Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Theranostics ; 12(1): 143-166, 2022.
Article in English | MEDLINE | ID: mdl-34987639

ABSTRACT

Aims: Hypoxic-ischaemic encephalopathy (HIE) is one of the most serious complications in neonates and infants. Mesenchymal stromal cell (MSC)-based therapy is emerging as a promising treatment avenue for HIE. However, despite its enormous potential, the clinical application of MSCs is limited by cell heterogeneity, low isolation efficiency and unpredictable effectiveness. In this study, we examined the therapeutic effects and underlying mechanisms of human pluripotent stem cell-derived ectomesenchymal stromal cells (hPSC-EMSCs) in a rat model of HIE. Methods: hPSC-EMSCs were induced from either human embryonic stem cells or induced pluripotent stem cells. Stem cells or the conditioned medium (CM) derived from stem cells were delivered intracranially or intranasally to neonatal rats with HIE. Human umbilical cord-derived MSCs (hUC-MSCs) were used as the therapeutic comparison control and phosphate-buffered saline (PBS) was used as a negative control. Lesion size, apoptosis, neurogenesis, astrogliosis and microgliosis were evaluated. The rotarod test and Morris water maze were used to determine brain functional recovery. The PC-12 cell line, rat primary cortical neurons and neural progenitor cells were used to evaluate neurite outgrowth and the neuroprotective and neurogenesis effects of hPSC-EMSCs/hUC-MSCs. RNA-seq and enzyme-linked immunosorbent assays were used to determine the secretory factors that were differentially expressed between hPSC-EMSCs and hUC-MSCs. The activation and suppression of extracellular signal-regulated kinase (ERK) and cAMP response element-binding protein (CREB) were characterised using western blotting and immunofluorescent staining. Results: hPSC-EMSCs showed a higher neuroprotective potential than hUC-MSCs, as demonstrated by a more significant reduction in lesion size and apoptosis in the rat brain following hypoxia-ischaemia (HI). Compared with PBS treatment, hPSC-EMSCs promoted endogenous neurogenesis and alleviated astrogliosis and microgliosis. hPSC-EMSCs were more effective than hUC-MSCs. hPSC-EMSCs achieved a greater recovery of brain function than hUC-MSCs and PBS in rats with HIE. CM derived from hPSC-EMSCs had neuroprotective and neurorestorative effects in vitro through anti-apoptotic and neurite outgrowth- and neurogenesis-promoting effects. Direct comparisons between hPSC-EMSCs and hUC-MSCs revealed the significant enrichment of a group of secretory factors in hPSC-EMSCs, including nerve growth factor (NGF), platelet-derived growth factor-AA and transforming growth factor-ß2, which are involved in neurogenesis, synaptic transmission and neurotransmitter transport, respectively. Mechanistically, the CM derived from hPSC-EMSCs was found to potentiate NGF-induced neurite outgrowth and the neuronal differentiation of NPCs via the ERK/CREB pathway. Suppression of ERK or CREB abolished CM-potentiated neuritogenesis and neuronal differentiation. Finally, intranasal delivery of the CM derived from hPSC-EMSCs significantly reduced brain lesion size, promoted endogenous neurogenesis, mitigated inflammatory responses and improved functional recovery in rats with HIE. Conclusion: hPSC-EMSCs promote functional recovery after HI through multifaceted neuromodulatory activities via paracrine/trophic mechanisms. We propose the use of hPSC-EMSCs for the treatment of HIE, as they offer an excellent unlimited cellular source of MSCs.


Subject(s)
Hypoxia-Ischemia, Brain/therapy , Mesenchymal Stem Cell Transplantation/methods , Animals , Cells, Cultured , Female , Humans , Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Middle Aged , PC-3 Cells , Primary Cell Culture , Rats
2.
Neuro Oncol ; 24(6): 872-885, 2022 06 01.
Article in English | MEDLINE | ID: mdl-34655293

ABSTRACT

BACKGROUND: Neuroblastoma (NB) is a common childhood malignant tumor of neural crest (NC) origin with remarkable heterogeneity in outcomes. Amplification of the oncogene MYCN is strongly associated with highly malignant behaviour and poor prognosis. METHODS: This study aims to use a human embryonic stem cell (hESC)-derived NC model to identify novel downstream effectors of MYCN that can be potentially used as prognostic marker and/or therapeutic target. RESULTS: We show that MYCN-driven NB derived from human neural crest cells (hNCCs) recapitulate the pathological and molecular features of MYCN-amplified neuroblastoma (MNA-NB). By using this platform, we identify a group of 14 surface protein-encoding genes that are associated with MYCN expression level in MNA-NB. Among these genes, high CD55 expression is correlated with poor survival in MNA-NB but not in non-MNA-NB. Furthermore, CD55 promotes tumorigenesis, tumor growth, and cancer stemness in MNA-NB cell lines (MNA-NBL) through regulating the JNK pathway. Mechanistically, MYCN binds to both canonical and noncanonical E-boxes on the promoter of CD55 to regulate its transcriptional expression. Finally, neutralizing antibody targeting CD55 significantly attenuates cancer stemness, suppresses tumor growth, and improves survival exclusively in MNA-NBL-inoculated mice. CONCLUSION: MYCN shapes CD55 into a cancer stem cell regulator which represents a prognostic marker and therapeutic target of MNA-NB. The hESC-derived NC model serves as a valuable platform for investigating NB initiation and progression and developing potential therapeutic targets.


Subject(s)
Human Embryonic Stem Cells , Neuroblastoma , Animals , Cell Line, Tumor , Child , Gene Expression Regulation, Neoplastic , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/pathology , Humans , Mice , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism , N-Myc Proto-Oncogene Protein/therapeutic use , Neoplastic Stem Cells/metabolism , Neural Crest/metabolism , Neural Crest/pathology , Neuroblastoma/genetics , Transcription Factors/genetics
3.
Stem Cell Res Ther ; 11(1): 279, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32660632

ABSTRACT

BACKGROUND: Stem cell senescence has been proposed as one of the major drivers of aging, and MSC senescence contributes to aging-related diseases. Activation of mTORC1 pathway and heterochromatin organization have been characterized as two characteristics of senescent cells; however, whether mTORC1 pathway interacts with heterochromatin organization and contributes to MSC senescence remains unknown. In this study, we investigated the interaction between heterochromatin organization and mTORC1/p70S6K pathway in stress-induced MSC senescence. METHODS: The stress-induced senescence models were established in human umbilical cord-derived MSCs by doxorubicin (Dox) or H2O2. Cellular senescence was evaluated by ß-Gal activity, upregulation of cell cycle suppressor genes, and expression of SASP. Activation of heterochromatin organization and mTORC1 pathway was determined by Western blot and immunofluorescent staining. A D-galactose (D-Gal)-induced aging model was established in rats to evaluate the crosstalk between heterochromatin and mTORC1 pathway in vivo. RESULTS: We found that heterochromatin organization was provoked at the early stage of Dox- or H2O2-induced senescence. Disruption of heterochromatin organization led to robust DNA damage response and exacerbated cellular senescence. Suppression of mTORC1/p70S6K pathway by either rapamycin or p70S6K knockdown promoted heterochromatin organization and ameliorated Dox- or H2O2-induced DNA damage and senescence. In contrast, direct activation of mTORC1 by MHY1485 impaired heterochromatin organization and aggravated stress-induced senescence. Moreover, concomitant activation of mTORC1 pathway and heterochromatin organization was found in D-galactose-induced osteoporosis model in rats. Rapamycin alleviated cellular senescence and promoted heterochromatin organization in BMSCs derived from D-galactose-treated rats. CONCLUSIONS: Altogether, our study indicates the existence of a complex interplay between the mTORC1/p70S6K pathway and the heterochromatin organization during stress-induced MSC senescence, with important implications for the understanding of aging as well as for its prevention and treatment.


Subject(s)
Heterochromatin , Ribosomal Protein S6 Kinases, 70-kDa , Animals , Cellular Senescence , Heterochromatin/genetics , Hydrogen Peroxide , Mechanistic Target of Rapamycin Complex 1/genetics , Rats
4.
iScience ; 21: 375-390, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31704649

ABSTRACT

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.

5.
Cancer Cell Int ; 18: 156, 2018.
Article in English | MEDLINE | ID: mdl-30337838

ABSTRACT

BACKGROUND: As newly identified Wnt enhancer, R-spondin gene family members have been linked to various cancers; however, their role in isocitrate dehydrogenase-wildtype subtype of human glioblastoma (GBM) cells remains unknown. METHODS: Human U87 and U251 cell lines were used to perform the experiments. GBM stem-like cells were enriched in stem cell growth media and induced to differentiate using retinoid acid or growth factor deprivation. Wnthigh and Wntlow subpopulations were isolated and evaluated by MTS, sphere formation, transwell migration and xenograft formation assays. RESULTS: R-spondin 2 but not R-spondin 3 potentiates Wnt/ß-catenin signaling in GBM cell lines. While R-spondin 2 does not affect cell growth, it induces the expression of pluripotent stem cell markers in combination with Wnt3A. GBM stem-like cells are endowed with intrinsic high activity of ß-catenin signaling, which can be further intensified by R-spondin 2. In addition, R-spondin2 promotes stem cell self-renewal and suppresses retinoid acid- or growth factor deprivation-induced differentiation, indicating R-spondin 2 maintains stem cell traits in GBM. On the other hand, we identify subpopulations of GBM cells that show distinctive responsiveness to Wnt/ß-catenin signaling. Interestingly, Wnthigh and Wntlow cells display distinctive biologic properties. Moreover, Wnthigh cell-inoculated xenografts exhibit enhanced tumorigenicity and increased expression levels of R-spondin 2 compared to Wntlow cell-inoculated xenografts. CONCLUSION: Our study reveals a novel regulatory mechanisms underlying the over-activation of ß-catenin-mediated signaling in the pathogenesis of GBM.

6.
Cell Transplant ; 27(9): 1352-1367, 2018 09.
Article in English | MEDLINE | ID: mdl-30095002

ABSTRACT

Mesenchymal stem cell (MSC)-based cell therapy has been demonstrated as a promising strategy in the treatment of inflammatory bowel disease (IBD), which is considered an immune disease. While the exact mechanisms underlying the therapeutic effect of MSCs are still unclear, MSCs display anti-inflammatory and immunomodulatory effects by interacting with various immunoregulatory cells. Our previous studies have shown that MSCs can be preconditioned and deconditioned with enhanced cell survival, differentiation and migration. In this study, we evaluated the effect of preconditioning on the immunoregulatory function of human umbilical cord-derived MSCs (hUCMSCs) and their therapeutic effect on treating IBD. Our results show that intraperitoneal administration of deconditioned hUCMSCs (De-hUCMSCs) reduces the disease activity index (DAI), histological colitis score and destruction of the epithelial barrier, and increases the body weight recovery more intensively than that of un-manipulated hUCMSCs. In addition, De-hUCMSCs but not hUCMSCs elicit anti-apoptotic effects via induction of the ERK pathway during the early stage of IBD development. In vitro co-culture studies indicate that De-hUCMSCs suppress T-cell proliferation and activation more markedly than hUCMSCs. Moreover, De-hUCMSCs block the induction of inflammatory cytokines such as tumor necrosis factor (TNF)α and interleukin (IL)-2, while promoting the secretion of the anti-inflammatory cytokine IL-10 in T-cells. Mechanically, we find that prostaglandin E2 (PGE2) secretion is significantly increased in De-hUCMSCs, the suppression of which dramatically abrogates the inhibitory effect of De-hUCMSCs on T-cell activation, implying that the crosstalk between De-hUCMSCs and T-cells is mediated by PGE2. Together, we have demonstrated that preconditioning enhances the immunosuppressive and therapeutic effects of hUCMSCs on treating IBD via increased secretion of PGE2.


Subject(s)
Colitis/therapy , Dinoprostone/immunology , Inflammatory Bowel Diseases/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Animals , Cell Line , Colitis/immunology , Colitis/pathology , Female , HCT116 Cells , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Jurkat Cells , Mice, Inbred C57BL
7.
EMBO Mol Med ; 10(10)2018 10.
Article in English | MEDLINE | ID: mdl-30154237

ABSTRACT

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.


Subject(s)
Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Labor, Obstetric , Animals , Female , Gene Expression Profiling , Humans , Mice , Models, Animal , Pregnancy , Uterus/physiology
8.
Oncotarget ; 8(40): 66951-66959, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28978008

ABSTRACT

Endometriotic tissues exhibit high migration ability with the underlying mechanisms remain elusive. Our previous studies have demonstrated that cystic fibrosis transmembrane conductance regulator (CFTR) acts as a tumor suppressor regulating cell migration. In the present study, we explored whether CFTR plays a role in the development of human endometriosis. We found that both mRNA and protein expression levels of CFTR and urokinase-type plasminogen activator receptor (uPAR) were significantly increased in ectopic endometrial tissues from patients with endometriosis compared to normal endometrial tissues from women without endometriosis and positively correlated. In human endometrial Ishikawa (ISK) cells, overexpression of CFTR stimulated cell migration with upregulated NFκB p65 and uPAR. Knockdown of CFTR inhibited cell migration. Furthermore, inhibition of NFκB with its inhibitors (curcumin or Bay) significantly reduced the expression of uPAR and cell migration in the CFTR-overexpressing ISK cells. Collectively, the present results suggest that the CFTR-NFκB-uPAR signaling may contribute to the progression of human endometriosis, and indicate potential targets for diagnosis and treatment.

9.
Sci Rep ; 7(1): 5233, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701694

ABSTRACT

Cystic fibrosis transmembrane conductance regulator (CFTR), known as a cAMP-activated Cl- channel, is widely expressed at the apical membrane of epithelial cells in a wide variety of tissues. Of note, despite the abundant expression of CFTR in mammalian kidney, the role of CFTR in kidney disease development is unclear. Here, we report that CFTR expression is downregulated in the UUO (unilateral ureteral obstruction)-induced kidney fibrosis mouse model and human fibrotic kidneys. Dysfunction or downregulation of CFTR in renal epithelial cells leads to alteration of genes involved in Epithelial-Mesenchymal Transition (EMT) and kidney fibrosis. In addition, dysregulation of CFTR activates canonical Wnt/ß-catenin signaling pathways, whereas the ß-catenin inhibitor reverses the effects of CFTR downregulation on EMT marker. More interestingly, CFTR interacts with Dishevelled 2 (Dvl2), a key component of Wnt signaling, thereby suppressing the activation of ß-catenin. Compared to wild type, deltaF508 mice with UUO treatment exhibit significantly higher ß-catenin activity with aggregated kidney fibrogenesis, which is reduced by forced overexpression of CFTR. Taken together, our study reveals a novel mechanism by which CFTR regulates Wnt/ß-catenin signaling pertinent to progression of kidney fibrosis and indicates a potential treatment target.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Fibrosis/pathology , Kidney Diseases/pathology , Mutation , beta Catenin/metabolism , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells , Epithelial-Mesenchymal Transition , Female , Fibrosis/genetics , Fibrosis/metabolism , Humans , Kidney Diseases/genetics , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Wnt Signaling Pathway , beta Catenin/genetics
10.
Cell Death Dis ; 8(5): e2819, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28542126

ABSTRACT

Mesenchymal stem cells (MSCs) extensively interact with cancer cells and other stroma cells in the tumor microenvironment. However, the role of MSCs in colorectal cancer (CRC) progression and metastasis is controversial. This study was designed to identify the role of inflammation-activated-MSCs in CRC development. Our results show that tumor necrosis factor (TNF)-α-preactivated-hMSCs significantly promote the progression of colon cancer cells by enhancing cell proliferation, epithelial-mesenchymal transition, migration, and invasion. TNF-α-primed-hMSCs secrete high level of CCL5, which interacts with its receptor CCR1 expressed in colon cancer cells. Interestingly, the stimulation of colon cancer cell progression by TNF-α-primed hMSCs is associated with the upregulation of ß-catenin signaling pathway. Blocking ß-catenin pathway significantly decreases the TNF-α-primed-conditioned medium or CCL5-mediated cancer cell progression by decreasing the enhancement of Slug, suggesting that the CCL5/ß-catenin/Slug pathway plays a critical role in hMSC-mediated cancer progression. Furthermore, in vivo model in nude mice confirms the ability of hMSCs to promote the proliferation and progression of colon cancer cells, and the upregulation of CCl5/ß-catenin/Slug pathway. Taken together, the present study has demonstrated a novel pathway involving CCl5/CCR1/ß-catenin/Slug, via which hMSCs promotes CRC development.


Subject(s)
Chemokine CCL5/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Progression , Epithelial-Mesenchymal Transition , Mesenchymal Stem Cells/metabolism , Snail Family Transcription Factors/metabolism , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Phenotype , Receptors, CCR1/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
11.
Stem Cell Reports ; 8(3): 743-757, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28216148

ABSTRACT

The success of stem cell-mediated gene therapy in cancer treatment largely depends on the specific homing ability of stem cells. We have previously demonstrated that after in vitro induction of neuronal differentiation and dedifferentiation, bone marrow stromal cells (BMSCs) revert to a primitive stem cell population (De-neu-BMSCs) distinct from naive BMSCs. We report here that De-neu-BMSCs express significantly higher levels of chemokines, and display enhanced homing abilities to glioma, the effect of which is mediated by the activated CCL5/CCR1/ERK axis. Intriguingly, we find that the activated chemokine axis in De-neu-BMSCs is epigenetically regulated by histone modifications. On the therapeutic front, we show that De-neu-BMSCs elicit stronger homing and glioma-killing effects together with cytosine deaminase/5-fluorocytosine compared with unmanipulated BMSCs in vivo. Altogether, the current study provides an insight into chemokine regulation in BMSCs, which may have more profound effects on BMSC function and their application in regenerative medicine and cancer targeting.


Subject(s)
Chemokine CCL5/metabolism , Epigenesis, Genetic , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioma/genetics , Glioma/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Receptors, CCR1/metabolism , Animals , Cell Dedifferentiation , Cell Movement/genetics , Cellular Reprogramming , Chemokines/metabolism , Histones/metabolism , Humans , Mice , Signal Transduction
12.
Oncotarget ; 7(39): 64030-64042, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27588407

ABSTRACT

While inflammation with aberrant activation of NF-κB pathway is a hallmark of cystic fibrosis (CF), the molecular mechanisms underlying the link between CFTR defect and activation of NF-κB-mediated pro-inflammatory response remain elusive. Here, we investigated the link between CFTR defect and NF-κB activation in ΔF508cftr-/- mouse intestine and human intestinal epithelial cell lines. Our results show that the NF-κB/COX-2/PGE2 pathway is activated whereas the ß-catenin pathway is suppressed in CF mouse intestine and CFTR-knockdown cells. Activation of ß-catenin pathway by GSK3 inhibitors suppresses CFTR mutation/knockdown-induced NF-κB/COX-2/PGE2 pathway in ΔF508 mouse intestine and CFTR-knockdown cells. In contrast, suppression of ß-catenin signaling induces the nuclear translocation of NF-κB. In addition, CFTR co-localizes and interacts with ß-catenin while CFTR mutation disrupts the interaction between NF-κB and ß-catenin in mouse intestine. Treatment with proteasome inhibitor MG132 completely reverses the reduced expression of ß-catenin in Caco-2 cells. Collectively, these results indicate that CFTR stabilizes ß-catenin and prevents its degradation, defect of which results in the activation of NF-κB-mediated inflammatory cascade. The present study has demonstrated a previously unsuspected interaction between CFTR and ß-catenin that regulates NF-κB nuclear translocation in mouse intestine. Therefore, our study provides novel insights into the physiological function of CFTR and pathogenesis of CF-related diseases in addition to the NF-κB-mediated intestinal inflammation seen in CF.


Subject(s)
Active Transport, Cell Nucleus , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/genetics , Inflammation , Intestine, Small/pathology , beta Catenin/metabolism , Animals , Caco-2 Cells , Cystic Fibrosis/pathology , Glycogen Synthase Kinase 3/metabolism , Humans , Intestine, Small/metabolism , Leupeptins/chemistry , Mice , Mice, Inbred CFTR , Mutation , NF-kappa B p50 Subunit/metabolism , Signal Transduction
14.
PLoS One ; 11(1): e0148062, 2016.
Article in English | MEDLINE | ID: mdl-26815535

ABSTRACT

The biologic studies of human neural crest stem cells (hNCSCs) are extremely challenging due to the limited source of hNCSCs as well as ethical and technical issues surrounding isolation of early human embryonic tissues. On the other hand, vast majority of studies on MycN have been conducted in human tumor cells, thus, the role of MycN in normal human neural crest development is completely unknown. In the present study, we determined the role of MycN in hNCSCs isolated from in vitro-differentiating human embryonic stem cells (hESCs). For the first time, we show that suppression of MycN in hNCSCs inhibits cell growth and cell cycle progression. Knockdown of MycN in hNCSCs increases the expression of Cdkn1a, Cdkn2a and Cdkn2b, which encodes the cyclin-dependent kinases p21CIP1, p16 INK4a and p15INK4b. In addition, MycN is involved in the regulation of human sympathetic neurogenesis, as knockdown of MycN enhances the expression of key transcription factors involved in sympathetic neuron differentiation, including Phox2a, Phox2b, Mash1, Hand2 and Gata3. We propose that unlimited source of hNCSCs provides an invaluable platform for the studies of human neural crest development and diseases.


Subject(s)
Cell Proliferation , Human Embryonic Stem Cells/cytology , Neural Crest/cytology , Neural Stem Cells/cytology , Neurogenesis , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Cell Cycle , Cell Line , Cells, Cultured , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Human Embryonic Stem Cells/metabolism , Humans , N-Myc Proto-Oncogene Protein , Neural Crest/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism
15.
Sci Rep ; 5: 15946, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26515683

ABSTRACT

The mechanism underlying pulmonary inflammation in thermal inhalation injury remains elusive. Cystic fibrosis, also hallmarked with pulmonary inflammation, is caused by mutations in CFTR, the expression of which is temperature-sensitive. We investigated whether CFTR is involved in heat-induced pulmonary inflammation. We applied heat-treatment in 16HBE14o- cells with CFTR knockdown or overexpression and heat-inhalation in rats in vivo. Heat-treatment caused significant reduction in CFTR and, reciprocally, increase in COX-2 at early stages both in vitro and in vivo. Activation of ERK/JNK, NF-κB and COX-2/PGE2 were detected in heat-treated cells, which were mimicked by knockdown, and reversed by overexpression of CFTR or VX-809, a reported CFTR mutation corrector. JNK/ERK inhibition reversed heat-/CFTR-knockdown-induced NF-κB activation, whereas NF-κB inhibitor showed no effect on JNK/ERK. IL-8 was augmented by heat-treatment or CFTR-knockdown, which was abolished by inhibition of NF-κB, JNK/ERK or COX-2. Moreover, in vitro or in vivo treatment with curcumin, a natural phenolic compound, significantly enhanced CFTR expression and reversed the heat-induced increases in COX-2/PGE2/IL-8, neutrophil infiltration and tissue damage in the airway. These results have revealed a CFTR-regulated MAPK/NF-κB pathway leading to COX-2/PGE2/IL-8 activation in thermal inhalation injury, and demonstrated therapeutic potential of curcumin for alleviating heat-induced pulmonary inflammation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Cell Line , Curcumin/pharmacology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Dinoprostone/analysis , Enzyme-Linked Immunosorbent Assay , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Hot Temperature , Inflammation/etiology , Inflammation/metabolism , Inhalation , Interleukin-8/analysis , Interleukin-8/genetics , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Diseases/etiology , Lung Diseases/metabolism , Male , Microscopy, Fluorescence , NF-kappa B/antagonists & inhibitors , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Up-Regulation/drug effects
16.
J Clin Endocrinol Metab ; 100(7): E955-63, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25996258

ABSTRACT

CONTEXT: Human endometriosis (EMS) is characterized by insufficient apoptosis. Our previous studies have shown elevated CD147 expression in human endometriotic tissues and its involvement in endometrial cell apoptosis. However, the exact underlying mechanism remains elusive. OBJECTIVE: The objective was to examine the correlation of the highly expressed CD147 with anti-apoptotic factor Bcl-2 in human endometriotic tissues and to determine the CD147-regulated apoptotic pathway in human endometrial epithelial cell line (HES). DESIGN: This was a laboratory study using human tissue analysis and HES cell culture. SETTING: The setting was an academic research center and hospital. PATIENTS: Patients were 30 women with ovarian EMS and 12 women without EMS. INTERVENTIONS: mRNA levels of CD147 and Bcl-2 were evaluated in endometriotic tissues by quantitative real-time PCR. HES cells were transfected with pcDNA3.0-CD147 overexpressing plasmid or immune-depleted by CD147 antibody. MAIN OUTCOME MEASURES: Main outcome measures were reverse transcription, quantitative real-time PCR, 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay, and Western blotting. RESULTS: In human endometriotic tissues, Bcl-2 was up-regulated and positively correlated with CD147 expression, accompanied by activated ERK signaling. In HES cells, overexpression of CD147 increased viable cells and up-regulated Bcl-2 expression by activation of ERK signaling. Interference with CD147 function suppressed ERK signaling and decreased Bcl-2 expression, followed by accumulation of apoptotic factors, including cleaved caspase-9, cleaved caspase-3, and cleaved poly ADP-ribose polymerase. CONCLUSIONS: The presently found strong correlations between Bcl-2 and CD147, ERK, and CD147 in human endometriotic lesions and the demonstrated reduced cell apoptosis through CD147-ERK-Bcl-2 intrinsic apoptosis signaling axis suggest that this CD147-regulated signaling may contribute to the enhanced cell survival in the progression of human EMS.


Subject(s)
Basigin/physiology , Endometriosis/genetics , Ovarian Diseases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Adult , Cell Survival/genetics , Cells, Cultured , Endometriosis/metabolism , Endometriosis/pathology , Enzyme Activation/genetics , Female , Humans , MAP Kinase Signaling System/physiology , Middle Aged , Ovarian Diseases/metabolism , Ovarian Diseases/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Up-Regulation/genetics
17.
Int J Oncol ; 46(5): 2107-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25760446

ABSTRACT

An increased risk of non-small cell lung cancer (NSCLC) in cystic fibrosis (CF) patients and carriers of CF transmembrane conductance regulator (CFTR) mutations has been proposed. However, the role of CFTR in lung cancer remains controversial. In the present study, CFTR expression was assessed in 165 NSCLC tumors and 22 normal lung samples with validation in an independent series of 131 samples. The effect of gain and loss of CFTR on the malignant behavior of NSCLC was examined. The effect of CFTR manipulation on tumor metastasis was examined in a mouse model. Expression of CFTR was downregulated in NSCLC (p=0.041). Low CFTR expression was correlated with advanced stage (p<0.001) and lymph node metastasis (p=0.009). Low CFTR expression was significantly associated with poor prognosis (overall survival: 45 vs. 36 months, p<0.0001; progression-free survival: 41 vs. 30 months, p=0.007). Knockdown of CFTR in NSCLC cells enhanced malignant behavior (epithelial-mesenchymal transition, invasion and migration); in contrast, overexpression of CFTR suppressed cancer progression in vitro and in vivo. The tumor-suppressing effect of CFTR was associated with inhibition of multiple uPA/uPAR-mediated malignant traits in culture. These results show that CFTR plays a role in inhibition of NSCLC metastasis and suggest that CFTR may serve as a novel indicator for predicting adverse prognosis and metastasis in NSCLC patients.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/biosynthesis , Lung Neoplasms/pathology , Adult , Aged , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Down-Regulation , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Proportional Hazards Models , Real-Time Polymerase Chain Reaction
18.
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
19.
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
20.
Nat Commun ; 5: 4420, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25025956

ABSTRACT

The cause of insulin insufficiency remains unknown in many diabetic cases. Up to 50% adult patients with cystic fibrosis (CF), a disease caused by mutations in the gene encoding the CF transmembrane conductance regulator (CFTR), develop CF-related diabetes (CFRD) with most patients exhibiting insulin insufficiency. Here we show that CFTR is a regulator of glucose-dependent electrical acitivities and insulin secretion in ß-cells. We demonstrate that glucose elicited whole-cell currents, membrane depolarization, electrical bursts or action potentials, Ca(2+) oscillations and insulin secretion are abolished or reduced by inhibitors or knockdown of CFTR in primary mouse ß-cells or RINm5F ß-cell line, or significantly attenuated in CFTR mutant (DF508) mice compared with wild-type mice. VX-809, a newly discovered corrector of DF508 mutation, successfully rescues the defects in DF508 ß-cells. Our results reveal a role of CFTR in glucose-induced electrical activities and insulin secretion in ß-cells, shed light on the pathogenesis of CFRD and possibly other idiopathic diabetes, and present a potential treatment strategy.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Enzyme-Linked Immunosorbent Assay , Insulin Secretion , Insulin-Secreting Cells/drug effects , Male , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...