Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
EMBO J ; 36(4): 441-457, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28003315

ABSTRACT

The transport protein particle (TRAPP) was initially identified as a vesicle tethering factor in yeast and as a guanine nucleotide exchange factor (GEF) for Ypt1/Rab1. In mammals, structures and functions of various TRAPP complexes are beginning to be understood. We found that mammalian TRAPPII was a GEF for both Rab18 and Rab1. Inactivation of TRAPPII-specific subunits by various methods including siRNA depletion and CRISPR-Cas9-mediated deletion reduced lipolysis and resulted in aberrantly large lipid droplets. Recruitment of Rab18 onto lipid droplet (LD) surface was defective in TRAPPII-deleted cells, but the localization of Rab1 on Golgi was not affected. COPI regulates LD homeostasis. We found that the previously documented interaction between TRAPPII and COPI was also required for the recruitment of Rab18 to the LD We hypothesize that the interaction between COPI and TRAPPII helps bring TRAPPII onto LD surface, and TRAPPII, in turn, activates Rab18 and recruits it on the LD surface to facilitate its functions in LD homeostasis.


Subject(s)
Coat Protein Complex I/metabolism , Lipid Droplets/metabolism , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Cell Line , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Lipolysis , Vesicular Transport Proteins/genetics , rab1 GTP-Binding Proteins/metabolism
2.
J Cell Mol Med ; 24(13): 7301-7312, 2020 07.
Article in English | MEDLINE | ID: mdl-32463592

ABSTRACT

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.


Subject(s)
Apoptosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Glioma/genetics , Glioma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Up-Regulation/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude
3.
Biochim Biophys Acta Mol Cell Res ; 1865(4): 605-615, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29326073

ABSTRACT

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.


Subject(s)
Cell Differentiation/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Embryonic Development/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Xenopus laevis/embryology , Animals , Base Sequence , Cell Line , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Humans , Male , Mice, Inbred C57BL , Retinoic Acid Receptor alpha/metabolism , Signal Transduction/drug effects , Spermatogonia/cytology , Stem Cells/drug effects , Xenopus laevis/metabolism , beta Catenin/metabolism
4.
Cell Mol Life Sci ; 75(10): 1737-1756, 2018 05.
Article in English | MEDLINE | ID: mdl-29411041

ABSTRACT

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.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Neoplasms/genetics , Neoplasms/pathology , Animals , Cell Movement/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Humans
5.
Reproduction ; 156(3): 261-268, 2018 09.
Article in English | MEDLINE | ID: mdl-29930176

ABSTRACT

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.


Subject(s)
Cell Movement/physiology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Embryo, Nonmammalian/physiology , Germ Cells/physiology , Zebrafish/embryology , Animals , Base Sequence , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Embryonic Development , Frameshift Mutation , Zebrafish/genetics , Zebrafish/physiology
6.
FASEB J ; 31(9): 3800-3815, 2017 09.
Article in English | MEDLINE | ID: mdl-28495756

ABSTRACT

Tendons are a mechanosensitive tissue, which enables them to transmit to bone forces that are derived from muscle. Patients with tendon injuries, such as tendinopathy or tendon rupture, were often observed with matrix degeneration, and the healing of tendon injuries remains a challenge as a result of the limited understanding of tendon biology. Our study demonstrates that the stretch-mediated activation channel, cystic fibrosis transmembrane conductance regulator (CFTR), was up-regulated in tendon-derived stem cells (TDSCs) during tenogenic differentiation under mechanical stretching. Tendon tissues in CFTR-dysfunctional DF508 mice exhibited irregular cell arrangement, uneven fibril diameter distribution, weak mechanical properties, and less matrix formation in a tendon defect model. Moreover, both tendon tissues and TDSCs isolated from DF508 mice showed significantly decreased levels of tendon markers, such as scleraxis, tenomodulin, Col1A1 (collagen type I α 1 chain), and decorin Furthermore, by RNA sequencing analysis, we demonstrated that Wnt/ß-catenin signaling was abnormally activated in TDSCs from DF508 mice, thereby further activating the pERK1/2 signaling pathway. Of most importance, we found that intervention in pERK1/2 signaling could promote tenogenic differentiation and tendon regeneration both in vitro and in vivo Taken together, our study demonstrates that CFTR plays an important role in tenogenic differentiation and tendon regeneration by inhibiting the ß-catinin/pERK1/2 signaling pathway. The therapeutic strategy of intervening in the CFTR/ß-catenin/pERK1/2 regulatory axis may be helpful for accelerating tendon injury healing, which has implications for tendon injury management.-Liu, Y., Xu, J., Xu, L., Wu, T., Sun, Y., Lee, Y.-W., Wang, B., Chan, H.-C., Jiang, X., Zhang, J., Li, G. Cystic fibrosis transmembrane conductance regulator mediates tenogenic differentiation of tendon-derived stem cells and tendon repair: accelerating tendon injury healing by intervening in its downstream signaling.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Signal Transduction/physiology , Tendon Injuries/therapy , Tendons/cytology , Animals , Biomechanical Phenomena , Cell Differentiation , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Male , Mice , Mice, Inbred CFTR , Rats , Rats, Sprague-Dawley , Stem Cell Transplantation , Stem Cells/metabolism , Up-Regulation , beta Catenin/genetics , beta Catenin/metabolism
7.
Proc Natl Acad Sci U S A ; 112(15): 4743-8, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25825716

ABSTRACT

Polycystic ovary syndrome (PCOS) is one of the most common female endocrine disorders and a leading cause of female subfertility. The mechanism underlying the pathophysiology of PCOS remains to be illustrated. Here, we identify two alternative splice variants (ASVs) of the androgen receptor (AR), insertion and deletion isoforms, in granulosa cells (GCs) in ∼62% of patients with PCOS. AR ASVs are strongly associated with remarkable hyperandrogenism and abnormalities in folliculogenesis, and are absent from all control subjects without PCOS. Alternative splicing dramatically alters genome-wide AR recruitment and androgen-induced expression of genes related to androgen metabolism and folliculogenesis in human GCs. These findings establish alternative splicing of AR in GCs as the major pathogenic mechanism for hyperandrogenism and abnormal folliculogenesis in PCOS.


Subject(s)
Alternative Splicing , INDEL Mutation , Polycystic Ovary Syndrome/genetics , Receptors, Androgen/genetics , Adult , Base Sequence , Cells, Cultured , Dehydroepiandrosterone/blood , Female , Gene Expression Profiling , Genome-Wide Association Study , Granulosa Cells/metabolism , HEK293 Cells , Humans , Hyperandrogenism/blood , Hyperandrogenism/genetics , Oogenesis/genetics , Ovarian Follicle/physiopathology , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/physiopathology , Protein Isoforms/genetics , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/blood
8.
Clin Exp Pharmacol Physiol ; 44 Suppl 1: 78-85, 2017 12.
Article in English | MEDLINE | ID: mdl-28345252

ABSTRACT

The cAMP/PKA pathway is one of the most important signalling pathways widely distributed in most eukaryotic cells. The activation of the canonical cAMP/PKA pathway depends on transmembrane adenylyl cyclase (tmAC). Recently, soluble adenylyl cyclase (sAC), which is activated by HCO3- or Ca2+ , emerges to provide an alternative way to activate cAMP/PKA pathway with the cystic fibrosis transmembrane conductance regulator (CFTR), a cAMP-activated Cl- /HCO3- -conducting anion channel, as a key player. This review summarizes new progress in the investigation of the CFTR/HCO3- -dependent sAC signalling and its essential role in various reproductive processes, particularly in ovarian functions. We present the evidence for a CFTR/HCO3- -dependent nuclear sAC signalling cascade that amplifies the FSH-stimulated cAMP/PKA pathway, traditionally thought to involve tmAC, in granulosa for the regulation of oestrogen production and granulosa cell proliferation. The implication of the CFTR/HCO3- /sAC pathway in amplifying other receptor-activated cAMP/PKA signalling in a wide variety of cell types and pathophysiological processes, including aging, is also discussed.


Subject(s)
Adenylyl Cyclases/metabolism , Cell Nucleus/enzymology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Follicle Stimulating Hormone, Human/metabolism , Ovary/enzymology , Animals , Bicarbonates/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Humans , Ovary/physiopathology , Polycystic Ovary Syndrome/enzymology , Polycystic Ovary Syndrome/physiopathology , Second Messenger Systems
9.
Semin Cell Dev Biol ; 29: 31-42, 2014 May.
Article in English | MEDLINE | ID: mdl-24594193

ABSTRACT

Spermatogenesis is a multistep process that supports the production of millions of sperm daily. Understanding of the molecular mechanisms that regulate spermatogenesis has been a major focus for decades. Yet, the regulators involved in different cellular processes of spermatogenesis remain largely unknown. Human diseases that result in defective spermatogenesis have provided hints on the molecular mechanisms regulating this process. In this review, we have summarized recent findings on the function and signaling mechanisms of several genes that are known to be associated with disease or pathological processes, including CFTR, CD147, YWK-II and CT genes, and discuss their potential roles in regulating different processes of spermatogenesis.


Subject(s)
Sertoli Cells/physiology , Spermatogenesis/physiology , Spermatozoa/cytology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Basigin/genetics , Basigin/metabolism , Cell Movement/genetics , Cryptorchidism/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Sertoli Cells/cytology , Signal Transduction
10.
J Cell Sci ; 127(Pt 20): 4396-408, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25107366

ABSTRACT

Mutations in CFTR lead to dysfunction of tubular organs, which is currently attributed to impairment of its conductive properties. We now show that CFTR regulates tight junction assembly and epithelial cell differentiation through modulation of the ZO-1-ZONAB pathway. CFTR colocalizes with ZO-1 at the tight junctions of trachea and epididymis, and is expressed before ZO-1 in Wolffian ducts. CFTR interacts with ZO-1 through the CTFR PDZ-binding domain. In a three-dimensional (3D) epithelial cell culture model, CFTR regulates tight junction assembly and is required for tubulogenesis. CFTR inhibition or knockdown reduces ZO-1 expression and induces the translocation of the transcription factor ZONAB (also known as YBX3) from tight junctions to the nucleus, followed by upregulation of the transcription of CCND1 and downregulation of ErbB2 transcription. The epididymal tubules of cftr(-/-) and cftr(ΔF508) mice have reduced ZO-1 levels, increased ZONAB nuclear expression, and decreased epithelial cell differentiation, illustrated by the reduced expression of apical AQP9 and V-ATPase. This study provides a new paradigm for the etiology of diseases associated with CFTR mutations, including cystic fibrosis.


Subject(s)
Cell Nucleus/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , DNA-Binding Proteins/metabolism , Epithelial Cells/physiology , Tight Junctions/physiology , Transcription Factors/metabolism , Zonula Occludens-1 Protein/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organogenesis/genetics , Protein Binding , Protein Transport/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Zonula Occludens-1 Protein/genetics
11.
Biochim Biophys Acta ; 1843(3): 618-28, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24373847

ABSTRACT

How mutations or dysfunction of CFTR may increase the risk of malignancies in various tissues remains an open question. Here we report the interaction between CFTR and an adherens junction molecule, AF-6/afadin, and its involvement in the development of colon cancer. We have found that CFTR and AF-6/afadin are co-localized at the cell-cell contacts and physically interact with each other in colon cancer cell lines. Knockdown of CFTR results in reduced epithelial tightness and enhanced malignancies, with increased degradation and reduced stability of AF-6/afadin protein. The enhanced invasive phenotype of CFTR-knockdown cells can be completely reversed by either AF-6/afadin over-expression or ERK inhibitor, indicating the involvement of AF-6/MAPK pathway. More interestingly, the expression levels of CFTR and AF-6/afadin are significantly downregulated in human colon cancer tissues and lower expression of CFTR and/or AF-6/afadin is correlated with poor prognosis of colon cancer patients. The present study has revealed a previously unrecognized interaction between CFTR and AF-6/afadin that is involved in the pathogenesis of colon cancer and indicated the potential of the two as novel markers of metastasis and prognostic predictors for human colon cancer.


Subject(s)
Colonic Neoplasms/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Kinesins/genetics , Microfilament Proteins/genetics , Myosins/genetics , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , HEK293 Cells , Humans , Kinesins/metabolism , MAP Kinase Signaling System , Microfilament Proteins/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Myosins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phenotype , Prognosis , Tight Junctions/genetics , Tight Junctions/metabolism , Tight Junctions/pathology
12.
J Cell Physiol ; 230(8): 1906-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25546515

ABSTRACT

Benign prostatic hyperplasia (BPH) is a hyper-proliferative disease of the aging prostate; however, the exact mechanism underlying the development of BPH remains incompletely understood. The present study investigated the possible involvement of the cystic fibrosis transmembrane conductance regulator (CFTR), which has been previously shown to negatively regulate nuclear factor-κB (NF-κB)/cyclooxygenase 2 (COX2)/prostaglandin E2 (PGE2) pathway, in the pathogenesis of BPH. Our results showed decreasing CFTR and increasing COX2 expression in rat prostate tissues with aging. Furthermore, suppression of CFTR led to increased expression of COX2 and over-production of PGE2 in a normal human prostate epithelial cell line (PNT1A) with elevated NF-κB activity. PGE2 stimulated the proliferation of primary rat prostate stromal cells but not epithelial cells, with increased PCNA expression. In addition, the condition medium from PNT1A cells after inhibition or knockdown of CFTR promoted cell proliferation of prostate stromal cells which could be reversed by COX2 or NF-κB inhibitor. More importantly, the involvement of CFTR in BPH was further demonstrated by the down-regulation of CFTR and up-regulation of COX2/NF-κB in human BPH samples. The present results suggest that CFTR may be involved in regulating PGE2 production through its negative regulation on NF-κB/COX2 pathway in prostate epithelial cells, which consequently stimulates cell growth of prostate stromal cells. The overstimulation of prostate stromal cell proliferation by down-regulation of CFTR-enhanced PGE2 production and release during aging may contribute to the development of BPH.


Subject(s)
Aging/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Prostatic Hyperplasia/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cyclooxygenase 2/metabolism , Dinoprostone/biosynthesis , Disease Models, Animal , Down-Regulation , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Male , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transfection
13.
J Cell Physiol ; 230(9): 2049-58, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25641604

ABSTRACT

The physiological role of cystic fibrosis transmembrane conductance regulator (CFTR) in keratinocytes and skin wound healing is completely unknown. The present study shows that CFTR is expressed in the multiple layers of keratinocytes in mouse epidermis and exhibits a dynamic expression pattern in a dorsal skin wound healing model, with diminishing levels observed from day 3 to day 5 and re-appearing from day 7 to day 10 after wounding. Knockdown of CFTR in cultured human keratinocytes promotes cell migration but inhibits differentiation, while overexpression of CFTR suppresses migration but enhances differentiation, indicating an important role of CFTR in regulating keratinocyte behavior. In addition, we have demonstrated a direct association of CFTR with epithelial junction formation as knockdown of CFTR downregulates the expression of adhesion molecules, such as E-cadherin, ZO-1 and ß-catenin, and disrupts the formation of cell junction, while overexpression of CFTR enhances cell junction formation. More importantly, we have shown that ΔF508cftr-/- mice with defective CFTR exhibit delayed wound healing as compared to wild type mice, indicating that normal function of CFTR is critical for wound repair. Taken together, the present study has revealed a previously undefined role of CFTR in regulating skin wound healing processes, which may have implications in injury repair of other epithelial tissues.


Subject(s)
Cell Differentiation/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Skin/metabolism , Wound Healing/genetics , Animals , Cadherins/biosynthesis , Cell Line , Cell Movement/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Epithelium/metabolism , Epithelium/pathology , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Skin/injuries , Skin/pathology , beta Catenin/biosynthesis
14.
Reproduction ; 149(5): 393-401, 2015 May.
Article in English | MEDLINE | ID: mdl-25646509

ABSTRACT

Polycystic ovarian syndrome (PCOS) is one of the most frequent causes of female infertility, featured by abnormal hormone profile, chronic oligo/anovulation, and presence of multiple cystic follicles in the ovary. However, the mechanism underlying the abnormal folliculogenesis remains obscure. We have previously demonstrated that CFTR, a cAMP-dependent Cl(-) and HCO3 (-) conducting anion channel, is expressed in the granulosa cells and its expression is downregulated in PCOS rat models and human patients. In this study, we aimed to investigate the possible involvement of downregulation of CFTR in the impaired follicle development in PCOS using two rat PCOS models and primary culture of granulosa cells. Our results indicated that the downregulation of CFTR in the cystic follicles was accompanied by reduced expression of proliferating cell nuclear antigen (PCNA), in rat PCOS models. In addition, knockdown or inhibition of CFTR in granulosa cell culture resulted in reduced cell viability and downregulation of PCNA. We further demonstrated that CFTR regulated both basal and FSH-stimulated granulosa cell proliferation through the HCO3 (-)/sAC/PKA pathway leading to ERK phosphorylation and its downstream target cyclin D2 (Ccnd2) upregulation. Reduced ERK phosphorylation and CCND2 were found in ovaries of rat PCOS model compared with the control. This study suggests that CFTR is required for normal follicle development and that its downregulation in PCOS may inhibit granulosa cell proliferation, resulting in abnormal follicle development in PCOS.


Subject(s)
Cell Proliferation , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Granulosa Cells/pathology , Ovarian Follicle/pathology , Polycystic Ovary Syndrome/pathology , Animals , Blotting, Western , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Female , Fluorescent Antibody Technique , Granulosa Cells/metabolism , Humans , Mice , Mice, Inbred ICR , Ovarian Follicle/metabolism , Phosphorylation , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
15.
Biochim Biophys Acta ; 1833(12): 2961-2969, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23916755

ABSTRACT

The epithelial-to-mesenchymal transition (EMT), a process involving the breakdown of cell-cell junctions and loss of epithelial polarity, is closely related to cancer development and metastatic progression. While the cystic fibrosis transmembrane conductance regulator (CFTR), a Cl(-) and HCO3(-) conducting anion channel expressed in a wide variety of epithelial cells, has been implicated in the regulation of epithelial polarity, the exact role of CFTR in the pathogenesis of cancer and its possible involvement in EMT process have not been elucidated. Here we report that interfering with CFTR function either by its specific inhibitor or lentiviral miRNA-mediated knockdown mimics TGF-ß1-induced EMT and enhances cell migration and invasion in MCF-7. Ectopic overexpression of CFTR in a highly metastatic MDA-231 breast cancer cell line downregulates EMT markers and suppresses cell invasion and migration in vitro, as well as metastasis in vivo. The EMT-suppressing effect of CFTR is found to be associated with its ability to inhibit NFκB targeting urokinase-type plasminogen activator (uPA), known to be involved in the regulation of EMT. More importantly, CFTR expression is found significantly downregulated in primary human breast cancer samples, and is closely associated with poor prognosis in different cohorts of breast cancer patients. Taken together, the present study has demonstrated a previously undefined role of CFTR as an EMT suppressor and its potential as a prognostic indicator in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Down-Regulation , Epithelial-Mesenchymal Transition , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplasm Invasiveness , Phenotype , Prognosis , Transforming Growth Factor beta1/pharmacology , Urokinase-Type Plasminogen Activator/metabolism
16.
J Cell Sci ; 125(Pt 18): 4219-29, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22641691

ABSTRACT

YWK-II protein/APLP2 is a member of an evolutionarily conserved protein family that includes amyloid precursor protein (APP) and amyloid precursor-like protein-1 (APLP1). We have previously demonstrated that YWK-II/APLP2 functions as a novel G(0)-protein-coupled receptor for Müllerian inhibiting substance (MIS) in cell survival. However, factors regulating the stability and turnover of YWK-II/APLP2 have not been identified. Here we present evidence that human leukocyte antigen-B-associated transcript 3 (Bat3), an important regulator involved in apoptosis, can interact with YWK-II/APLP2 and enhance its stability by reducing its ubiquitylation and degradation by the ubiquitin-proteasome system. Coexpression of different Bat3 domain deletion constructs with YWK-II/APLP2 reveals that the proline-rich domain of Bat3 is required for its binding to YWK-II/APLP2. In addition, we find that the protein levels of YWK-II/APLP2 could be enhanced by nuclear export of Bat3 under apoptotic stimulation. We also find elevated levels of Bat3 and YWK-II/APLP2 in human colorectal cancer with a positive correlation between the two. Taken together, these results have revealed a previously undefined mechanism regulating cell apoptosis and suggest that aberrant enhancement of YWK-II/APLP2 by nuclear export of Bat3 may play a role in cancer development by inhibiting cell apoptosis.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Apoptosis , Molecular Chaperones/metabolism , Nerve Tissue Proteins/metabolism , Active Transport, Cell Nucleus , Animals , CHO Cells , Cell Nucleus/metabolism , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cricetinae , Cricetulus , Female , Gene Knockdown Techniques , HCT116 Cells , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Protein Binding , Protein Stability , Ubiquitination , Up-Regulation , Xenograft Model Antitumor Assays
17.
Reproduction ; 148(6): 559-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25187622

ABSTRACT

In our previous study, we have demonstrated that the epithelial sodium channel (ENaC) mediates the embryo-derived signals leading to the activation of CREB and upregulation of cyclooxygenase type 2 (COX2) required for embryo implantation. This study aims to investigate whether microRNAs (miRNAs) are involved in the ENaC-induced upregulation of COX2 during embryo implantation. The results show that the levels of miR-101 and miR-199a-3p, two COX2 targeting miRNAs, are reduced by ENaC activation, and increased by ENaC inhibition or knock-down of ENaC subunit (ENaCα) in human endometrial surface epithelial (HES) cells or in mouse uteri during implantation. Phosphorylation of CREB is induced by the activation of ENaC, and blocked by ENaC inhibition or knockdown in HES cells. Knockdown of ENaCα or CREB in HES cells or in mouse uterus in vivo results in increases in miR-101 and miR-199a-3p, accompanied with decreases in COX2 protein levels and reduction in implantation rate. The downregulation of COX2 caused by knockdown of ENaC or CREB can be recovered by the inhibitors of miR-101 or miR-199a-3p in HES cells. These results reveal a novel molecular mechanism modulating COX2 expression during embryo implantation via ENaC-dependent CREB activation and COX2-targeting miRNAs.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Embryo Implantation/physiology , Epithelial Sodium Channels/physiology , MicroRNAs/physiology , Animals , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclooxygenase 2/physiology , Embryo, Mammalian/physiology , Endometrium/cytology , Epithelial Cells/cytology , Female , Humans , In Vitro Techniques , Mice , Mice, Inbred ICR , Models, Animal , Phosphorylation/physiology , RNA, Small Interfering/pharmacology , Signal Transduction/physiology
18.
Cell Biol Int ; 38(1): 1-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24115633

ABSTRACT

The solute carrier 26 (SLC26) family emerges as a distinct class of anion transporters with its members SLC26A3 (Slc26a3) and SLC26A6 (Slc26a6) reported to be electrogenic Cl(-)/HCO3(-) exchangers. While it is known that uterine fluid has high HCO3(-) content and that HCO3(-) is essential for sperm capacitation, the molecular mechanisms underlying the transport of HCO3(-) across uterine epithelial cells and sperm have not been fully investigated. The present review re-examines the results from early reports studying anion transport, finding clues for the involvement of Cl(-)/HCO3(-) anion exchangers in electrogenic HCO3(-) transport across endometrial epithelium. We also summarise recent work on Slc26a3 and Slc26a6 in uterine epithelial cells and sperm, revealing their functional role in working closely with the cystic fibrosis transmembrane conductance regulator (CFTR) for HCO3(-) transport in these cells. The possible involvement of these anion exchangers in other HCO3(-) dependent reproductive processes and their implications for infertility are also discussed.


Subject(s)
Anion Transport Proteins/metabolism , Epithelial Cells/metabolism , Spermatozoa/metabolism , Bicarbonates/metabolism , Chlorides/metabolism , Epithelial Cells/cytology , Female , Humans , Male , Uterus/cytology , Uterus/metabolism
19.
Cell Biol Int ; 38(10): 1183-93, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24890676

ABSTRACT

Maternal caffeine exposure may be one of the causes for intrauterine growth retardation and low birth weight (LBW), and increased risk of type 2 diabetes mellitus (T2DM) in the adulthood has been associated with LBW. However, whether maternal caffeine exposure contributes to T2DM development of her offspring has not been fully investigated. We have investigated the influence of maternal caffeine exposure on glucose homeostasis in vivo and effects of long-term caffeine load on insulin secretion of ß-cells. The intake of caffeine during gestation markedly decreases birth weight and postnatal body weight of the offspring. Serum insulin levels of adult offspring after oral glucose tolerance test (OGTT) were significantly lower in the caffeine group compared to the control, although plasma glucose levels were not significantly altered. Proteome analysis of pancreas of adult offspring identified 24 proteins that were differentially expressed between the caffeine and control groups, including proteins involved in energy metabolism. In a rat pancreatic ß-cell line Rin-5f cells, caffeine downregulated expression of one of the proteins involved in insulin synthesis, P4hb, and there was reduced transcriptional expression of insulin. While basal insulin secretion of caffeine-treated cells was elevated, insulin secretion after glucose challenge in long-term caffeine-treated cells was significantly reduced, with increased apoptosis of ß-cells. These results indicate that maternal caffeine exposure may result in potentially abnormal glucose homeostasis and increase the risk of T2DM in the offspring adulthood.


Subject(s)
Caffeine/toxicity , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Maternal Exposure , Prenatal Exposure Delayed Effects , Animals , Apoptosis/drug effects , Birth Weight/drug effects , Blood Glucose/analysis , Cells, Cultured , Diabetes Mellitus, Type 2/etiology , Electrophoresis, Gel, Two-Dimensional , Female , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Pregnancy , Proteome/analysis , Rats , Rats, Sprague-Dawley , Risk Factors
20.
Clin Exp Pharmacol Physiol ; 41(9): 615-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24909068

ABSTRACT

Although microRNAs (miRNAs) have been recognized as one of the important epigenetic mechanisms that regulate gene expression in response to changes in the environment, the links between environmental cues and changes in miRNAs remain largely unknown. Localized to the cell membrane and recognized as an anion channel, the cystic fibrosis transmembrane conductance regulator (CFTR) has recently been shown to mediate important signalling pathways leading to activation of miRNAs. This brief review summarizes the related findings and discusses the emerging role of CFTR as an epigenetic regulator, possibly involved in a wide spectrum of physiological and pathological processes.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , MicroRNAs/genetics , Cell Membrane/physiology , Cues , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Environment , Gene Expression Regulation/physiology , Humans , MicroRNAs/physiology , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL