcinogenesis of the intestinal-type GC.


Assuntos
Hormônios Gastrointestinais/biossíntese , Infecções por Helicobacter/complicações , Peptídeos Natriuréticos/biossíntese , Receptores Acoplados a Guanilato Ciclase/biossíntese , Receptores de Peptídeos/biossíntese , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiologia , Biomarcadores Tumorais/análise , Western Blotting , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Hormônios Gastrointestinais/análise , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori , Humanos , Imuno-Histoquímica , Ligantes , Peptídeos Natriuréticos/análise , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/microbiologia , Lesões Pré-Cancerosas/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase/análise , Receptores de Peptídeos/análise , Neoplasias Gástricas/patologia
17.
Thyroid ; 21(4): 391-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21385081

RESUMO

BACKGROUND: Endocrine gland-derived vascular endothelial growth factor (Prok1) and prokineticin 2 (Prok2) are involved in the organ-specific regulation of angiogenesis, which is a crucial step toward cancer progression in most tumors, including those of thyroid gland. The oncogene BRAF V600E mutation is associated with poor clinical outcome of papillary thyroid cancer (PTC) and can independently predict its recurrence. DESIGN: Our hypothesis was that Prok1 and Prok2 expression levels associated with BRAF mutations can be prognostic factors for PTC outcome. Prok1 and Prok2 were examined in PTC, a cell line derived from a human PTC (designated FB-2), euthyroid multinodular goiter (MNG), Graves' disease (GD), and contralateral normal thyroid (NT) tissues from PTC cases. We evaluated BRAF mutation and its relationship with Prok1 expression pattern in PTC. METHODS: We studied Prok1 and Prok2 mRNAs by real-time polymerase chain reaction and BRAF mutation by mutant allele-specific polymerase chain reaction amplification. Formalin-fixed, paraffin-embedded blocks of PTC and NT were used for the immunohistochemical determination of Prok1 using anti-endocrine gland vascular endothelial growth factor primary antibody. RESULTS: Prok1 and Prok2 transcripts were both present in thyroid tissues, and Prok1 was differentially expressed in PTC compared to MNG, GD, and NT. Prok1 mRNA levels were very low in NT and MNG and significantly higher in PTC, FB-2, and GD (p<0.05). Prok1 protein was almost undetectable in NT but was highly expressed in all PTC samples having an infiltrative pattern of growth and lymph node metastases ( p<0.05). Further, the expression of Prok1 in PTC was associated with 60% of the samples being positive for the BRAF mutation ( p<0.05). CONCLUSIONS: We found that Prok1 is significantly increased in PTC, and its expression in PTC is related to BRAF mutation. These results suggest that Prok1 could be a new useful marker for thyroid cancer progression. Prok1 therefore could also be a potential target for novel therapeutic strategies, although the lack of functional data suggests caution against generalization of this assumption


Assuntos
Hormônios Gastrointestinais/biossíntese , Proteínas Proto-Oncogênicas B-raf/genética , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Adulto , Idoso , Carcinoma , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Carcinoma Papilar/patologia , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neuropeptídeos/biossíntese , Câncer Papilífero da Tireoide , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Regulação para Cima
18.
PLoS One ; 4(9): e7151, 2009 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-19784373

RESUMO

The suprachiasmatic nucleus (SCN) in the hypothalamus is the predominant circadian clock in mammals. To function as a pacemaker, the intrinsic timing signal from the SCN must be transmitted to different brain regions. Prokineticin 2 (PK2) is one of the candidate output molecules from the SCN. In this study, we investigated the efferent projections of PK2-expressing neurons in the SCN through a transgenic reporter approach. Using a bacterial artificial chromosome (BAC) transgenic mouse line, in which the enhanced green fluorescence protein (EGFP) reporter gene expression was driven by the PK2 promoter, we were able to obtain an efferent projections map from the EGFP-expressing neurons in the SCN. Our data revealed that EGFP-expressing neurons in the SCN, hence representing some of the PK2-expressing neurons, projected to many known SCN target areas, including the ventral lateral septum, medial preoptic area, subparaventricular zone, paraventricular nucleus, dorsomedial hypothalamic nucleus, lateral hypothalamic area and paraventricular thalamic nucleus. The efferent projections of PK2-expressing neurons supported the role of PK2 as an output molecule of the SCN.


Assuntos
Hormônios Gastrointestinais/biossíntese , Regulação da Expressão Gênica , Neurônios/metabolismo , Neuropeptídeos/biossíntese , Núcleo Supraquiasmático/metabolismo , Animais , Encéfalo/metabolismo , Cromossomos Artificiais Bacterianos , Genes Reporter , Proteínas de Fluorescência Verde/metabolismo , Hipotálamo/metabolismo , Hibridização in Situ Fluorescente , Camundongos , Camundongos Transgênicos , Oscilometria , Regiões Promotoras Genéticas , Vasopressinas/metabolismo
20.
Pancreatology ; 9(1-2): 165-72, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19077468

RESUMO

AIMS: Endocrine gland-derived vascular endothelial growth factor (EG-VEGF)/prokineticins have been identified as tissue-specific angiogenic factors. This study investigates the expression and localization of EG-VEGF and its receptors in pancreatic tissues and pancreatic stellate cells (PSCs). METHODS: mRNA levels of EG-VEGF/prokineticin 1 (PK1), prokineticin 2 (PK2) and their receptors 1 (PKR1) and 2 (PKR2) were measured in pancreatic tissues, pancreatic cancer cell lines and PSCs by quantitative reverse-transcriptase polymerase chain reaction (QRT-PCR). Protein expression of PK1, PKR1 and PKR2 was assessed in pancreatic tissues by immunohistochemistry. Growth factor-induced secretion of EG-VEGF was measured by ELISA. RESULTS: QRT-PCR analysis in bulk tissues of normal pancreas, chronic pancreatitis and pancreatic ductal adenocarcinoma showed no significant difference of PK1 mRNA levels, whereas PK2 mRNA was barely detectable. High PK1 mRNA levels were observed only in cultured PSCs and microdissected islet cells, but not in cancer cells, and PK1 protein was localized mainly in islets and cancer-associated stromal cells. PKR1 and PKR2 proteins were present in endothelial cells of small blood vessels. TGF-beta(1) and PDGF-BB specifically stimulated PK1 secretion in PSCs. CONCLUSIONS: Islet and/or PSC-derived PK1 might act through its receptors on endothelial cells to increase angiogenesis in pancreatic diseases.


Assuntos
Ilhotas Pancreáticas/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/biossíntese , Linhagem Celular Tumoral , Células Cultivadas , Hormônios Gastrointestinais/biossíntese , Humanos , Neuropeptídeos/biossíntese , Pâncreas/citologia , Pancreatite Crônica/metabolismo , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Peptídeos/biossíntese , Fator de Crescimento do Endotélio Vascular Derivado de Glândula Endócrina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...