Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 168
Filtrar
1.
Cell Rep Med ; 3(12): 100825, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36513070

RESUMEN

Sperm chemotaxis is required for guiding sperm toward the egg. However, the molecular identity of physiological chemoattractant and its involvement in infertility remain elusive. Here, we identify DEFB19/119 (mouse/human orthologs) as a physiological sperm chemoattractant. The epithelia of the female reproductive tract and the cumulus-oocyte complex secrete DEFB19/119 that elicits calcium mobilization via the CatSper channel and induces sperm chemotaxis in capacitated sperm. Manipulating the level of DEFB19 in mice determines the number of sperm arriving at the fertilization site. Importantly, we identify exon mutations in the DEFB119 gene in idiopathic infertile women with low level of DEFB119 in the follicular fluid. The level of DEFB119 correlates with the chemotactic potency of follicular fluid and predicts the infertile outcome with positive correlation. This study reveals the pivotal role of DEFB19/119 in sperm chemotaxis and demonstrates its potential application in the diagnosis of idiopathic infertility.


Asunto(s)
Infertilidad Femenina , beta-Defensinas , Humanos , Masculino , Femenino , Animales , Ratones , Quimiotaxis/fisiología , Semen/metabolismo , Espermatozoides/metabolismo , Factores Quimiotácticos/metabolismo
2.
Transl Cancer Res ; 11(3): 436-443, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35402186

RESUMEN

Background: The role of cystic fibrosis transmembrane conductance regulator (CFTR) in hematopoiesis and adult leukemia has been demonstrated using a zebrafish model and leukemia cell lines in our previous works. Here, we continue to explore the association between CFTR and human childhood B-cell acute lymphoblastic leukemia (B-ALL). Methods: We continued to collect the peripheral blood and bone marrows of human childhood patients diagnosed with primary B-ALL as well as non-leukemia controls and isolated lymphocytes for analysis using western blotting and quantitative real-time polymerase chain reaction (qPCR) assay. Then, we used immunofluorescence, co-immunoprecipitation, western blotting, luciferase, 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assays to identify the interaction of CFTR with Wnt signaling in B-ALL. Finally, we established B-ALL xenograft model in non-obese diabetic/severe combined immunodeficiency (NOD/SCID) mice using SUP-B15 cells, and examined whether the CFTR inhibitor CFTR-inh172 could active against SUP-B15-Dependent B-ALL in vivo. Results: Highly expressed CFTR protein and mRNA are associated with primary childhood B-ALL patients. Aberrantly upregulated CFTR and Wnt signaling, our previously reported CFTR-Dvl2-ß-catenin pathway, is found in human childhood B-ALL patients. Interference with CFTR in B-ALL cell lines induces the downregulation of DVL2/ß-catenin and Wnt downstream target accompanied by a reduction of cell proliferation. Furthermore, B-ALL cell lines SUP-B15 cell-transplanted NOD/SCID mice treated with CFTR inhibitor CFTRinh-172 had significantly longer survival and slower leukemia progression compared with mice treated with vehicle dimethyl sulfoxide (DMSO). Conclusions: These findings demonstrate that highly expressed CFTR is associated with human childhood B-ALL and the potential of CFTR inhibitor CFTR-inh172 for the treatment of human B-ALL.

3.
Bioact Mater ; 8: 95-108, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34541389

RESUMEN

Magnesium metal and its alloys are being developed as effective orthopedic implants; however, the mechanisms underlying the actions of magnesium on bones remain unclear. Cystic fibrosis, the most common genetic disease in Caucasians caused by the mutation of CFTR, has shown bone disorder as a key clinical manifestation, which currently lacks effective therapeutic options. Here we report that implantation of magnesium-containing implant stimulates bone formation and improves bone fracture healing in CFTR-mutant mice. Wnt/ß-catenin signaling in the bone is enhanced by the magnesium implant, and inhibition of Wnt/ß-catenin by iCRT14 blocks the magnesium implant to improve fracture healing in CFTR-mutant mice. We further demonstrate that magnesium ion enters osteocytes, increases intracellular cAMP level and activates ATF4, a key transcription factor known to regulate Wnt/ß-catenin signaling. In vivo knockdown of ATF4 abolishes the magnesium implant-activated ß-catenin in bones and reverses the improved-fracture healing in CFTR-mutant mice. In addition, oral supplementation of magnesium activates ATF4 and ß-catenin as well as enhances bone volume and density in CFTR-mutant mice. Together, these results show that magnesium implantation or supplementation may serve as a potential anabolic therapy for cystic fibrosis-related bone disease. Activation of ATF4-dependent Wnt/ß-catenin signaling in osteocytes is identified as a previously undefined mechanism underlying the beneficial effect of magnesium on bone formation.

4.
Endocr Connect ; 11(1)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34825893

RESUMEN

Objective: The beneficial effect of angiotensin(1-7) (Ang(1-7)), via the activation of its receptor, MAS-1, has been noted in diabetes treatment; however, how Ang(1-7) or MAS-1 affects insulin secretion remains elusive and whether the endogenous level of Ang(1-7) or MAS-1 is altered in diabetic individuals remains unexplored. We recently identified an important role of cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated Cl- channel, in the regulation of insulin secretion. Here, we tested the possible involvement of CFTR in mediating Ang(1-7)'s effect on insulin secretion and measured the level of Ang(1-7), MAS-1 as well as CFTR in the blood of individuals with or without type 2 diabetes. Methods: Ang(1-7)/MAS-1/CFTR pathway was determined by specific inhibitors, gene manipulation, Western blotting as well as insulin ELISA in a pancreatic ß-cell line, RINm5F. Human blood samples were collected from 333 individuals with (n = 197) and without (n = 136) type 2 diabetes. Ang(1-7), MAS-1 and CFTR levels in the human blood were determined by ELISA. Results: In RINm5F cells, Ang(1-7) induced intracellular cAMP increase, cAMP-response element binding protein (CREB) activation, enhanced CFTR expression and potentiated glucose-stimulated insulin secretion, which were abolished by a selective CFTR inhibitor, RNAi-knockdown of CFTR, or inhibition of MAS-1. In human subjects, the blood levels of MAS-1 and CFTR, but not Ang(1-7), were significantly higher in individuals with type 2 diabetes as compared to those in non-diabetic healthy subjects. In addition, blood levels of MAS-1 and CFTR were in significant positive correlation in type-2 diabetic but not non-diabetic subjects. Conclusion: These results suggested that MAS-1 and CFTR as key players in mediating Ang(1-7)-promoted insulin secretion in pancreatic ß-cells; MAS-1 and CFTR are positively correlated and both upregulated in type 2 diabetes.

5.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166242, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34389474

RESUMEN

Endoplasmic reticulum (ER) stress, with adaptive unfolded protein response (UPR), is a key link between obesity, insulin resistance and type 2 diabetes, all of which are often present in the most common endocrine-metabolic disorder in women of reproductive age, polycystic ovary syndrome (PCOS), which is characterized with hyperandrogenism. However, the link between excess androgen and endoplasmic reticulum (ER) stress/insulin resistance in patients with polycystic ovary syndrome (PCOS) is unknown. An unexpected role of kisspeptin was reported in the regulation of UPR pathways and its involvement in the androgen-induced ER stress in hypothalamic neuronal cells. To evaluate the relationship of kisspeptin and ER stress, we detected kisspeptin and other factors in blood plasm of PCOS patients, rat models and hypothalamic neuronal cells. We detected higher testosterone and lower kisspeptin levels in the plasma of PCOS than that in non-PCOS women. We established a PCOS rat model by dihydrotestosterone (DHT) chronic exposure, and observed significantly downregulated kisspeptin expression and activated UPR pathways in PCOS rat hypothalamus compared to that in controls. Inhibition or knockdown of kisspeptin completely mimicked the enhancing effect of DHT on UPR pathways in a hypothalamic neuronal cell line, GT1-7. Kp10, the most potent peptide of kisspeptin, effectively reversed or suppressed the activated UPR pathways induced by DHT or thapsigargin, an ER stress activator, in GT1-7 cells, as well as in the hypothalamus in PCOS rats. Similarly, kisspeptin attenuated thapsigargin-induced Ca2+ response and the DHT- induced insulin resistance in GT1-7 cells. Collectively, the present study has revealed an unexpected protective role of kisspeptin against ER stress and insulin resistance in the hypothalamus and has provided a new treatment strategy targeting hypothalamic ER stress and insulin resistance with kisspeptin as a potential therapeutic agent.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Kisspeptinas/sangre , Neuronas/metabolismo , Síndrome del Ovario Poliquístico/genética , Andrógenos/efectos adversos , Animales , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Femenino , Hipotálamo/metabolismo , Hipotálamo/patología , Resistencia a la Insulina/genética , Kisspeptinas/genética , Neuronas/patología , Obesidad/metabolismo , Obesidad/patología , Síndrome del Ovario Poliquístico/sangre , Síndrome del Ovario Poliquístico/patología , Ratas , Testosterona/sangre , Respuesta de Proteína Desplegada/genética
6.
Mech Dev ; 163: 103627, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32574800

RESUMEN

Mutations in the CFTR gene cause cystic fibrosis (CF) with myocardial dysfunction. However, it remains unknown whether CF-related heart disease is a secondary effect of pulmonary disease, or an intrinsic primary defect in the heart. Here, we used zebrafish, which lack lung tissue, to investigate the role of CFTR in cardiogenesis. Our findings demonstrated that the loss of CFTR impairs cardiac development from the cardiac progenitor stage, resulting in cardiac looping defects, a dilated atrium, pericardial edema, and a decrease in heart rate. Furthermore, we found that cardiac development was perturbed in wild-type embryos treated with a gating-specific CFTR channel inhibitor, CFTRinh-172, at the blastula stage of development, but not at later stages. Gene expression analysis of blastulas indicated that transcript levels, including mRNAs associated with cardiovascular diseases, were significantly altered in embryos derived from cftr mutants relative to controls. To evaluate the role of CFTR in human heart failure, we performed a genetic association study on individuals with dilated cardiomyopathy and found that the I556V mutation in CFTR, which causes a channel defect, was associated with the disease. Similar to other well-studied channel-defective CFTR mutants, CFTR I556V mRNA failed to restore cardiac dysplasia in mutant embryos. The present study revealed an important role for the CFTR ion channel in regulating cardiac development during early embryogenesis, supporting the hypothesis that CF-related heart disease results from an intrinsic primary defect in the heart.


Asunto(s)
Cardiomiopatía Dilatada/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Corazón/crecimiento & desarrollo , Pez Cebra/genética , Animales , Cardiomiopatía Dilatada/fisiopatología , Modelos Animales de Enfermedad , Embrión no Mamífero/metabolismo , Embrión no Mamífero/fisiopatología , Desarrollo Embrionario/genética , Corazón/fisiopatología , Humanos , Mutación/genética , Pez Cebra/crecimiento & desarrollo
7.
J Cell Mol Med ; 24(13): 7301-7312, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32463592

RESUMEN

Cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated Cl- channel, is extensively expressed in the epithelial cells of various tissues and organs. Accumulating evidence indicates that aberrant expression or mutation of CFTR is related to carcinoma development. Malignant gliomas are the most common and aggressive intracranial tumours; however, the role of CFTR in the development of malignant gliomas is unclear. Here, we report that CFTR is expressed in malignant glioma cell lines. Suppression of CFTR channel function or knockdown of CFTR suppresses glioma cell viability whereas overexpression of CFTR promotes it. Additionally, overexpression of CFTR suppresses apoptosis and promotes glioma progression in both subcutaneous and orthotopic xenograft models. Cystic fibrosis transmembrane conductance regulator activates Akt/Bcl2 pathway, and suppression of PI3K/Akt pathway abolishes CFTR overexpression-induced up-regulation of Bcl2 (MK-2206 and LY294002) and cell viability (MK-2206). More importantly, the protein expression level of CFTR is significantly increased in glioblastoma patient samples. Altogether, our study has revealed a mechanism by which CFTR promotes glioma progression via up-regulation of Akt/Bcl2-mediated anti-apoptotic pathway, which warrants future studies into the potential of using CFTR as a therapeutic target for glioma treatment.


Asunto(s)
Apoptosis/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Glioma/genética , Glioma/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Regulación hacia Arriba/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Desnudos
8.
Sci China Life Sci ; 63(3): 388-400, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31197761

RESUMEN

The pathophysiology of preeclampsia (PE) remains unclear. PE spiral artery remodeling dysfunction and PE offspring cardiovascular future development has been a worldwide concern. We collected placental and umbilical artery samples from nor-motensive and PE pregnancies. Mineralocorticoid receptor (MR) and its alternative splicing variant (ASV) expression and their biological effects on PE were examined. An MR ASV was found to be highly expressed in all PE samples and slightly expressed in about half of the normotensive samples (umbilical artery, ~57.58%; placenta, ~36.84%). The MR ASV expression was positively associated with blood pressure in both groups. The MR ASV protein changed the aldosterone-induced expression pattern of MR target genes related to ion exchanges and cell signaling pathways. The MR ASV can also impair the proliferation, migration, and tube formation ability of endothelial cells. These findings indicate that MR ASV in PE placenta plays a pathogenic role in PE pathophysiology, especially in endothelial dysfunction, and the existence of the MR ASV in PE umbilical artery provides a new direction in the study of PE offspring with increased risk of cardiovascular diseases.


Asunto(s)
Empalme Alternativo/genética , Preeclampsia/tratamiento farmacológico , Receptores de Mineralocorticoides/metabolismo , Enfermedades Vasculares/tratamiento farmacológico , Adulto , Aldosterona/metabolismo , Presión Sanguínea , ADN Complementario/metabolismo , Células Endoteliales/metabolismo , Femenino , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Placenta/metabolismo , Factor de Crecimiento Placentario , Embarazo , Proteínas Gestacionales , ARN/metabolismo , Receptores de Mineralocorticoides/genética , Factores de Riesgo , Enfermedades Vasculares/metabolismo
9.
iScience ; 21: 448-457, 2019 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-31707258

RESUMEN

Ultrasound brain stimulation is a promising modality for probing brain function and treating brain disease non-invasively and with high spatiotemporal resolution. However, the mechanism underlying its effects remains unclear. Here, we examine the role that the mouse piezo-type mechanosensitive ion channel component 1 (Piezo1) plays in mediating the in vitro effects of ultrasound in mouse primary cortical neurons and a neuronal cell line. We show that ultrasound alone could activate heterologous and endogenous Piezo1, initiating calcium influx and increased nuclear c-Fos expression in primary neurons but not when pre-treated with a Piezo1 inhibitor. We also found that ultrasound significantly increased the expression of the important proteins phospho-CaMKII, phospho-CREB, and c-Fos in a neuronal cell line, but Piezo1 knockdown significantly reduced this effect. Our findings demonstrate that the activity of mechanosensitive ion channels such as Piezo1 stimulated by ultrasound is an important contributor to its ability to stimulate cells in vitro.

10.
iScience ; 21: 375-390, 2019 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-31704649

RESUMEN

Epigenomic changes and stem cell deterioration are two hallmarks of aging. Accumulating evidence suggest that senescence of mesenchymal stromal cells (MSCs) perpetuates aging or age-related diseases. Here we report that two H3K9 demethylases, KDM3A and KDM4C, regulate heterochromatin reorganization via transcriptionally activating condensin components NCAPD2 and NCAPG2 during MSC senescence. Suppression of KDM3A or KDM4C by either genetic or biochemical approach leads to robust DNA damage response and aggravates cellular senescence, whereas overexpression of KDM3A/KDM4C or NCAPD2 promotes heterochromatin reorganization and blunts DNA damage response. Moreover, MSCs derived from Kdm3a-/- mice exhibit defective chromosome organization and exacerbated DNA damage response, which are associated with accelerated bone aging. Consistently, analysis of human bone marrow MSCs and transcriptome database reveals inverse correlation of KDM3A/KDM4C and/or NCAPD2/NCAPG2 with aging. Taken together, the present finding unveils that H3K9 demethylases function as a surveillance mechanism to restrain DNA damage accumulation in stem cells during aging.

11.
Leuk Res ; 86: 106225, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31541940

RESUMEN

Our previous studies have demonstrated that a previously unrecognized role of CFTR in hematopoiesis and acute leukemia. Here, we show that CFTR inhibitor CFTR-inh172 possesses ability to inhibit human T-cell acute lymphoblastic leukemia cells. In detail, CFTR-inh172 inhibited cell proliferation, promoted apoptosis and arrested the cell cycle in human T-cell acute lymphoblastic leukemia cell CCRF-CEM, JURKAT and MOLT-4. Furthermore, transcriptome analysis reveals that CFTR-inh172 induces significant alteration of gene expression related to apoptosis and proliferation. These findings demonstrate the potential of CFTR inhibitor CFTR-inh172 in human T-cell acute lymphoblastic leukemia treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzoatos/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulación de la Expresión Génica/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Tiazolidinas/farmacología , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Transducción de Señal , Transcriptoma , Células Tumorales Cultivadas
12.
Theranostics ; 9(17): 5049-5064, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31410201

RESUMEN

Rationale: Abnormal Wnt/ß-catenin signaling in the endometrium can lead to both embryo implantation failure and severe pathogenic changes of the endometrium such as endometrial cancer and endometriosis. However, how Wnt/ß-catenin signaling is regulated in the endometrium remains elusive. We explored possible regulation of Wnt/ß-catenin signaling by multi-drug resistance protein 4 (MRP4), a potential target in cancer chemotherapy, and investigated the mechanism. Methods: Knockdown of MRP4 was performed in human endometrial cells in vitro or in a mouse embryo-implantation model in vivo. Immunoprecipitation, immunoblotting and immunofluorescence were used to assess protein interaction and stability. Wnt/ß-catenin signaling was assessed by TOPflash reporter assay and quantitative PCR array. Normal and endometriotic human endometrial tissues were examined. Data from human microarray or RNAseq databases of more than 100 participants with endometriosis, endometrial cancer or IVF were analyzed. In vitro and in vivo tumorigenesis was performed. Results: MRP4-knockdown, but not its transporter-function-inhibition, accelerates ß-catenin degradation in human endometrial cells. MRP4 and ß-catenin are co-localized and co-immunoprecipitated in mouse and human endometrium. MRP4-knockdown in mouse uterus reduces ß-catenin levels, downregulates a series of Wnt/ß-catenin target genes and impairs embryo implantation, which are all reversed by blocking ß-catenin degradation. Analysis of human endometrial biopsy samples and available databases reveals significant and positive correlations of MRP4 with ß-catenin and Wnt/ß-catenin target genes in the receptive endometrium in IVF, ectopic endometriotic lesions and endometrial cancers. Knockdown of MRP4 also inhibits in vitro and in vivo endometrial tumorigenesis. Conclusion: A previously undefined role of MRP4 in stabilizing ß-catenin to sustain Wnt/ß-catenin signaling in endometrial cells is revealed for both embryo implantation and endometrial disorders, suggesting MRP4 as a theranostic target for endometrial diseases associated with Wnt/ß-catenin signaling abnormality.


Asunto(s)
Neoplasias Endometriales/metabolismo , Endometriosis/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Embarazo/metabolismo , Vía de Señalización Wnt , Adulto , Animales , Línea Celular Tumoral , Endometrio/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos ICR , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , beta Catenina/metabolismo
13.
J Clin Med ; 8(7)2019 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-31336927

RESUMEN

Human cardiac progenitor cells isolated from the same host may have advantages over other sources of stem cells. The aim of this study is to establish a new source of human progenitor cells collected from a waste product, pericardiac effusion fluid, after open-heart surgery in children with congenital heart diseases. The fluid was collected every 24 h for 2 days after surgery in 37 children. Mononuclear cells were isolated and expanded in vitro. These pericardial effusion-derived progenitor cells (PEPCs) exhibiting cardiogenic lineage markers, were highly proliferative and enhanced angiogenesis in vitro. Three weeks after stem cell transplantation into the ischemic heart in mice, cardiac ejection fraction was improved significantly without detectable progenitor cells. Gene expression profiles of the repaired hearts revealed activation of several known repair mechanisms including paracrine effects, cell migration, and angiogenesis. These progenitor cells may have the potential for heart regeneration.

14.
EMBO Mol Med ; 10(10)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30154237

RESUMEN

The shift of cytokine profile from anti- to pro-inflammatory is the most recognizable sign of labor, although the underlying mechanism remains elusive. Here, we report that the epithelial sodium channel (ENaC) is upregulated and activated in the uterus at labor in mice. Mechanical activation of ENaC results in phosphorylation of CREB and upregulation of pro-inflammatory cytokines as well as COX-2/PGE2 in uterine epithelial cells. ENaC expression is also upregulated in mice with RU486-induced preterm labor as well as in women with preterm labor. Interference with ENaC attenuates mechanically stimulated uterine contractions and significantly delays the RU486-induced preterm labor in mice. Analysis of a human transcriptome database for maternal-fetus tissue/blood collected at onset of human term and preterm births reveals significant and positive correlation of ENaC with labor-associated pro-inflammatory factors in labored birth groups (both term and preterm), but not in non-labored birth groups. Taken together, the present finding reveals a pro-inflammatory role of ENaC in labor at term and preterm, suggesting it as a potential target for the prevention and treatment of preterm labor.


Asunto(s)
Citocinas/metabolismo , Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Trabajo de Parto , Animales , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Modelos Animales , Embarazo , Útero/fisiología
15.
Reproduction ; 156(3): 261-268, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29930176

RESUMEN

Mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene affect fertility in both sexes. However, the involvement of CFTR in regulating germ cell development remains largely unknown. Here, we used zebrafish model to investigate the role of CFTR in primordial germ cells (PGCs) development. We generated a cftr frameshift mutant zebrafish line using CRISPR/Cas9 technique and investigated the migration of PGCs during early embryo development. Our results showed that loss of Cftr impairs the migration of PGCs from dome stages onward. The migration of PGCs was also perturbed by treatment of CFTRinh-172, a gating-specific CFTR channel inhibitor. Moreover, defected PGCs migration in cftr mutant embryos can be partially rescued by injection of WT but not other channel-defective mutant cftr mRNAs. Finally, we observed the elevation of cxcr4b, cxcl12a, rgs14a and ca15b, key factors involved in zebrafish PGCs migration, in cftr-mutant zebrafish embryos. Taken together, the present study revealed an important role of CFTR acting as an ion channel in regulating PGCs migration during early embryogenesis. Defect of which may impair germ cell development through elevation of key factors involved in cell motility and response to chemotactic gradient in PGCs.


Asunto(s)
Movimiento Celular/fisiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Embrión no Mamífero/fisiología , Células Germinativas/fisiología , Pez Cebra/embriología , Animales , Secuencia de Bases , Regulador de Conductancia de Transmembrana de Fibrosis Quística/antagonistas & inhibidores , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Desarrollo Embrionario , Mutación del Sistema de Lectura , Pez Cebra/genética , Pez Cebra/fisiología
16.
Proteomics Clin Appl ; 12(6): e1700162, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29781578

RESUMEN

PURPOSE: A previous study reported an elevated inflammation during tendon injury in mice with cystic fibrosis (CF), indicating the inadequate management of inflammation due to dysfunction of the cystic fibrosis transmembrane conductance regulator (CFTR). The objective of this study is to identify the targets of CFTR that contribute to the abnormal inflammation during tendon injury. EXPERIMENTAL DESIGN: A 2D gel electrophoresis and mass-spectrometry-based comparative proteomics is performed to find the molecular targets of CFTR. And the targeted protein is further confirmed at both mRNA and protein levels. RESULTS: It is identified that 14 proteins are differentially expressed, with annexin A1 being one of the most significantly downregulated protein. Further confirmation shows that annexin A1 is significantly decreased in TDSCs isolated from DF508 mice. As an essential anti-inflammation mediator, it is also downregulated in the injured tendon tissue of DF508 mice when compared with WT mice. CONCLUSIONS AND CLINICAL RELEVANCE: Decreased annexin A1 expression can contribute to the elevated inflammation in DF508 mice during tendon injury. Therefore, annexin A1 can be considered as a new potential biomarker or drug target for a possible therapeutic approach in clinical practice.


Asunto(s)
Anexina A1/genética , Diferenciación Celular/genética , Fibrosis Quística/genética , Inflamación/genética , Animales , Fibrosis Quística/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Espectrometría de Masas , Ratones , Proteómica/métodos , Transducción de Señal/genética , Células Madre/citología , Tendones/citología
17.
Cell Death Dis ; 9(3): 275, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29449653

RESUMEN

Mutations of cystic fibrosis transmembrane conductance regulator (CFTR) cause cystic fibrosis (CF) with a multitude of clinical manifestations. Some CF patients develop clinically significant anemia, suggesting that CFTR may regulate hematopoiesis. Here, we report that cftr mutant zebrafish model exhibits primitive and definitive hematopoietic defects with impaired Wnt signaling. Cftr is found to interact, via its PDZ-binding domain (PDZBD), with Dishevelled (Dvl), a key component of Wnt signaling required for hematopoietic progenitor specification, thus protecting Dvl from Dapper1 (Dpr1)-induced lysosomal degradation. Defective hematopoiesis and impaired Wnt signaling in cftr mutant can be rescued by overexpression of wild-type or channel function-defective G551D mutant CFTR with an intact PDZBD, but not Cftr with mutations in the PDZBD. Analysis of human database ( http://r2.amc.nl ) shows that CFTR is positively correlated with DVL2 and Wnt-related hematopoietic factors in human blood system. The results reveal a previously unrecognized role of CFTR, which is independent of its channel function, in regulating DVL degradation and thus Wnt signaling required for hematopoiesis in both zebrafish and humans, providing an explanation for the anemic phenotype of CF patients.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Hematopoyesis , Proteínas de la Membrana/metabolismo , Mutación , Vía de Señalización Wnt , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Proteínas Dishevelled/genética , Proteínas Dishevelled/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Dominios PDZ , Proteolisis , Pez Cebra/embriología , Pez Cebra/genética , beta Catenina/genética , beta Catenina/metabolismo
18.
Cell Mol Life Sci ; 75(10): 1737-1756, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29411041

RESUMEN

Mutations of cystic fibrosis transmembrane conductance regulator (CFTR) cause cystic fibrosis, the most common life-limiting recessive genetic disease among Caucasians. CFTR mutations have also been linked to increased risk of various cancers but remained controversial for a long time. Recent studies have begun to reveal that CFTR is not merely an ion channel but also an important regulator of cancer development and progression with multiple signaling pathways identified. In this review, we will first present clinical findings showing the correlation of genetic mutations or aberrant expression of CFTR with cancer incidence in multiple cancers. We will then focus on the roles of CFTR in fundamental cellular processes including transformation, survival, proliferation, migration, invasion and epithelial-mesenchymal transition in cancer cells, highlighting the signaling pathways involved. Finally, the association of CFTR expression levels with patient prognosis, and the potential of CFTR as a cancer prognosis indicator in human malignancies will be discussed.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Neoplasias/genética , Neoplasias/patología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Humanos
19.
Biochim Biophys Acta Mol Cell Res ; 1865(4): 605-615, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29326073

RESUMEN

Retinoic acid (RA) plays a pivotal role in many cellular processes; however, the signaling mechanisms mediating the effect of RA are not fully understood. Here, we show that RA transcriptionally upregulates cystic fibrosis transmembrane conductance regulator (Cftr) by promoting the direct binding of its receptor RARα to Cftr promoter in mouse spermatogonia and embryonic stem (ES) cells. The RA/CFTR pathway is involved in the differentiation of spermatogonia and organogenesis during the embryo development of Xenopus laevis. Loss of CFTR by siRNA-mediated knockdown blunts the RA-induced spermatogonial differentiation. Overexpression of CFTR mimics the effect of RA on the induction of spermatogonial differentiation or restores the developmental defects induced by the knockdown of RARα in spermatogonial cells and Xenopus laevis. Analysis of the human database shows that the expression of CFTR positively correlates with RARα in brain tissues, stem cells as well as cancers, supporting the role of RA/CFTR pathway in various developmental processes in humans. Together, our study discovers an essential role of CFTR in mediating the RA-dependent signaling for stem cell differentiation and embryonic development.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Desarrollo Embrionario/efectos de los fármacos , Células Madre/citología , Células Madre/metabolismo , Transcripción Genética/efectos de los fármacos , Tretinoina/farmacología , Xenopus laevis/embriología , Animales , Secuencia de Bases , Línea Celular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos C57BL , Receptor alfa de Ácido Retinoico/metabolismo , Transducción de Señal/efectos de los fármacos , Espermatogonias/citología , Células Madre/efectos de los fármacos , Xenopus laevis/metabolismo , beta Catenina/metabolismo
20.
Oncotarget ; 8(53): 91445-91458, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29207656

RESUMEN

CatSper channel has been considered the principal sperm Ca2+ channel responsible for the cytosolic Ca2+ elevation required for various sperm functions necessary for fertilization [1-4]. However, the mechanism underlying the activation of CatSper channel by various physiological ligands remain incompletely understood. We have recently demonstrated the expression of C-C chemokine receptor 6 (CCR6) in sperm and Ca2+ influx upon binding of human ß-defensin 1 (DEFB1) to CCR6, which is important for sperm motility [5]. In the present study, we have demonstrated that CCR6 receptor and CatSper channel are both required for the Ca2+ entry/current induced by physiological ligands DEFB1, chemokine (C-C motif) ligand 20 (CCL20) and progesterone in human sperm. CCR6 is co-localized and interacts with CatSper in human sperm. Ca2+ influx mediated by CCR6 and CatSper is required for essential sperm functions, including motility, hyperactivation and acrosome reaction, which are impaired in infertile sperm showing reduced levels of CCR6 and CatSper. The present finding suggests a critical role of CCR6 receptor in mediating ligand-induced, CatSper-dependent Ca2+ influx required for various sperm functions and thus male fertility.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...