Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 527
Filter
1.
Life Sci ; 264: 118688, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33130074

ABSTRACT

AIMS: Many gastrointestinal (GI) disorders are developmental in origin and are caused by abnormal enteric nervous system (ENS) formation. Maternal vitamin A deficiency (VAD) during pregnancy affects multiple central nervous system developmental processes during embryogenesis and fetal life. Here, we evaluated whether maternal diet-induced VAD during pregnancy alone can cause changes in the ENS that lead to GI dysfunction in rat offspring. MAIN METHODS: Rats were selected to construct animal models of normal VA, VA deficiency and VA supplementation. The fecal water content, total gastrointestinal transmission time and colonic motility were measured to evaluate gastrointestinal function of eight-week-old offspring rats. The expression levels of RARß, SOX10, cholinergic (ChAT) and nitrergic (nNOS) enteric neurons in colon tissues were detected through western blot and immunofluorescence. Primary enteric neurospheres were treated with retinoic acid (RA), infection with Ad-RARß and siRARß adenovirus, respectively. KEY FINDINGS: Our data revealed marked reductions in the mean densities of cholinergic and nitrergic enteric neurons in the colon and GI dysfunction evidenced by mild intestinal flatulence, increased fecal water content, prolonged total GI transit time and reduced colon motility in adult offspring of the VAD group. Interestingly, maternal VA supplementation (VAS) during pregnancy rescued these changes. In addition, in vitro experiments demonstrated that exposure to appropriate doses of RA promoted enteric neurosphere differentiation into cholinergic and nitrergic neurons, possibly by upregulating RARß expression, leading to enhanced SOX10 expression. SIGNIFICANCE: Maternal VAD during pregnancy is an environmental risk factor for GI dysfunction in rat offspring.


Subject(s)
Cholinergic Neurons/metabolism , Gastrointestinal Diseases/metabolism , Gastrointestinal Tract/metabolism , Nitrergic Neurons/metabolism , Receptors, Retinoic Acid/biosynthesis , Vitamin A Deficiency/blood , Animals , Cells, Cultured , Cholinergic Neurons/pathology , Female , Gastrointestinal Diseases/pathology , Gastrointestinal Tract/pathology , Nitrergic Neurons/pathology , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/antagonists & inhibitors , Vitamin A Deficiency/complications
2.
Mol Biol Rep ; 47(8): 6105-6110, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32780252

ABSTRACT

The etiology of pterygium remains unclear, but ultraviolet (UV) radiation is generally considered to be major risk factor. Pterygium has similarity features with many cancers, including inflammation, invasion, cell proliferation, anti-apoptosis, angiogenesis and recurrence after resection. Retinoic acid via cellular retinoic acid binding protein 2 (CRABP2) is involved in cell cycle arrest, apoptosis and differentiation, while it via fatty acid binding protein 5 (FABP5) is involved in survival, cell proliferation and angiogenesis, which pathway gets activated depends on the CRABP2/FABP5 ratio. Alterations of retinoid signaling were found in many cancer types. The deregulated retinoid signaling may also contribute to the development and/or recurrence of pterygium. The aim of our study was to determine mRNA and protein expressions of CRABP2 and FABP5 and ratio of CRABP2/FABP5 in primer and recurrent pterygium tissues. Pterygia tissues were collected from 30 eyes of 30 patients undergoing pterygium excision. CRABP2 and FABP5 mRNA and protein expression were assessed using Real-time PCR and Western blotting through examination of excised specimens from pterygium and conjunctiva tissues. The ratio of CRABP2/FABP5 gene expression was not altered when primary pterygium tissues compared normal conjunctival tissues (1.00-fold change). Whereas the ratio of CRABP2/ FABP5 gene expression was decreased when recurrent pterygium tissues compared normal conjunctival tissues (0.81-fold change). Understanding etiopathogenesis of pterygium may aid in the find of more promising treatments to prevent pterygium in earlier stages.


Subject(s)
Eye Proteins/genetics , Fatty Acid-Binding Proteins/genetics , Pterygium/genetics , Receptors, Retinoic Acid/genetics , Aged , Conjunctiva/metabolism , Eye Proteins/metabolism , Fatty Acid-Binding Proteins/biosynthesis , Female , Humans , Male , Middle Aged , Pterygium/metabolism , RNA, Messenger/biosynthesis , Real-Time Polymerase Chain Reaction , Receptors, Retinoic Acid/biosynthesis , Recurrence
3.
Anticancer Drugs ; 30(1): 56-64, 2019 01.
Article in English | MEDLINE | ID: mdl-30198914

ABSTRACT

The vitamin A derivative 9-cis-retinoic acid (9-cis-RA) has been used for the treatment and prevention of cutaneous T-cell lymphoma (CTCL). However, the precise mechanism by which 9-cis-RA treatment ameliorates CTCL remains elusive. Our research shows that 9-cis-RA inhibits proliferation and induces apoptosis in CTCL cells in a dose-dependent and time-dependent manner. 9-Cis-RA also induced G0/G1 cell cycle arrest by downregulation of cyclin D1. We confirmed that 9-cis-RA significantly decreased phosphorylation of JAK1, STAT3, and STAT5 and downregulated Bcl-xL and cyclin D1, indicating that 9-cis-RA inhibited the activation of JAK/STAT signaling. Meanwhile, 9-cis-RA also activated classical RA-mediated transcription by retinoic acid receptors (RAR) and/or retinoid X receptors (RXR) in a CTCL cell line. Thus, 9-cis-RA may be effective for chemotherapy and may prevent human CTCL by inhibiting proliferation and inducing apoptosis by inhibition of the JAK/STAT pathway and activation of the RAR/RXR pathway.


Subject(s)
Alitretinoin/pharmacology , Lymphoma, T-Cell, Cutaneous/drug therapy , Skin Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/biosynthesis , Janus Kinases/genetics , Janus Kinases/metabolism , Lymphoma, T-Cell, Cutaneous/genetics , Lymphoma, T-Cell, Cutaneous/metabolism , Lymphoma, T-Cell, Cutaneous/pathology , Nitriles , Pyrazoles/pharmacology , Pyrimidines , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Resting Phase, Cell Cycle/drug effects , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/biosynthesis , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Skin Neoplasms/pathology
4.
Diagn Pathol ; 13(1): 9, 2018 Jan 22.
Article in English | MEDLINE | ID: mdl-29378601

ABSTRACT

BACKGROUND: Nephroblastoma or Wilms tumor is the most frequent kidney cancer in children and accounts for 98% of kidney tumors in this age group. Despite favorable prognosis, a subgroup of these patients progresses to recurrence and death. The retinoic acid (RA) pathway plays a role in the chemoprevention and treatment of tumors due to its effects on cell differentiation and its antiproliferative, anti-oxidant, and pro-apoptotic activities. Reports describe abnormal cellular retinoic acid-binding protein 2 (CRABP2) expression in neoplasms and its correlation with prognostic factors and clinical and pathological characteristics. The aim of this study was to evaluate the immunohistochemical expression of retinoic acid receptor alpha (RARA) and CRABP2 in paraffin-embedded samples of nephroblastomas via semiquantitative and quantitative analyses and to correlate this expression with prognostic factors. METHODS: Seventy-seven cases of nephroblastomas were selected from pediatric oncology services. The respective medical records and surgical specimens were reviewed. Three representative tumor samples and one non-tumor renal tissue sample were selected for the preparation of tissue microarrays (TMA). The Allred scoring system was used for semiquantitative immunohistochemical analyses, whereas a morphometric analysis of the stained area was employed for quantitative evaluation. The nonparametric Mann-Whitney test was used for comparisons between two groups, while the nonparametric Kruskal-Wallis test was used to compare three or more groups. RESULTS: Immunopositivity for RARA and CRABP2 was observed in both the nucleus and cytoplasm. All histological components of the nephroblastoma (blastema, epithelium, and stroma) were positive for both markers. RARA, based on semiquantitative analyses, and CRABP2, bases on quantitative analyses, exhibited increased immunohistochemical expression in patients with metastasis, with p values of 0.0247 and 0.0128, respectively. These findings were similar to the results of the quantitative analysis of RARA expression, showing greater immunopositivity in tumor samples of patients subjected to pre-surgical chemotherapy. No significant correlation was found with the other variables studied, such as disease stage, anaplasia, risk group, histological type, nodal involvement, and clinical evolution. CONCLUSIONS: Semiquantitative and quantitative analyses of the markers RARA and CRABP2 indicate their potential as biomarkers for tumor progression and their participation in nephroblastoma tumorigenesis.


Subject(s)
Biomarkers, Tumor/analysis , Kidney Neoplasms/pathology , Receptors, Retinoic Acid/biosynthesis , Retinoic Acid Receptor alpha/biosynthesis , Wilms Tumor/pathology , Child, Preschool , Female , Humans , Infant , Male
5.
Eur Rev Med Pharmacol Sci ; 21(19): 4304-4310, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29077166

ABSTRACT

OBJECTIVE: To explore the retinoic acid receptor-ß (RAR-ß) expression in lung squamous cell carcinoma (LSCC) tissues and its prognosis. PATIENTS AND METHODS: SP assay was used to detect the RAR-ß expression in 100 cases of surgically resected LSCC tissues and 20 cases of peritumoral normal lung tissues, and prognosis follow-up was conducted. RESULTS: The overall positive expression rate of RAR-ß was 54.00%, which was not correlated with age, gender, phase and pathological type (p>0.05). Stratified analysis showed that the prognosis of patients with positive IRAR-ß expression in phase I was significantly better than that of those with negative IRAR-ß expression, in which the median survival times were 31 and 22 months respectively (p=0.022). In contrast, the prognosis of patients with negative RAR-ß expression was better than that of those with positive RAR-ß expression in phase II and III A. The median survival times were 23 and 16 months respectively in phase II p = 0.008, and 19 and 7 months respectively in phase III A (p=0.019). CONCLUSIONS: RAR-ß is expressed in LSCC tumor tissues. RAR-ß expression, which is not significantly correlated with the clinicopathological characteristics of patients, affects the postoperative survival of LSCC patients in phase I and II-III A dually. RAR-ß expression state is one of the independent factors for the prognosis of LSCC patients.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Receptors, Retinoic Acid/biosynthesis , Adult , Biomarkers, Tumor/analysis , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/pathology , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Survival Analysis
6.
Tumour Biol ; 39(6): 1010428317712135, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28639915

ABSTRACT

TIG3 (tazarotene-induced gene 3) has been reported to suppress the progression of several malignancies, where this gene is universally downregulated. However, the expression of TIG3 in primary glioblastoma and its relevance to patient's prognosis have not been elaborated. Thus, this study was aimed to evaluate TIG3 expression level in primary glioblastoma and investigate the prognostic value of TIG3 for patients. The Cancer Genome Atlas database was first utilized to analyze the expression and prognostic potential of TIG3 in 528 glioblastoma cases. Compared with control group, glioblastoma showed significantly elevated TIG3 expression (p < 0.001). Log-rank analysis revealed that higher expression of TIG3 was associated with shorter overall survival (358vs 383 days, p = 0.039). Furthermore, TIG3 protein expression detected by immunohistochemistry confirmed positive correlation of TIG3 expression and glioma grade and upregulation of TIG3 in our cohort of 101 primary glioblastoma patients compared to 16 normal brains. Finally, Kaplan-Meier analysis and Cox regression analysis identified high TIG3 expression as an independent risk factor for overall survival of primary glioblastoma patients (overall survival, 10 vs 13 months, p = 0.033; hazard ratio = 1.542, p = 0.046). Together, this study indicated that increased expression of TIG3 in primary glioblastoma is a novel biomarker for predicting poor outcome of patients. We then hypothesize that TIG3 may function in a different pattern in glioblastoma.


Subject(s)
Biomarkers, Tumor/biosynthesis , Glioblastoma/genetics , Prognosis , Receptors, Retinoic Acid/biosynthesis , Adult , Aged , Biomarkers, Tumor/genetics , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Receptors, Retinoic Acid/genetics
7.
Int J Gynecol Cancer ; 27(4): 643-650, 2017 05.
Article in English | MEDLINE | ID: mdl-28375930

ABSTRACT

OBJECTIVE: Several studies have reported that retinoic acid (RA) might be used to treat malignancies. The effects of RA are mediated by the RA receptor (RAR), and RARα/RARß especially acts as a tumor suppressor. However, little is known about its role in human endometrial cancer. MATERIALS AND METHODS: In this study, we examined the effects of all-trans RA (ATRA) on progression of human endometrial cancer cell line, RL95-2 and Hec1A. We then examined the expression of RARα and RARß in 50 endometrial cancer tissues by using immunohistochemistry. RESULTS: We found inhibitory effects of ATRA on cell proliferation, apoptosis, and migration in RL95-2 cells, but not in Hec1A cells. RARα or RARß knockdown individually could not cancel out the inhibition of cell proliferation by ATRA in RL95-2 cells, but simultaneous knockdown of RARα and RARß could block its effect on proliferation. RARα and RARß knockdown dose dependently reduced the inhibition of migration by ATRA, but the effect was more pronounced with RARß knockdown than with RARα knockdown. We confirmed that RARß gene was directly regulated by ATRA in microarray and real-time reverse transcription polymerase chain reaction. Furthermore, the RARß agonist (BMS453) significantly suppressed proliferation of RL95-2 cells. In immunohistochemical analysis, RARα expression was positively correlated with tumor grade, and RARß showed the opposite tendency in endometrial cancer. CONCLUSIONS: Retinoic acid might have multiple antitumor effects, and RARß may be a potent therapeutic target in RA treatment for endometrial cancers.


Subject(s)
Endometrial Neoplasms/drug therapy , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Molecular Targeted Therapy , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha/biosynthesis , Retinoic Acid Receptor alpha/deficiency , Retinoic Acid Receptor alpha/genetics , Retinoic Acid Receptor alpha/metabolism
8.
J Biol Chem ; 291(36): 18991-9005, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27402828

ABSTRACT

All-trans-retinoic acid may be an important molecular signal in the postnatal control of eye size. The goal of this study was to identify retinoic acid-binding proteins secreted by the choroid and sclera during visually guided ocular growth. Following photoaffinity labeling with all-trans-[11,12-(3)H]retinoic acid, the most abundant labeled protein detected in the conditioned medium of choroid or sclera had an apparent Mr of 27,000 Da. Following purification and mass spectrometry, the Mr 27,000 band was identified as apolipoprotein A-I. Affinity capture of the radioactive Mr 27,000 band by anti-chick apolipoprotein A-I antibodies confirmed its identity as apolipoprotein A-I. Photoaffinity labeling and fluorescence quenching experiments demonstrated that binding of retinoic acid to apolipoprotein A-I is 1) concentration-dependent, 2) selective for all-trans-retinoic acid, and 3) requires the presence of apolipoprotein A-I-associated lipids for retinoid binding. Expression of apolipoprotein A-I mRNA and protein synthesis were markedly up-regulated in choroids of chick eyes during the recovery from induced myopia, and apolipoprotein A-I mRNA was significantly increased in choroids following retinoic acid treatment. Together, these data suggest that apolipoprotein A-I may participate in a regulatory feedback mechanism with retinoic acid to control the action of retinoic acid on ocular targets during postnatal ocular growth.


Subject(s)
Apolipoprotein A-I/biosynthesis , Avian Proteins/biosynthesis , Choroid/metabolism , Eye Proteins/biosynthesis , Gene Expression Regulation/drug effects , Receptors, Retinoic Acid/biosynthesis , Tretinoin/pharmacology , Animals , Apolipoprotein A-I/chemistry , Avian Proteins/chemistry , Chickens , Choroid/chemistry , Eye Proteins/chemistry , Receptors, Retinoic Acid/chemistry , Tretinoin/chemistry
9.
J Chem Neuroanat ; 77: 80-92, 2016 11.
Article in English | MEDLINE | ID: mdl-27242163

ABSTRACT

Retinoic acid (RA) is important during development, in neuronal plasticity, and also in peripheral nervous system regeneration. Here we use the frog visual system as a model to investigate the changes in RA signaling that take place after axonal injury to the central nervous system. Immunocytochemistry was used to localize different components of RA signaling within sections of the retina and optic tectum, namely, the synthetic enzyme retinaldehyde dehydrogenase (RALDH), the RA binding proteins CRABPI and II, the retinoic acid receptors RARα, ß and γ, and finally the catabolic enzyme CYP26A1. The levels of these proteins were quantified in extracts of retina and tectum using Western blotting. Animals were studied at 1 week, 3 weeks and 6 weeks after optic nerve transection. At the latter time point the RGC axons were re-entering the optic tectum. All the components of RA signaling were present at low to moderate levels in retinas and tecta of control, unoperated animals. In retina, soon after optic nerve injury there was a large increase in RALDH, some increase in the CRABPs, and a large increase in RGC RARß and (expression. These increases continued as the RGC axons were regenerating, with the addition of later RARα expression at 6 weeks. At no stage did CYP26A1 expression significantly change. In the tectum the levels of RALDH increased after axotomy and during regrowth of axons (3 weeks), then decreased at 6 weeks, at which time the levels of CYP26A1 increased. Axotomy did not cause an immediate increase in tectal RAR levels but RARα and RARß increased after 3 weeks and RARγ only after 6 weeks. These results are consistent with RA signaling playing an important role in the survival and regeneration of frog RGCs.


Subject(s)
Optic Nerve Injuries/physiopathology , Signal Transduction , Tretinoin/metabolism , Visual Pathways/physiopathology , Animals , Female , Gene Expression Regulation , Immunohistochemistry , Male , Rana pipiens , Receptors, Retinoic Acid/biosynthesis , Retina/physiopathology , Retinal Dehydrogenase/biosynthesis , Retinal Ganglion Cells/metabolism , Retinoic Acid 4-Hydroxylase/biosynthesis , Retinoic Acid 4-Hydroxylase/genetics , Retinoid X Receptors/biosynthesis , Superior Colliculi/physiopathology
10.
Expert Opin Ther Pat ; 26(8): 957-71, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27336223

ABSTRACT

INTRODUCTION: Differentiation therapy using all-trans retinoic acid (ATRA) revolutionised the treatment of acute promyelocytic leukaemia to such an extent that it is now one of the most curable types of leukaemia, with ATRA and anthracycline-based chemotherapy providing cure rates above 80%. Isotretinoin is used to treat chronic acne. Here, we examine the information described in recent patents and the extent to which new findings are influencing extending retinoid-based differentiation therapy to other cancers, as well as the development of new therapies for other disorders. AREAS COVERED: A search has been performed on the literature and worldwide patents filed during 2014 to the present time, focusing on synthetic agonists and antagonists of retinoic acid receptors and novel compositions for the delivery of these agents. EXPERT OPINION: New potential therapeutic applications have been described, including lung, breast and head and neck cancers, T cell lymphoma and neurodegenerative, metabolic, ophthalmic, muscle, and inflammatory disorders. Recent patents have described the means to maximise retinoid activity. Two decades of efforts to extend retinoid-based therapies have been disappointing and new synthetic retinoids, target diseases and modes of delivery may well resolve this long standing issue.


Subject(s)
Receptors, Retinoic Acid/agonists , Receptors, Retinoic Acid/biosynthesis , Retinoids/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Design , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/pathology , Patents as Topic , Receptors, Retinoic Acid/antagonists & inhibitors , Retinoids/pharmacology , Retinoids/therapeutic use
11.
Mol Biol (Mosk) ; 50(2): 255-65, 2016.
Article in Russian | MEDLINE | ID: mdl-27239845

ABSTRACT

Retinoids are signaling molecules that control a wide variety of cellular processes and possess antitumor activity. This work presents a comprehensive description of changes in the expression of 23 genes that regulate retinoid metabolism and signaling in non-small-cell lung cancer tumors compared to adjacent normal tissues obtained using RT-PCR. Even at early stages of malignant transformation, a significant decrease in ADH1B, ADH3, RDHL, and RALDH1 mRNA levels was observed in 82, 79, 73, and 64% of tumor specimens, respectively, and a considerable increase in AKR1B10 mRNA content was observed in 80% of tumors. Dramatic changes in the levels of these mRNAs can impair the synthesis of all-trans retinoic acid, a key natural regulatory retinoid. Apart from that, it was found that mRNA levels of nuclear retinoid receptor genes RXRγ, RARα, RXRα, and gene RDH11 were significantly decreased in 80, 67, 57, and 66% of tumor specimens, respectively. Thus, neoplastic transformation of lung tissue cells is accompanied with deregulated expression of key genes of retinoid metabolism and function.


Subject(s)
3-Hydroxysteroid Dehydrogenases/biosynthesis , Alcohol Dehydrogenase/biosynthesis , Aldehyde Dehydrogenase/biosynthesis , Aldehyde Reductase/biosynthesis , Carcinoma, Non-Small-Cell Lung/genetics , 3-Hydroxysteroid Dehydrogenases/genetics , Alcohol Dehydrogenase/genetics , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Aldehyde Reductase/genetics , Aldo-Keto Reductases , Carcinoma, Non-Small-Cell Lung/pathology , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Humans , RNA, Messenger/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Retinal Dehydrogenase , Retinoic Acid Receptor alpha , Retinoids/genetics , Retinoids/metabolism , Signal Transduction/genetics , Tretinoin/metabolism
12.
Oncotarget ; 7(18): 25350-65, 2016 May 03.
Article in English | MEDLINE | ID: mdl-26992218

ABSTRACT

Hepatocellular carcinoma (HCC) is the third cause of cancer-related death worldwide. Accumulating studies have demonstrated that aberrant expression of several lncRNAs was found to be involved in the hepatocarcinogenesis. In this study, a lncRNA Ftx was chosen to investigate its effects on HCC cells, and clarify the possible mechanism. We demonstrated that the lncRNA Ftx and Ftx-derived miR-545 were up-regulated in both HCC tissues and cells. MiR-545 was positively correlated with lncRNA Ftx expression. Notably, clinical association analysis revealed that the high expression of lncRNA Ftx and miR-545 was associated with poor prognostic features, and conferred a reduced 5-year overall survival (OS) and disease-free survival (DFS) of HCC patients. We found that miR-545 was a pivotal mediator in Ftx-induced promotion of HCC cell growth. Subsequently, we identified RIG-I as a direct target of miR-545. The expression of RIG-I was downregulated in HCC tissues and was inversely correlated with miR-545 expression. Our data revealed that ectopic expression of RIG-I abrogated the effects of lncRNA Ftx or miR-545 on HCC cells. LncRNA Ftx/miR-545-mediated downregulation of RIG-I led to increased Akt phosphorylation in vitro and in vivo. Inhibition of Akt phosphorylation abolished the effects of lncRNA Ftx/miR-545 on HCC cells. In conclusion, our study demonstrates that the novel pathway lncRNA Ftx/miR-545/RIG-I promotes HCC development by activating PI3K/Akt signaling, and it may serve as a novel prognostic biomarker and therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Receptors, Retinoic Acid/genetics , Adult , Aged , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Cell Proliferation/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Male , Middle Aged , Receptors, Retinoic Acid/biosynthesis
13.
PLoS One ; 11(2): e0148381, 2016.
Article in English | MEDLINE | ID: mdl-26839961

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the predominant pathotype of esophageal carcinoma (EC) in China, especially in Henan province, with poor prognosis and limited 5-year survival rate. Cellular retinoic acid binding protein 2 (CRABP2) is a member of the retinoic acid (RA) and lipocalin/cytosolic fatty-acid binding protein family and plays a completely contrary role in tumorigenesis through the retinoid signaling pathway, depending on the nuclear RA receptors (RAR) and PPARbeta/delta receptors. Presently, the biological role of CRABP2 in the development of ESCC has never been reported. Here, we firstly evaluated the expression of CRABP2 at both mRNA and protein levels and showed that it was remarkably downregulated in clinical ESCC tissues and closely correlated with the occurrence position, pathology, TNM stage, size, infiltration depth and cell differentiation of the tumor. Additionally, the biological function assays demonstrated that CRABP2 acted as a tumor suppressor in esophageal squamous carcinogenesis by significantly inhibiting cell growth, inducing cell apoptosis and blocking cell metastasis both in vitro and in vivo. All in all, our finding simplicate that CRABP2 is possibly an efficient molecular marker for diagnosing and predicting the development of ESCC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Receptors, Retinoic Acid/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Animals , Apoptosis/genetics , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Down-Regulation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Receptors, Retinoic Acid/genetics , Tumor Suppressor Proteins/genetics
14.
BMC Cancer ; 16: 2, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26728137

ABSTRACT

BACKGROUND: Chemotherapy is the only therapy option for the majority of AML patients, however, there are several limitations for this treatment. Our aim was to find a new chemotherapy strategy that is more effective and less toxic. METHODS: MTT assays and a xenograft mouse model were employed to evaluate the synergistic activity of all-trans retinoic acid (ATRA) combined with topotecan (TPT). Drug-induced DNA damage and apoptosis were determined by flow cytometry analysis with PI and DAPI staining, the comet assay and Western blots. Short hairpin RNA (shRNA) and a RARα plasmid were used to determine whether RARα expression influenced DNA damage and apoptosis. RESULTS: We found that ATRA exhibited synergistic activity in combination with Topotecan in AML cells, and the enhanced apoptosis induced by Topotecan plus ATRA resulted from caspase pathway activation. Mechanistically, ATRA dramatically down regulated RARα protein levels and led to more DNA damage and ultimately resulted in the synergism of these two agents. In addition, the increased antitumor efficacy of Topotecan combined with ATRA was further validated in the HL60 xenograft mouse model. CONCLUSIONS: Our data demonstrated, for the first time, that the combination of TPT and ATRA showed potential benefits in AML, providing a novel insight into clinical treatment strategies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Drug Synergism , Leukemia, Myeloid, Acute/drug therapy , Topotecan/administration & dosage , Tretinoin/administration & dosage , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA Damage/drug effects , Gene Expression Regulation, Leukemic/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , RNA, Small Interfering , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Xenograft Model Antitumor Assays
15.
Oncotarget ; 6(34): 35830-42, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26416422

ABSTRACT

The transcription factor Kruppel-like factor 2 (KLF2) displays anticarcinogenic activities but the mechanism that underlies this activity is unknown. We show here that KLF2 is markedly downregulated in human breast cancers and that its expression positively correlates with breast cancer patient survival. We show further that KLF2 suppresses tumor development by controlling the transcriptional activity of the vitamin A metabolite retinoic acid (RA). RA regulates gene transcription by activating two types of nuclear receptors: RA receptors (RARs), which inhibit tumor development, and peroxisome proliferator-activated receptor ß/δ (PPARß/δ), which promotes tumorigenesis. The partitioning of RA between these receptors is regulated by two carrier proteins: cellular retinoic acid-binding protein 2 (CRABP2), which delivers RA to RARs, and fatty acid-binding protein 5 (FABP5), which shuttles ligands to PPARß/δ. We show that KLF2 induces the expression of CRABP2 and RARγ and inhibits the expression FABP5 and PPARß/δ thereby shifting RA signaling from the pro-carcinogenic FABP5/PPARß/δ to the growth-suppressing CRABP2/RAR path. The data thus reveal that KLF2 suppresses tumor growth by controlling the transcriptional activities of RA.


Subject(s)
Breast Neoplasms/metabolism , Kruppel-Like Transcription Factors/metabolism , Tretinoin/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Cell Transformation, Neoplastic , Down-Regulation , Fatty Acid-Binding Proteins/antagonists & inhibitors , Female , Heterografts , Humans , Kruppel-Like Transcription Factors/genetics , MCF-7 Cells , Mice , Mice, Nude , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Signal Transduction , Transcription, Genetic , Retinoic Acid Receptor gamma
16.
EMBO Mol Med ; 7(7): 950-72, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25888236

ABSTRACT

Forty-two cell lines recapitulating mammary carcinoma heterogeneity were profiled for all-trans retinoic acid (ATRA) sensitivity. Luminal and ER(+) (estrogen-receptor-positive) cell lines are generally sensitive to ATRA, while refractoriness/low sensitivity is associated with a Basal phenotype and HER2 positivity. Indeed, only 2 Basal cell lines (MDA-MB157 and HCC-1599) are highly sensitive to the retinoid. Sensitivity of HCC-1599 cells is confirmed in xenotransplanted mice. Short-term tissue-slice cultures of surgical samples validate the cell-line results and support the concept that a high proportion of Luminal/ER(+) carcinomas are ATRA sensitive, while triple-negative (Basal) and HER2-positive tumors tend to be retinoid resistant. Pathway-oriented analysis of the constitutive gene-expression profiles in the cell lines identifies RARα as the member of the retinoid pathway directly associated with a Luminal phenotype, estrogen positivity and ATRA sensitivity. RARα3 is the major transcript in ATRA-sensitive cells and tumors. Studies in selected cell lines with agonists/antagonists confirm that RARα is the principal mediator of ATRA responsiveness. RARα over-expression sensitizes retinoid-resistant MDA-MB453 cells to ATRA anti-proliferative action. Conversely, silencing of RARα in retinoid-sensitive SKBR3 cells abrogates ATRA responsiveness. All this is paralleled by similar effects on ATRA-dependent inhibition of cell motility, indicating that RARα may mediate also ATRA anti-metastatic effects. We define gene sets of predictive potential which are associated with ATRA sensitivity in breast cancer cell lines and validate them in short-term tissue cultures of Luminal/ER(+) and triple-negative tumors. In these last models, we determine the perturbations in the transcriptomic profiles afforded by ATRA. The study provides fundamental information for the development of retinoid-based therapeutic strategies aimed at the stratified treatment of breast cancer subtypes.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Receptors, Retinoic Acid/biosynthesis , Tretinoin/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Gene Expression Profiling , Gene Silencing , Humans , Retinoic Acid Receptor alpha , Transplantation, Heterologous , Tretinoin/therapeutic use
17.
Acta Haematol ; 133(3): 279-86, 2015.
Article in English | MEDLINE | ID: mdl-25413479

ABSTRACT

BACKGROUND: This study was conducted to determine the antineoplastic activities of 5-aza-2'-deoxycytidine (decitabine; DAC) and all-trans retinoic acid (ATRA), administered either alone or in combination, on in vitro cultured SHI-1 cells as well as their effects on the expression of the tumor suppressor gene p16(INK4a) (p16) and the retinoic acid receptor (RAR)-ß. METHODS: Cell growth inhibition, differentiation and apoptosis were determined in SHI-1 cells treated with DAC and/or ATRA, and the combination index of the two compounds was calculated. Methylation of the p16 and RAR-ß genes in SHI-1 cells was detected by methylation-specific polymerase chain reaction (PCR). Real-time quantitative reverse transcriptase PCR was used to detect mRNA expression of the p16 and RAR-ß genes, and Western blot analysis was performed for protein expression. RESULTS: The drug combination had a synergistic effect on growth inhibition, differentiation and apoptosis of SHI-1 cells, and the effects of DAC and ATRA were dependent on time. DAC, either alone or in combination with ATRA, induced demethylation of the genes p16 and RAR-ß, whereas ATRA alone had no effect on methylation. The RAR-ß gene was reexpressed following DAC-ATRA combination treatment, and both agents had no effect on p16 expression. CONCLUSION: The results revealed that DAC used in combination with ATRA has significant clinical potential in the treatment of acute monocytic leukemia.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Leukemia, Monocytic, Acute/drug therapy , Receptors, Retinoic Acid/biosynthesis , Azacitidine/agonists , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Decitabine , Humans , Leukemia, Monocytic, Acute/metabolism , Leukemia, Monocytic, Acute/pathology , Tretinoin/agonists , Tretinoin/pharmacology
18.
Mol Oncol ; 9(2): 355-64, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25300573

ABSTRACT

The early steps of mammary tumorigenesis include loss of epithelial cell polarity, escape from anoikis, and acquisition of proliferative capacity. The genes responsible for these processes are predicted to be early diagnostic markers or new therapeutic targets. Here we tested 51 genes coamplified with ERBB2 in the 17q12-21 amplicon for these tumorigenic activities using an MCF10A 3D culture-based screening system. We found that overexpression of retinoic acid receptor α (RARA) disrupted normal acinar structure and induced epithelial-to-mesenchymal transition (EMT). The mRNA levels of known EMT-inducing factors, including SLUG, FOXC2, ZEB1, and ZEB2, were significantly increased upon RARA overexpression. Knockdown of ZEB1 suppressed the RARA-mediated EMT phenotype. These results suggest that overexpression of RARA enhances malignant transformation during mammary tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Epithelial-Mesenchymal Transition , Mammary Glands, Human/metabolism , Neoplasm Proteins/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Glands, Human/pathology , Neoplasm Proteins/genetics , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha
19.
Cell Mol Life Sci ; 72(11): 2187-98, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25516021

ABSTRACT

The pharmacological modulation of hypoxia-inducible factor-1α (HIF-1α) and HIF-1α-regulated vascular endothelial growth factor-A (VEGF-A) in the kidney has therapeutic interest. Although it is assumed that prostaglandin E(2) (PGE(2)) exerts its biological effects from the extracellular medium through activation of EP receptors located at the cell membrane, we have shown in human renal proximal tubular HK-2 cells (and other cell lines) that intracellular PGE(2) regulates the expression of HIF-1α expression and the production of VEGF-A. Here, we have found--through experiments involving EP receptors agonists, EP receptor gene silencing and inhibition of the prostaglandin uptake transporter--that these biological effects of PGE(2) are mediated by intracellular EP(2) receptors. In sharp contrast with cell membrane EP(2), whose activation results in increased production of cAMP, intracellular EP(2) signaling was independent of cAMP. Instead, it involved c-src-dependent transactivation of epidermal growth factor receptor, which led to p38/ERK1/2-dependent activation of mitogen- and stress-activated kinase-1 (MSK-1) and to MSK-1-dependent-histone H3 phosphorylation and transcriptional up-regulation of retinoic acid receptor-ß. Even more important, this signaling pathway was fully reproduced in nuclei isolated from HK-2 cell, which highlights the relevance of nuclear EP receptors in the up-regulation of HIF-1α. These results open the possibility that signal cascades that proceed entirely in the cell nucleus might be responsible for several PGE(2) effects that are assumed to be due to cell membrane EP receptors.


Subject(s)
ErbB Receptors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Transcriptional Activation/genetics , Vascular Endothelial Growth Factor A/biosynthesis , Cell Nucleus/metabolism , Cyclic AMP/metabolism , Enzyme Activation , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Histones/metabolism , Humans , Kidney Tubules, Proximal/cytology , Phosphorylation , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Retinoic Acid/biosynthesis , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
20.
Tumour Biol ; 35(10): 10295-300, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25034531

ABSTRACT

The impairment of retinoic acid (RA)-dependent signaling is a frequent event during carcinogenesis. Cellular retinoic acid-binding proteins (CRABP1 and CRABP2) are important modulators of RA activity. Up to date, the role of these proteins in cancer progression remains poorly investigated. Here, we studied for the first time the simultaneous messenger RNA (mRNA) and protein expression of CRABPs in non-small cell lung cancer (NSCLC) samples. CRABP1 and CRABP2 mRNA levels were elevated in 42 and 56 % of NSCLC samples, respectively. Decrease of CRABP2 mRNA expression was significantly associated with the presence of lymph node metastases. Protein expression of CRABP1 and CRABP2 was detected in 50 and 56 % of tumor samples, respectively. We also found a positive correlation between CRABP1 and CRABP2 expression. Taken together, we demonstrated significant changes in CRABP expression in NSCLC samples. Importantly, the presented data provide the first evidence of potential involvement of CRABP2 in lung cancer metastasis.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Receptors, Retinoic Acid/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...