Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 261
Filtrar
1.
Cell ; 181(6): 1291-1306.e19, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32407674

RESUMEN

Enteroendocrine cells (EECs) sense intestinal content and release hormones to regulate gastrointestinal activity, systemic metabolism, and food intake. Little is known about the molecular make-up of human EEC subtypes and the regulated secretion of individual hormones. Here, we describe an organoid-based platform for functional studies of human EECs. EEC formation is induced in vitro by transient expression of NEUROG3. A set of gut organoids was engineered in which the major hormones are fluorescently tagged. A single-cell mRNA atlas was generated for the different EEC subtypes, and their secreted products were recorded by mass-spectrometry. We note key differences to murine EECs, including hormones, sensory receptors, and transcription factors. Notably, several hormone-like molecules were identified. Inter-EEC communication is exemplified by secretin-induced GLP-1 secretion. Indeed, individual EEC subtypes carry receptors for various EEC hormones. This study provides a rich resource to study human EEC development and function.


Asunto(s)
Células Enteroendocrinas/metabolismo , ARN Mensajero/genética , Células Cultivadas , Hormonas Gastrointestinales/genética , Tracto Gastrointestinal/metabolismo , Péptido 1 Similar al Glucagón/genética , Humanos , Organoides/metabolismo , Factores de Transcripción/genética , Transcriptoma/genética
2.
EMBO J ; 40(20): e108614, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34487375

RESUMEN

Circadian rhythms in mammals are governed by the hypothalamic suprachiasmatic nucleus (SCN), in which 20,000 clock cells are connected together into a powerful time-keeping network. In the absence of network-level cellular interactions, the SCN fails as a clock. The topology and specific roles of its distinct cell populations (nodes) that direct network functions are, however, not understood. To characterise its component cells and network structure, we conducted single-cell sequencing of SCN organotypic slices and identified eleven distinct neuronal sub-populations across circadian day and night. We defined neuropeptidergic signalling axes between these nodes, and built neuropeptide-specific network topologies. This revealed their temporal plasticity, being up-regulated in circadian day. Through intersectional genetics and real-time imaging, we interrogated the contribution of the Prok2-ProkR2 neuropeptidergic axis to network-wide time-keeping. We showed that Prok2-ProkR2 signalling acts as a key regulator of SCN period and rhythmicity and contributes to defining the network-level properties that underpin robust circadian co-ordination. These results highlight the diverse and distinct contributions of neuropeptide-modulated communication of temporal information across the SCN.


Asunto(s)
Relojes Circadianos/genética , Ritmo Circadiano/genética , Hormonas Gastrointestinales/genética , Neuropéptidos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Péptidos/genética , Núcleo Supraquiasmático/metabolismo , Transcriptoma , Animales , Péptido Liberador de Gastrina/genética , Péptido Liberador de Gastrina/metabolismo , Hormonas Gastrointestinales/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Neuronas/citología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Receptores de Bombesina/genética , Receptores de Bombesina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Receptores de Vasopresinas/genética , Receptores de Vasopresinas/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Núcleo Supraquiasmático/citología , Péptido Intestinal Vasoactivo/genética , Péptido Intestinal Vasoactivo/metabolismo , Vasopresinas/genética , Vasopresinas/metabolismo
3.
J Biol Chem ; 299(1): 102814, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36539034

RESUMEN

Energy homeostasis is a complex system involving multiple hormones, neuropeptides, and receptors. Prokineticins (PK1 and PK2) are agonists to two G protein-coupled receptors, prokineticin receptor 1 and 2 (PKR1 and PKR2), which decrease food intake when injected in rodents. The relative contribution of PKR1 and PKR2 to the anorexigenic effect of PK2 and their site of action in the brain have not yet been elucidated. While PKR1 and PKR2 are both expressed in the hypothalamus, a central region involved in the control of energy homeostasis, PKR2 is also present in the amygdala, which has recently been shown to regulate food intake in response to several anorexigenic signals. PKR trafficking and signaling are inhibited by the melanocortin receptor accessory protein 2 (MRAP2), thus suggesting that MRAP2 has the potential to alter the anorexigenic activity of PK2 in vivo. In this study, we investigated the importance of PKR1 and PKR2 for PK2-mediated inhibition of food intake, the brain region involved in this function, and the effect of MRAP2 on PK2 action in vivo. Using targeted silencing of PKR2 and chemogenetic manipulation of PKR2 neurons, we show that the anorexigenic effect of PK2 is mediated by PKR2 in the amygdala and that altering MRAP2 expression in PKR2 neurons modulates the activity of PK2. Collectively, our results provide evidence that inhibition of food intake by PKs is not mediated through activation of hypothalamic neurons but rather amygdala PKR2 neurons and further establishes the importance of MRAP2 in the regulation of energy homeostasis.


Asunto(s)
Hormonas Gastrointestinales , Neuropéptidos , Proteínas Portadoras/metabolismo , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo , Hormonas Gastrointestinales/farmacología , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal
4.
Int J Med Sci ; 21(1): 27-36, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38164347

RESUMEN

Prokineticin 1 (PROK1) is a secreted protein involved in a range of physiological activities such as cell proliferation, migration, angiogenesis, and neuronal cell proliferation. Emerging evidences show that PROK1/PROK receptors (PROKRs) are expressed by trophoblasts, and decidual stroma cells at the maternal-fetal interface. PROK1 plays a critical role in successful pregnancy establishment by regulating the decidualization, implantation and placental development. Dysregulation of prokineticin signaling has been described in certain pathological states associated with pregnancy, including pre-eclampsia, recurrent miscarriage and fetal growth restriction. In this review, the expression and pleiotropic roles of PROK1 under physiological and pathological pregnancy conditions are discussed.


Asunto(s)
Hormonas Gastrointestinales , Preeclampsia , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina , Embarazo , Femenino , Humanos , Placenta/metabolismo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Transducción de Señal/genética , Trofoblastos , Preeclampsia/genética , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo
5.
Mol Hum Reprod ; 27(11)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34643696

RESUMEN

Evidence accumulated in recent years has revealed that neutrophils are involved in the initial establishment of endometriosis, which is well-known as a chronic inflammatory disease. So far, why and how neutrophils promote the formation of early endometriosis are still unclear. In this study, using a mouse model of endometriosis, we demonstrated that endometriosis mice (EMs mice) had a significantly increased number of neutrophils in peritoneal fluids and lesions, and increased levels of granulocyte colony-stimulating factor (G-CSF) and IL-6 in serum and peritoneal fluids compared to the control group. In the neutrophils and uterine fragments co-injection experiment, neutrophils regulated by G-CSF and IL-6 had a similar effect to neutrophils from EMs mice, increasing the number, area, weight and microvessel density (MVD) of endometriotic lesions. Blocking the effect of G-CSF and IL-6 in EMs mice resulted in a decrease in the number, area and weight of endometriotic lesions. Following the depletion of neutrophils in vivo using a anti-Ly6G antibody, the MVD in the lesions of mice treated with neutrophils from EMs mice and neutrophils from pG/pI6 mice were significantly reduced. Neutrophils from EMs mice and neutrophils from pG/pI6 mice altered the expression levels of Mmp9, Bv8 and Trail genes compared to the neutrophils from PBS-treated mice. IL-6 together with G-CSF induced a higher expression of phospho-STAT3 and STAT3 in neutrophils. These findings suggest that neutrophils modulated by G-CSF and IL-6 through the STAT3 pathway alter the expression levels of the angiogenesis-related genes Mmp9, Bv8 and Trail, and may promote the establishment of early endometriosis.


Asunto(s)
Endometriosis/metabolismo , Endometrio/irrigación sanguínea , Endometrio/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Interleucina-6/metabolismo , Neovascularización Patológica , Neutrófilos/metabolismo , Animales , Modelos Animales de Enfermedad , Endometriosis/inmunología , Endometriosis/patología , Endometrio/inmunología , Femenino , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo , Factor Estimulante de Colonias de Granulocitos/genética , Interleucina-6/genética , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Densidad Microvascular , Neuropéptidos/genética , Neuropéptidos/metabolismo , Neutrófilos/inmunología , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
6.
Cell Biol Int ; 45(2): 404-410, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33140874

RESUMEN

Kallmann syndrome (KS) is a rare human genetic disorder characterized by hypogonadotropic hypogonadism with the reduction or absence of olfactory sense. Mutations in multiple genes, including chemokine prokineticin-2 (PROK2), are considered to contribute to the abnormal migration of gonadotropin-releasing hormone neurons in the embryonic stage. However, the mechanisms of the different inheritance modes of KS have not been comprehensively determined. In this article, we present the case of one KS patient with the same mutation in PROK2 (c.223-4C>A) as his mother. RNA sequencing analysis of his leukocytes showed a new transcript of PROK2, which contained a partial intron (192 bp) compared to those of his parents. Furthermore, we observed that hsa-miR-3195 was expressed at low levels in his and his father's sera compared to his mother's. Unexpectedly, hsa-miR-3195 was also identified to specifically target the 192 bp intron of the aberrant PROK2 transcript of this patient. We determined that high expression of hsa-miR-3195 could efficiently target aberrant PROK2 and stabilize the normal function of PROK2 in vitro, which provided a probable explanation for the different phenotypes of the patient and his mother with the same genotype.


Asunto(s)
Hormonas Gastrointestinales/genética , Síndrome de Kallmann/genética , Neuropéptidos/genética , Adolescente , Femenino , Genotipo , Células HEK293 , Humanos , Masculino , MicroARNs/metabolismo , Mutación , Linaje , Fenotipo , Transcriptoma
7.
Cell Biochem Funct ; 39(2): 308-316, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32876972

RESUMEN

Cancer cell derived exosomes play important roles in cancer progression and modulation of the tumour microenvironment. This study aims to investigate the role of prokineticin receptor 1 (PKR1) positive exosomes on angiogenesis. In the present study, PKR1 expression in tumour samples from ovarian cancer patients were examined firstly. Then, two ovarian cancer cell lines, namely A2780 and HO-8910 cells, were used to isolate and obtain the PKR1 positive exosomes from the serum free medium. The function analysis of PKR1 positive exosomes on angiogenesis was conducted by cell proliferation and migration assay, tube formation analysis, and tumour volume assay. The results showed that PKR1 expression was down regulated in tumour samples of ovarian cancer patients compared with adjacent normal tissues. The intracellular expression of PKR1 could be detected in A2780 and HO-8910 cells. And, the isolated exosomes from the serum free medium were confirmed by transmission electron microscopic and NTA analysis, as well as the co-presence of PKR1 with exosome marker CD63. The function analysis of PKR1 positive exosomes on angiogenesis demonstrated the uptake of PKR1 positive exosomes by human umbilical vein endothelial cells through immunofluorescence staining. The angiogenesis assays in vitro indicated that PKR1 positive exosomes promoted migration and tube formation of HUVECs but not proliferation. The endogenous PKR1 was also verified to help to enhance migration and promote tube formation of vascular endothelial cells, which might involved in the phosphorylation of STAT3. Additionally, The tumour volume from exosomes treated A2780 tumour-bearing mice was significantly increased compared with the control group, accompanied with the induced PKR1 expression and phosphorylation of STAT3 level. SIGNIFICANCE OF THE STUDY: This study proved the important role of PKR1 positive exosomes released from ovarian cancer cells on promoting angiogenesis. The data indicated that PKR1 derived from ovarian cancer cells could act as an important tumour associated antigen and biomolecular factor for cellular communication in tumour microenvironment.


Asunto(s)
Exosomas/metabolismo , Hormonas Gastrointestinales/metabolismo , Neovascularización Fisiológica , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Exosomas/trasplante , Femenino , Hormonas Gastrointestinales/antagonistas & inhibidores , Hormonas Gastrointestinales/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Trasplante Heterólogo , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/antagonistas & inhibidores , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética
8.
Int J Mol Sci ; 22(21)2021 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-34769347

RESUMEN

Chemotherapy-induced neuropathy (CIN) is a major adverse effect associated with many chemotherapeutics, including bortezomib (BTZ). Several mechanisms are involved in CIN, and recently a role has been proposed for prokineticins (PKs), a chemokine family that induces proinflammatory/pro-algogen mediator release and drives the epigenetic control of genes involved in cellular differentiation. The present study evaluated the relationships between epigenetic mechanisms and PKs in a mice model of BTZ-induced painful neuropathy. To this end, spinal cord alterations of histone demethylase KDM6A, nuclear receptors PPARα/PPARγ, PK2, and pro-inflammatory cytokines IL-6 and IL-1ß were assessed in neuropathic mice treated with the PK receptors (PKRs) antagonist PC1. BTZ treatment promoted a precocious upregulation of KDM6A, PPARs, and IL-6, and a delayed increase of PK2 and IL-1ß. PC1 counteracted allodynia and prevented the increase of PK2 and of IL-1ß in BTZ neuropathic mice. The blockade of PKRs signaling also opposed to KDM6A increase and induced an upregulation of PPAR gene transcription. These data showed the involvement of epigenetic modulatory enzymes in spinal tissue phenomena associated with BTZ painful neuropathy and underline a role of PKs in sustaining the increase of proinflammatory cytokines and in exerting an inhibitory control on the expression of PPARs through the regulation of KDM6A gene expression in the spinal cord.


Asunto(s)
Bortezomib/toxicidad , Hormonas Gastrointestinales/metabolismo , Histona Demetilasas/metabolismo , Hiperalgesia/patología , Neuropéptidos/metabolismo , Dolor/patología , Enfermedades del Sistema Nervioso Periférico/patología , Médula Espinal/patología , Animales , Antineoplásicos/toxicidad , Citocinas/metabolismo , Hormonas Gastrointestinales/genética , Histona Demetilasas/genética , Hiperalgesia/inducido químicamente , Hiperalgesia/genética , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/genética , PPAR alfa/genética , PPAR alfa/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Dolor/inducido químicamente , Dolor/genética , Dolor/metabolismo , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/genética , Enfermedades del Sistema Nervioso Periférico/metabolismo , Médula Espinal/metabolismo
9.
Molecules ; 26(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33572808

RESUMEN

Type 2 diabetes (T2D) is a fast-increasing health problem globally, and it results from insulin resistance and pancreatic ß-cell dysfunction. The gastrointestinal (GI) tract is recognized as one of the major regulatory organs of glucose homeostasis that involves multiple gut hormones and microbiota. Notably, the incretin hormone glucagon-like peptide-1 (GLP-1) secreted from enteroendocrine L-cells plays a pivotal role in maintaining glucose homeostasis via eliciting pleiotropic effects, which are largely mediated via its receptor. Thus, targeting the GLP-1 signaling system is a highly attractive therapeutic strategy to treatment T2D. Polyphenols, the secondary metabolites from plants, have drawn considerable attention because of their numerous health benefits, including potential anti-diabetic effects. Although the major targets and locations for the polyphenolic compounds to exert the anti-diabetic action are still unclear, the first organ that is exposed to these compounds is the GI tract in which polyphenols could modulate enzymes and hormones. Indeed, emerging evidence has shown that polyphenols can stimulate GLP-1 secretion, indicating that these natural compounds might exert metabolic action at least partially mediated by GLP-1. This review provides an overview of nutritional regulation of GLP-1 secretion and summarizes recent studies on the roles of polyphenols in GLP-1 secretion and degradation as it relates to metabolic homeostasis. In addition, the effects of polyphenols on microbiota and microbial metabolites that could indirectly modulate GLP-1 secretion are also discussed.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Polifenoles/uso terapéutico , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Hormonas Gastrointestinales/genética , Péptido 1 Similar al Glucagón/genética , Humanos , Secreción de Insulina/genética , Células Secretoras de Insulina/patología
10.
J Cell Mol Med ; 24(5): 3242-3245, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31991505

RESUMEN

Prokineticin 1 (PROK1) is a key regulator of embryo implantation and placentation, and its dysregulation is associated with pregnancy complications, such as pre-eclampsia and foetal growth restriction. We have previously shown that insulin strongly enhances the expression of PROK1 in human decidualizing stromal cells. Here, we demonstrate that dihydrotestosterone (DHT), but not testosterone, potentiates insulin to up-regulate PROK1 in these cells. However, the androgens alone do not influence the expression of PROK1. Our findings suggest that insulin and androgens both are involved in the regulation of PROK1 that could have implications for normal and pathological pregnancies.


Asunto(s)
Dihidrotestosterona/farmacología , Endometrio/metabolismo , Hormonas Gastrointestinales/genética , Insulina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Adolescente , Adulto , Andrógenos/genética , Biopsia , Células Cultivadas , Decidua/metabolismo , Decidua/patología , Implantación del Embrión/genética , Endometrio/efectos de los fármacos , Endometrio/crecimiento & desarrollo , Células Epiteliales/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Placentación/genética , Preeclampsia , Embarazo , Complicaciones del Embarazo/tratamiento farmacológico , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/patología , Cultivo Primario de Células , Transducción de Señal/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Activación Transcripcional , Adulto Joven
11.
J Cell Physiol ; 235(4): 3776-3789, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31602654

RESUMEN

Fatty acids (FAs) play a crucial role in the development of clear cell renal cell carcinoma (ccRCC), FAs function requires the participation of fatty-acid-binding protein (FABP). Current studies have shown that different members of the FABP's family play different roles in the tumorigenesis of ccRCC. Therefore, the systematic analysis of FABPs will be of great significance. However, the diverse expression patterns and prognostic values of nine FABPs have yet to be elucidated. In this study, through multiple analysis and verification of multiple databases, such as ONCOMINE, The Human Protein Atlas, UALCAN, Gene Expression Profiling Interactive Analysis, and cBioPortal, we found that the expression of FABP1 was significantly downregulated and the expression of FABP5/6/7 was significantly upregulated in ccRCC compared with renal tissues, and the patients with high messenger RNA (mRNA) levels of the FABP5/6/7 or low mRNA levels of FABP1 were predicted to have a lower overall survival or disease-free survival. Further analysis by the protein-protein interaction (PPI), Gene Ontology pathway, and Kyoto Encyclopedia of Genes and Genomes pathway showed that FABPs were mainly involved in the peroxisome proliferator-activated receptor (PPAR) pathway. In coexpression analysis, we found that FABP1/5/6/7 was coexpressed with transforming growth factor-ß1 (TGF-ß1), PPARA, and LPL. This study implied that FABP1/5/6/7 could act as an important tumor biomarker of ccRCC; the role of FABPs may be related to PPAR or TGF-ß pathway.


Asunto(s)
Carcinoma de Células Renales/genética , Proteína de Unión a los Ácidos Grasos 7/genética , Proteínas de Unión a Ácidos Grasos/genética , Hormonas Gastrointestinales/genética , Proteínas Supresoras de Tumor/genética , Carcinogénesis/genética , Carcinoma de Células Renales/epidemiología , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Pronóstico , Mapas de Interacción de Proteínas
12.
PLoS Pathog ; 14(4): e1006955, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29621339

RESUMEN

Macrolides are used to treat various inflammatory diseases owing to their immunomodulatory properties; however, little is known about their precise mechanism of action. In this study, we investigated the functional significance of the expansion of myeloid-derived suppressor cell (MDSC)-like CD11b+Gr-1+ cells in response to the macrolide antibiotic clarithromycin (CAM) in mouse models of shock and post-influenza pneumococcal pneumonia as well as in humans. Intraperitoneal administration of CAM markedly expanded splenic and lung CD11b+Gr-1+ cell populations in naïve mice. Notably, CAM pretreatment enhanced survival in a mouse model of lipopolysaccharide (LPS)-induced shock. In addition, adoptive transfer of CAM-treated CD11b+Gr-1+ cells protected mice against LPS-induced lethality via increased IL-10 expression. CAM also improved survival in post-influenza, CAM-resistant pneumococcal pneumonia, with improved lung pathology as well as decreased interferon (IFN)-γ and increased IL-10 levels. Adoptive transfer of CAM-treated CD11b+Gr-1+ cells protected mice from post-influenza pneumococcal pneumonia. Further analysis revealed that the CAM-induced CD11b+Gr-1+ cell expansion was dependent on STAT3-mediated Bv8 production and may be facilitated by the presence of gut commensal microbiota. Lastly, an analysis of peripheral blood obtained from healthy volunteers following oral CAM administration showed a trend toward the expansion of human MDSC-like cells (Lineage-HLA-DR-CD11b+CD33+) with increased arginase 1 mRNA expression. Thus, CAM promoted the expansion of a unique population of immunosuppressive CD11b+Gr-1+ cells essential for the immunomodulatory properties of macrolides.


Asunto(s)
Claritromicina/farmacología , Hormonas Gastrointestinales/metabolismo , Neuropéptidos/metabolismo , Infecciones por Orthomyxoviridae/complicaciones , Neumonía Neumocócica/tratamiento farmacológico , Factor de Transcripción STAT3/metabolismo , Choque Séptico/tratamiento farmacológico , Streptococcus pneumoniae/efectos de los fármacos , Animales , Antibacterianos/farmacología , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Hormonas Gastrointestinales/genética , Lipopolisacáridos/toxicidad , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Mieloides/efectos de los fármacos , Células Mieloides/microbiología , Células Mieloides/virología , Neuropéptidos/genética , Orthomyxoviridae/patogenicidad , Infecciones por Orthomyxoviridae/virología , Fagocitosis/efectos de los fármacos , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/virología , Factor de Transcripción STAT3/genética , Choque Séptico/inducido químicamente
13.
Dig Dis Sci ; 65(12): 3592-3604, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32671585

RESUMEN

BACKGROUND: Alcohol-associated liver disease accounts for half of cirrhosis-related deaths worldwide. The spectrum of disease varies from simple steatosis to fibrosis, cirrhosis and ultimately hepatocellular carcinoma. Understanding the disease on a molecular level helps us to develop therapeutic targets. AIM: We performed transcriptomic analysis in liver and ileum from chronic plus binge ethanol-fed mice, and we assessed the role of selected differentially expressed genes and their association with serum bile acids and gut microbiota. METHODS: Wild-type mice were subjected to a chronic Lieber-DeCarli diet model for 8 weeks followed by one binge of ethanol. RNA-seq analysis was performed on liver and ileum samples. Associations between selected differentially regulated genes and serum bile acid profile or fecal bacterial profiling (16S rDNA sequencing) were investigated. RESULTS: We provide a comprehensive transcriptomic analysis to identify differentially expressed genes, KEGG pathways, and gene ontology functions in liver and ileum from chronic plus binge ethanol-fed mice. In liver, we identified solute carrier organic anion transporter family, member 1a1 (Slco1a1; encoding for organic anion transporting polypeptides (OATP) 1A1), as the most down-regulated mRNA, and it is negatively correlated with serum cholic acid level. Prokineticin 2 (Prok2) mRNA, a cytokine-like molecule associated with gastrointestinal tract inflammation, was significantly down-regulated in ethanol-fed mice. Prok2 mRNA expression was negatively correlated with abundance of Allobaculum (genus), Coprococcus (genus), Lachnospiraceae (family), Lactococcus (genus), and Cobriobacteriaceae (family), while it positively correlated with Bacteroides (genus). CONCLUSIONS: RNA-seq analysis revealed unique transcriptomic signatures in the liver and intestine following chronic plus binge ethanol feeding.


Asunto(s)
Etanol/farmacología , Hormonas Gastrointestinales/genética , Microbioma Gastrointestinal , Intestinos , Hepatopatías Alcohólicas/metabolismo , Hígado , Neuropéptidos/genética , Proteínas de Transporte de Catión Orgánico/genética , Animales , Depresores del Sistema Nervioso Central/farmacología , Ácido Cólico/análisis , Correlación de Datos , Regulación hacia Abajo , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Perfilación de la Expresión Génica/métodos , Intestinos/efectos de los fármacos , Intestinos/microbiología , Intestinos/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Análisis de Secuencia de ARN/métodos
14.
Int J Mol Sci ; 21(17)2020 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-32887509

RESUMEN

Cervical cancer is the second most frequent type of gynecologic cancer worldwide. Prokineticin 2 (PROK2) is reported to be involved in tumor progression in some malignant tumors. However, the role of PROK2 in the development of cervical cancer remains unknown. Our results indicate that PROK2 is overexpressed in the human cervical cancer. Cervical cancer patients with high PROK2 expression have a shorter overall survival rate (OS) and disease-free survival rate (DFS). PROK2 acts as a potential biomarker for predicting OS and DFS of cervical cancer patients. We further show that PROK2 is important factor for oncogenic migration and invasion in human cervical cancer cells. Knockdown PROK2 significantly inhibited cell migration, invasion, and MMP15 protein expression in HeLa cells. High expression of MMP15 is confirmed in the human cervical cancer, is significantly associated with the shorter overall survival rate (OS) and is correlated with PROK2 expression. Overexpression of PROK2 using PROK2 plasmid significantly reverses the function of knockdown PROK2, and further upregulates MMP15 expression, migration and invasion of human cervical cancer cells. In conclusion, our findings are the first to demonstrate the role of PROK2 as a novel and potential biomarker for clinical use, and reveal the oncogenic functions of PROK2 as therapeutic target for cervical cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Hormonas Gastrointestinales/metabolismo , Regulación Neoplásica de la Expresión Génica , Metaloproteinasa 15 de la Matriz/metabolismo , Neuropéptidos/metabolismo , Neoplasias del Cuello Uterino/patología , Apoptosis , Biomarcadores de Tumor/genética , Ciclo Celular , Movimiento Celular , Proliferación Celular , Femenino , Hormonas Gastrointestinales/antagonistas & inhibidores , Hormonas Gastrointestinales/genética , Humanos , Metaloproteinasa 15 de la Matriz/química , Metaloproteinasa 15 de la Matriz/genética , Invasividad Neoplásica , Neuropéptidos/antagonistas & inhibidores , Neuropéptidos/genética , Pronóstico , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
15.
Am J Physiol Cell Physiol ; 317(6): C1239-C1246, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31553648

RESUMEN

Recently, we showed that double-transgenic rats overexpressing guanylin (Gn), a bioactive peptide, and its receptor, guanylyl cyclase-C (GC-C), specifically in macrophages demonstrate an antiobesity phenotype and low-expression levels of proinflammatory cytokines in the mesenteric fat even when fed a high-fat diet. Here, we examined the levels and mechanism of Gn and GC-C transcription following saturated fatty acid and lipopolysaccharide (LPS), an activator of Toll-like receptor 4 (TLR4), exposure by using the NR8383 macrophage cell line. In addition, the levels of guanylin and cGMP were increased by addition of either palmitic acid or LPS. Next, we investigated the interaction of the gene transcription and nuclear factor-κB (NF-κB) by using an NF-κB inhibitor and chromatin immunoprecipitation assay. We showed that palmitic acid induced Gn gene expression via TLR4 and NF-κB. Moreover, we demonstrated that NF-κB binding to the Gn promoter was responsible for the induction of gene transcription by palmitic acid or LPS. Our results indicate that saturated fatty acids such as palmitic acid activate Gn gene expression via the NF-κB pathway, raising the possibility that the activated Gn-GC-C system may contribute to the inhibition of high-fat diet-induced proinflammatory cytokines in macrophages.


Asunto(s)
Hormonas Gastrointestinales/genética , Lipopolisacáridos/farmacología , Macrófagos Alveolares/efectos de los fármacos , FN-kappa B/genética , Péptidos Natriuréticos/genética , Ácido Palmítico/farmacología , Receptor Toll-Like 4/genética , Animales , Línea Celular , GMP Cíclico/inmunología , GMP Cíclico/metabolismo , Hormonas Gastrointestinales/agonistas , Hormonas Gastrointestinales/inmunología , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Macrófagos Alveolares/citología , Macrófagos Alveolares/inmunología , Ratones , FN-kappa B/inmunología , Péptidos Natriuréticos/agonistas , Péptidos Natriuréticos/inmunología , Células RAW 264.7 , Ratas , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores Acoplados a la Guanilato-Ciclasa/inmunología , Transducción de Señal , Células THP-1 , Receptor Toll-Like 4/inmunología
16.
Circulation ; 138(9): 913-928, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-29545372

RESUMEN

BACKGROUND: Angiogenesis is integral for embryogenesis, and targeting angiogenesis improves the outcome of many pathological conditions in patients. TBX20 is a crucial transcription factor for embryonic development, and its deficiency is associated with congenital heart disease. However, the role of TBX20 in angiogenesis has not been described. METHODS: Loss- and gain-of-function approaches were used to explore the role of TBX20 in angiogenesis both in vitro and in vivo. Angiogenesis gene array was used to identify key downstream targets of TBX20. RESULTS: Unbiased gene array survey showed that TBX20 knockdown profoundly reduced angiogenesis-associated PROK2 (prokineticin 2) gene expression. Indeed, loss of TBX20 hindered endothelial cell migration and in vitro angiogenesis. In a murine angiogenesis model using subcutaneously implanted Matrigel plugs, we observed that TBX20 deficiency markedly reduced PROK2 expression and restricted intraplug angiogenesis. Furthermore, recombinant PROK2 administration enhanced angiogenesis and blood flow recovery in murine hind-limb ischemia. In zebrafish, transient knockdown of tbx20 by morpholino antisense oligos or genetic disruption of tbx20 by CRISPR/Cas9 impaired angiogenesis. Furthermore, loss of prok2 or its cognate receptor prokr1a also limited angiogenesis. In contrast, overexpression of prok2 or prokr1a rescued the impaired angiogenesis in tbx20-deficient animals. CONCLUSIONS: Our study identifies TBX20 as a novel transcription factor regulating angiogenesis through the PROK2-PROKR1 (prokineticin receptor 1) pathway in both development and disease and reveals a novel mode of angiogenic regulation whereby the TBX20-PROK2-PROKR1 signaling cascade may act as a "biological capacitor" to relay and sustain the proangiogenic effect of vascular endothelial growth factor. This pathway may be a therapeutic target in the treatment of diseases with dysregulated angiogenesis.


Asunto(s)
Hormonas Gastrointestinales/metabolismo , Isquemia/metabolismo , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Dominio T Box/metabolismo , Proteínas de Pez Cebra/metabolismo , Inductores de la Angiogénesis/farmacología , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Hormonas Gastrointestinales/genética , Regulación del Desarrollo de la Expresión Génica , Miembro Posterior , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isquemia/tratamiento farmacológico , Isquemia/genética , Isquemia/fisiopatología , Masculino , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Fisiológica/efectos de los fármacos , Neuropéptidos/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteínas de Dominio T Box/genética , Pez Cebra , Proteínas de Pez Cebra/genética
17.
Biol Reprod ; 101(4): 832-841, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31276578

RESUMEN

CONTEXT: Prokineticin 1 (PROK1) quantification in global follicular fluid (FF) has been recently reported as a predictive biomarker of in vitro fertilization (IVF) outcome. It is now necessary to evaluate its clinical usefulness in individual follicles. OBJECTIVES: To evaluate the clinical value of PROK1 secretion in individual FF to predict oocyte competence. To determine the impact of follicular size, oocyte maturity, and gonadotropin treatments on PROK1 secretion. DESIGN AND SETTING: Prospective cohort study from May 2015 to May 2017 at the University Hospital of Grenoble. PATIENTS: A total of 69 infertile couples underwent IVF. INTERVENTION(S): Collection of 298 individual FF from 44 women undergoing IVF; 52 individual cumulus cell (CC) samples and 15 CC primary cultures from 25 women undergoing IVF-intracytoplasmic sperm injection (ICSI). MAIN OUTCOME MEASURE(S): Oocyte competence was defined as the ability to sustain embryo development to the blastocyst stage. Follicular size was measured by 2D-sonography. PROK1 concentration was quantified by ELISA assay. RESULTS: PROK1 concentration was correlated to follicular size (r = 0.85, P = 2.2 × 10-16). Normalized PROK1 concentration in FF was predictive of subsequent oocyte competence (AUROC curve = 0.76 [95% CI, 0.69-0.83]; P = 1.7 × 10-9), irrespectively of day-2 embryo morphokinetic parameters. The expression and secretion of PROK1 were increased in FF and CC of mature oocytes (P < 0.01). Follicle Stimulating Hormone and hCG up-regulated PROK1 secretion in CC primary cultures (P < 0.01; P < 0.05), probably through the cAMP pathway (P < 0.01). CONCLUSIONS: PROK1 quantification in individual FF could constitute a new predictive biomarker of oocyte competence in addition with embryo morphokinetic parameters. TRIAL REGISTRATION NUMBER: none.


Asunto(s)
Biomarcadores/análisis , Desarrollo Embrionario , Líquido Folicular/química , Hormonas Gastrointestinales/análisis , Oocitos/fisiología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/análisis , Biomarcadores/metabolismo , Células Cultivadas , Estudios de Cohortes , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Fertilización In Vitro , Líquido Folicular/metabolismo , Francia , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/metabolismo , Expresión Génica/efectos de los fármacos , Hormonas/farmacología , Humanos , Recuperación del Oocito/normas , Oocitos/citología , Oogénesis/efectos de los fármacos , Oogénesis/genética , Oogénesis/fisiología , Embarazo , Índice de Embarazo , Pronóstico , Estudios Prospectivos , Control de Calidad , Inyecciones de Esperma Intracitoplasmáticas , Resultado del Tratamiento , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo
18.
Stem Cells ; 36(10): 1589-1602, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29873146

RESUMEN

Epicardial adipose tissues (EATs) and vascular tissues may both belong to the mesoepithelial lineage that develops from epicardium-derived progenitor cells (EPDCs) in developing and injured hearts. Very little is known of the molecular mechanisms of EPDC contribution in EAT development and neovascularization in adult heart, which the topic remains a subject of intense therapeutic interest and scientific debate. Here we studied the epigenetic control of stemness and anti-adipogenic and pro-vasculogenic fate of human EPDCs (hEPDCs), through investigating an angiogenic hormone, prokineticin-2 (PK2) signaling via its receptor PKR1. We found that hEPDCs spontaneously undergoes epithelial-to-mesenchymal transformation (EMT), and are not predestined for the vascular lineages. However, PK2 via a histone demethylase KDM6A inhibits EMT, and induces asymmetric division, leading to self-renewal and formation of vascular and epithelial/endothelial precursors with angiogenic potential capable of differentiating into vascular smooth muscle and endothelial cells. PK2 upregulates and activates KDM6A to inhibit repressive histone H3K27me3 marks on promoters of vascular genes (Flk-1 and SM22α) involved in vascular lineage commitment and maturation. In PK2-mediated anti-adipogenic signaling, KDM6A stabilizes and increases cytoplasmic ß-catenin levels to repress peroxisome proliferator-activated receptor-γ expression and activity. Our findings offer additional molecular targets to manipulate hEPDCs-involved tissue repair/regeneration in cardiometabolic and ischemic heart diseases. Stem Cells 2018;36:1589-1602.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Hormonas Gastrointestinales/metabolismo , Neuropéptidos/metabolismo , Pericardio/citología , Pericardio/metabolismo , Diferenciación Celular/fisiología , Epigénesis Genética , Transición Epitelial-Mesenquimal , Hormonas Gastrointestinales/genética , Histona Demetilasas/metabolismo , Humanos , Neuropéptidos/genética , Proteínas Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
19.
J Cell Mol Med ; 22(1): 163-172, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28782224

RESUMEN

Prokineticin 1 (PROK1), a hypoxia-regulated angiogenic factor, has emerged as a crucial regulator of embryo implantation and placentation. Dysregulation of PROK1 has been linked to recurrent pregnancy loss, pre-eclampsia, foetal growth restriction and preterm birth. These pregnancy complications are common in women with obesity and polycystic ovary syndrome, i.e. conditions associated with insulin resistance and compensatory hyperinsulinaemia. We investigated the effect of insulin on PROK1 expression during in vitro decidualization. Endometrial stromal cells were isolated from six healthy, regularly menstruating women and decidualized in vitro. Insulin induced a significant dose-dependent up-regulation of PROK1 on both mRNA and protein level in decidualizing endometrial stromal cells. This up-regulation was mediated by hypoxia-inducible factor 1-alpha (HIF1α) via the phosphatidylinositol 3-kinase (PI3K) pathway. Furthermore, we demonstrated that PROK1 did not affect the viability, but significantly inhibited the migration of endometrial stromal cells and the migratory and invasive capacity of trophoblast cell lines. This in vitro study provides new insights into the regulation of PROK1 by insulin in human decidualizing endometrial stromal cells, the action of PROK1 on migration of endometrial stromal cells, as well as migration and invasion of trophoblasts. We speculate that hyperinsulinaemia may be involved in the mechanisms by which PROK1 is linked to placenta-related pregnancy complications.


Asunto(s)
Decidua/citología , Decidua/metabolismo , Hormonas Gastrointestinales/genética , Insulina/farmacología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/genética , Adolescente , Adulto , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Coriocarcinoma/patología , Femenino , Hormonas Gastrointestinales/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Trofoblastos/citología , Factor de Crecimiento Endotelial Vascular Derivado de Glándula Endocrina/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Adulto Joven
20.
BMC Cancer ; 18(1): 1114, 2018 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-30442117

RESUMEN

BACKGROUND: To identify potential therapeutic target in clear cell renal cell carcinoma (ccRCC), we performed a transcriptome analysis. Our analysis showed that fatty acid binding protein 7 (FABP7) has the highest mean differential overexpression in ccRCC compared to normal kidney. We aimed to investigate the significance of FABP7 in ccRCC. METHODS: Immunohistochemical staining for 40 advanced ccRCC cases was performed to investigate correlation between clinicopathological parameters and FABP7. They were composed of 40-83 years old cases with 33 male, 22 cases with pT ≥ 3, 19 cases with M1, and 16 cases with grade 3. The effect of gene knockdown was analysed by a cell viability assay and invasion assay in FABP7-overexpressing cell lines (SKRC7 and SKRC10). RESULTS: Our immunohistochemical analysis showed that higher FABP7 expression significantly correlated with distant metastasis and poor cancer-specific survival (CSS; both p < 0.05). Functional suppression of FABP7 significantly inhibited SKRC10 cell growth (p < 0.05) and resulted in a significant reduction of the invasive potential (p < 0.01), but did not cause growth inhibition of SKRC7 cells. We found that The Cancer Genome Atlas Research Network (TCGA) database shows FABP6 and 7 as equally overexpressed in the FABP family. Functional suppression of fatty acid binding protein 6 (FABP6) resulted in significant growth inhibition of SKRC7 cells (p < 0.005). CONCLUSIONS: Functional suppression of FABP7 significantly reduced cell viability and invasive potential in a ccRCC cell line. FABP7 may play a role in progression in some metastatic ccRCCs. The suppressed function may be compensated by another FABP family member.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/patología , Proteína de Unión a los Ácidos Grasos 7/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Hormonas Gastrointestinales/metabolismo , Neoplasias Renales/patología , Proteínas Supresoras de Tumor/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/genética , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Línea Celular Tumoral , Supervivencia Celular , Progresión de la Enfermedad , Proteína de Unión a los Ácidos Grasos 7/antagonistas & inhibidores , Proteína de Unión a los Ácidos Grasos 7/genética , Proteínas de Unión a Ácidos Grasos/genética , Femenino , Hormonas Gastrointestinales/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Humanos , Riñón/patología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida/métodos , Invasividad Neoplásica/patología , Supervivencia sin Progresión , ARN Interferente Pequeño/metabolismo , Tasa de Supervivencia , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda