Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Metabolism ; 127: 154954, 2022 02.
Article in English | MEDLINE | ID: mdl-34875308

ABSTRACT

INTRODUCTION: Atherosclerotic Coronary Artery Disease (ASCAD) is the leading cause of mortality worldwide. Novel therapeutic approaches aiming to improve the atheroprotective functions of High Density Lipoprotein (HDL) include the use of reconstituted HDL forms containing human apolipoprotein A-I (rHDL-apoA-I). Given the strong atheroprotective properties of apolipoprotein E3 (apoE3), rHDL-apoE3 may represent an attractive yet largely unexplored therapeutic agent. OBJECTIVE: To evaluate the atheroprotective potential of rHDL-apoE3 starting with the unbiased assessment of global transcriptome effects and focusing on endothelial cell (EC) migration as a critical process in re-endothelialization and atherosclerosis prevention. The cellular, molecular and functional effects of rHDL-apoE3 on EC migration-associated pathways were assessed, as well as the potential translatability of these findings in vivo. METHODS: Human Aortic ECs (HAEC) were treated with rHDL-apoE3 and total RNA was analyzed by whole genome microarrays. Expression and phosphorylation changes of key EC migration-associated molecules were validated by qRT-PCR and Western blot analysis in primary HAEC, Human Coronary Artery ECs (HCAEC) and the human EA.hy926 EC line. The capacity of rHDL-apoE3 to stimulate EC migration was assessed by wound healing and transwell migration assays. The contribution of MEK1/2, PI3K and the transcription factor ID1 in rHDL-apoE3-induced EC migration and activation of EC migration-related effectors was assessed using specific inhibitors (PD98059: MEK1/2, LY294002: PI3K) and siRNA-mediated gene silencing, respectively. The capacity of rHDL-apoE3 to improve vascular permeability and hypercholesterolemia in vivo was tested in a mouse model of hypercholesterolemia (apoE KO mice) using Evans Blue assays and lipid/lipoprotein analysis in the serum, respectively. RESULTS: rHDL-apoE3 induced significant expression changes in 198 genes of HAEC mainly involved in re-endothelialization and atherosclerosis-associated functions. The most pronounced effect was observed for EC migration, with 42/198 genes being involved in the following EC migration-related pathways: 1) MEK/ERK, 2) PI3K/AKT/eNOS-MMP2/9, 3) RHO-GTPases, 4) integrin. rHDL-apoE3 induced changes in 24 representative transcripts of these pathways in HAEC, increasing the expression of their key proteins PIK3CG, EFNB2, ID1 and FLT1 in HCAEC and EA.hy926 cells. In addition, rHDL-apoE3 stimulated migration of HCAEC and EA.hy926 cells, and the migration was markedly attenuated in the presence of PD98059 or LY294002. rHDL-apoE3 also increased the phosphorylation of ERK1/2, AKT, eNOS and p38 MAPK in these cells, while PD98059 and LY294002 inhibited rHDL-apoE3-induced phosphorylation of ERK1/2, AKT and p38 MAPK, respectively. LY had no effect on rHDL-apoE3-mediated eNOS phosphorylation. ID1 siRNA markedly decreased EA.hy926 cell migration by inhibiting rHDL-apoE3-triggered ERK1/2 and AKT phosphorylation. Finally, administration of a single dose of rHDL-apoE3 in apoE KO mice markedly improved vascular permeability as demonstrated by the reduced concentration of Evans Blue dye in tissues such as the stomach, the tongue and the urinary bladder and ameliorated hypercholesterolemia. CONCLUSIONS: rHDL-apoE3 significantly enhanced EC migration in vitro, predominantly via overexpression of ID1 and subsequent activation of MEK1/2 and PI3K, and their downstream targets ERK1/2, AKT and p38 MAPK, respectively, and improved vascular permeability in vivo. These novel insights into the rHDL-apoE3 functions suggest a potential clinical use to promote re-endothelialization and retard development of atherosclerosis.


Subject(s)
Apolipoprotein E3/pharmacology , Endothelial Cells/drug effects , Lipoproteins, HDL/pharmacology , Animals , Apolipoprotein E3/metabolism , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/physiology , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/drug effects , Inhibitor of Differentiation Protein 1/genetics , Lipoproteins, HDL/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Mol Carcinog ; 60(2): 151-163, 2021 02.
Article in English | MEDLINE | ID: mdl-33428809

ABSTRACT

Regorafenib is approved for patients with unresectable hepatocellular carcinoma (HCC) following sorafenib. However, the effect of regorafenib on HCC metastasis and its mechanism are poorly understood. Here, our data showed that regorafenib significantly restrained the migration, invasion and vasculogenic mimicry (VM) of HCC cells, and downregulated the expression of epithelial-to-mesenchymal transition (EMT)/VM-related molecules. Using RNA-seq and cellular thermal shift assays, we found that inhibitor of differentiation 1 (ID1) was a key target of regorafenib. In HCC tissues, the protein expression of ID1 was positively correlated with EMT and VM formation (CD34- /PAS+ ). Functionally, ID1 knockdown inhibited HCC cell migration, invasion, metastasis, and VM formation in vitro and in vivo, with upregulation of E-cadherin and downregulation of Snail and VE-cadherin. Moreover, Snail overexpression promoted the migration, invasion, and VM formation of ID1 knockdown cells. Snail knockdown reduced the migration, invasion, and VM formation of ID1 overexpression cells. Finally, regorafenib suppressed VM formation and decreased the expression of ID1, VE-cadherin and Snail in HCC PDX model. In conclusion, we manifested that regorafenib distinctly inhibited EMT in HCC cells via targeting ID1, leading to the suppression of cell migration, invasion and VM formation. These findings suggest that regorafenib may be developed as a suitable therapeutic agent for HCC metastasis.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Liver Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Xenograft Model Antitumor Assays/methods , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Liver Neoplasms/blood supply , Liver Neoplasms/genetics , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Tumor Burden/drug effects , Tumor Burden/genetics
3.
Genes (Basel) ; 11(10)2020 09 27.
Article in English | MEDLINE | ID: mdl-32992599

ABSTRACT

Emerging evidence indicates that the host microRNAs (miRNAs) are important intracellular regulators and play pivotal roles in intricate host-pathogen interaction networks. In our previous studies, ssc-microRNA-4334-5p (miR-4334-5p) was identified as a differentially expressed miRNA in microarray-based miRNAs profiling experiment, but whether miR-4334-5p regulates foot and mouth disease virus (FMDV) propagation is less understood. Here, we demonstrated that miR-4334-5p expression level was up-regulated shortly after FMDV infection, transfection of miR-4334-5p mimics promoted, while inhibitor transfection suppressed FMDV replication correspondingly. Further bioinformatic analysis and experimental study suggested ID1 was the direct target of miR-4334-5p, suppressing FMDV replication by regulating interferon (IFN) pathways. These findings shed light on microRNAs-ID1-interferon axis in regulating FMDV replication.


Subject(s)
Foot-and-Mouth Disease Virus/pathogenicity , Foot-and-Mouth Disease/virology , Host-Pathogen Interactions , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Interferon Type I/antagonists & inhibitors , Kidney/virology , MicroRNAs/genetics , Animals , Cells, Cultured , Cricetinae , Foot-and-Mouth Disease/genetics , Foot-and-Mouth Disease/metabolism , Kidney/metabolism , MicroRNAs/metabolism , Signal Transduction , Swine
4.
Int J Med Sci ; 17(8): 995-1005, 2020.
Article in English | MEDLINE | ID: mdl-32410828

ABSTRACT

The inhibitor of DNA binding (Id) proteins are regulators of cell cycle and cell differentiation. Of all Id family proteins, Id1 is mostly linked to tumorigenesis, cellular senescence as well as cell proliferation and survival. Id1 is a stem cell-like gene more than a classical oncogene. Id1 is overexpressed in numerous types of cancers and exerts its promotion effect to these tumors through different pathways. Briefly, Id1 was found significantly correlated with EMT-related proteins, K-Ras signaling, EGFR signaling, BMP signaling, PI3K/Akt signaling, WNT and SHH signaling, c-Myc signaling, STAT3 signaling, RK1/2 MAPK/Egr1 pathway and TGF-ß pathway, etc. Id1 has potent effect on facilitating tumorous angiogenesis and metastasis. Moreover, high expression of Id1 plays a facilitating role in the development of drug resistance, including chemoresistance, radiation resistance and resistance to drugs targeting angiogenesis. However, controversial results were also obtained. Overall, Id1 represent a promising target of anti-tumor therapeutics based on its potent promotion effect to cancer. Numerous drugs were found exerting their anti-tumor function through Id1-related signaling pathways, such as fucoidan, berberine, tetramethylpyrazine, crizotinib, cannabidiol and vinblastine.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/pathology , Inhibitor of Differentiation Protein 1/metabolism , Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Senescence/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Neoplasms/drug therapy
5.
Cancer Res ; 79(16): 4057-4071, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31292163

ABSTRACT

Glioblastoma is the most common primary brain tumor in adults. While the introduction of temozolomide chemotherapy has increased long-term survivorship, treatment failure and rapid tumor recurrence remains universal. The transcriptional regulatory protein, inhibitor of DNA-binding-1 (ID1), is a key regulator of cell phenotype in cancer. We show that CRISPR-mediated knockout of ID1 in glioblastoma cells, breast adenocarcinoma cells, and melanoma cells dramatically reduced tumor progression in all three cancer systems through transcriptional downregulation of EGF, which resulted in decreased EGFR phosphorylation. Moreover, ID1-positive cells were enriched by chemotherapy and drove tumor recurrence in glioblastoma. Addition of the neuroleptic drug pimozide to inhibit ID1 expression enhanced the cytotoxic effects of temozolomide therapy on glioma cells and significantly prolonged time to tumor recurrence. Conclusively, these data suggest ID1 could be a promising therapeutic target in patients with glioblastoma. SIGNIFICANCE: These findings show that the transcriptional regulator ID1 is critical for glioblastoma initiation and chemoresistance and that inhibition of ID1 enhances the effect of temozolomide, delays tumor recurrence, and prolongs survival.


Subject(s)
Brain Neoplasms/drug therapy , Drug Resistance, Neoplasm/physiology , Glioblastoma/drug therapy , Inhibitor of Differentiation Protein 1/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , Melanoma/pathology , Mice, Inbred NOD , Phosphorylation , Pimozide/administration & dosage , Pimozide/pharmacology , Temozolomide/administration & dosage , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
6.
Cell Physiol Biochem ; 51(1): 487-500, 2018.
Article in English | MEDLINE | ID: mdl-30453304

ABSTRACT

BACKGROUND/AIMS: The underlying molecular mechanisms involved in sphingosine kinase 1 (SphK1)/sphingosine 1-phosphate (S1P) mediation of platelet-derived growth factor (PDGF)-induced pulmonary arterial smooth muscle cell (PASMC) proliferation are still unclear, and the present study aims to address this issue. METHODS: Small interfering RNA (siRNA) and microRNA inhibitor transfection was performed to block the expression of SphK1, bone morphogenetic protein receptor II (BMPRII) and microRNA-21 (miR-21). Gene expression levels of SphK1, BMPRII and inhibitor of DNA binding 1 (Id1) were detected by immunoblotting, miR-21 expression level was examined with qRT-PCR, and S1P production was measured by ELISA. Additionally, PASMC proliferation was determined by BrdU incorporation assay. RESULTS: Our results indicated that PDGF increased the expression of SphK1 protein and S1P production, up-regulated miR-21 expression, reduced BMPRII and Id1 expression, and promoted PASMCs proliferation. Pre-silencing of SphK1 with siRNA reversed PDGF-induced S1P production, miR-21 up-regulation, BMPRII and Id1 down-regulation, as well as PASMC proliferation. Pre-inhibition of miR-21 also blocked BMPRII and Id1 down-regulation as well as PASMC proliferation caused by PDGF. Knockdown of BMPRII down-regulated Id1 expression in PASMCs. We further found that inhibition of PI3K/Akt and ERK signaling pathways, particularly ERK cascade, suppressed PDGF-induced above changes. CONCLUSION: Our study indicates that SphK1/S1P pathway plays an important role in PDGF-induced PASMC proliferation via miR-21/BMPRII/Id1 axis and targeting against SphK1/S1P axis might be a novel strategy in the prevention and treatment of pulmonary arterial hypertension (PAH).


Subject(s)
Cell Proliferation/drug effects , Lysophospholipids/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Platelet-Derived Growth Factor/pharmacology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Animals , Antagomirs/metabolism , Bone Morphogenetic Protein Receptors, Type II/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/genetics , Pulmonary Artery/cytology , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Sphingosine/metabolism , Up-Regulation/drug effects
7.
J Photochem Photobiol B ; 178: 521-529, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29245122

ABSTRACT

The inhibitor of DNA binding and cell differentiation (Id) proteins are dominant negative regulators of the helix-loop-helix transcription factor family and play a key role during development as well as in vascular disorders and cancer. In fact, impairing the Id-protein activity in cancer cells reduces cell growth and even chemoresistance. Recently, we have shown that a synthetic Id-protein ligand (1Y) consisting of a cyclic nonapeptide can reduce the viability of the two breast cancer cell lines MCF-7 and T47D and of the bladder cancer cells T24 to about 50% at concentrations ≥100µM. Moreover, the cyclopeptide displays both proapoptotic and antiproliferative effects on MCF-7 cells. Herein, we show that the cyclopeptide does not induce cell death at the dose of 5µΜ, but it still inhibits MCF-7 and T24 cell proliferation, which correlates with an increased protein level of the cell-cycle regulator p27Kip1. Furthermore, 1Y-pretreated MCF-7, T47D, and T24 cells are more susceptible than untreated cells to the phototoxic effects of the three photosensitizers meta-tetra(hydroxyphenyl)chlorin, porfimer sodium, and hypericin, which are applied in photodynamic therapy (PDT). The combination of the Id-protein ligand with each of the light-activated photosensitizers shows synergistic effects on the reduction of cell viability. In conclusion, an Id-protein ligand with moderate cancer cell killing activity at concentrations ≥100µM can be applied at a 20-fold lower and barely toxic dose to raise the sensitivity of cancer cells towards phototoxicity associated with photodynamic treatment. This suggests the potential benefit of targeting the Id proteins in combined drug approaches for cancer therapy.


Subject(s)
Cell Proliferation/drug effects , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Peptides, Cyclic/toxicity , Photosensitizing Agents/toxicity , Anthracenes , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Dihematoporphyrin Ether/toxicity , Drug Synergism , Humans , Inhibitor of Differentiation Protein 1/metabolism , Light , MCF-7 Cells , Nanostructures/chemistry , Nanostructures/toxicity , Peptides, Cyclic/chemistry , Perylene/analogs & derivatives , Perylene/toxicity , Photosensitizing Agents/chemistry , Porphyrins/toxicity
8.
Acta Biochim Pol ; 64(2): 315-322, 2017.
Article in English | MEDLINE | ID: mdl-28510612

ABSTRACT

Adjuvant chemotherapy with 5-fluorouracil remains the basic treatment for patients with advanced colorectal carcinoma. The major obstacle in successful treatment is the ability of CRC cells to acquire chemoresistance. Here we examined the impact of ID1 silencing on the sensitivity of CRC cells to 5-FU. To suppress ID1 expression in HT-29 and HCT-116 cells the cells were transduced with a lentiviral vector carrying the ID1 silencing sequence. Cells with silenced ID1 showed altered expression of epithelial and mesenchymal markers and exhibited increased proliferation rate compared to the parental cells. HCT-116 cells with suppressed ID1 became sensitized to 5-FU and this was not observed in HT-29 cells. Silencing ID1 resulted in altered expression of genes encoding enzymes metabolizing 5-FU. HT-29 cells with suppressed ID1 had significantly reduced mRNA level for thymidine phosphorylase, uridine-cytydine kinase 2 and dihydropyrimidine dehydrogenase. ID1 suppression in HCT-116 cells resulted in an increase of mRNA level for thymidine phosphorylase, thymidine kinase and uridine-cytydine kinase 2 with concurrent drop of dihydropyrimidine dehydrogenase and thymidylate synthetase mRNA levels. In conclusion, ID1 expression impacts the sensitivity of colon cancer cells to 5-FU and may be considered as a potential predictive marker in CRC treatment.


Subject(s)
Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Fluorouracil/administration & dosage , Inhibitor of Differentiation Protein 1/genetics , Aged , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dihydrouracil Dehydrogenase (NADP)/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Male , RNA, Messenger/biosynthesis , Thymidine Phosphorylase/genetics , Uridine Kinase/genetics
9.
Int J Oncol ; 49(5): 1953-1962, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27633352

ABSTRACT

Ribosomal protein L9 (RPL9), a component of the 60S subunit for protein synthesis, is upregulated in human colorectal cancer. In the present study, we investigated whether RPL9 gained extraribosomal function during tumorigenesis and whether targeting of RPL9 with small interfering (si) RNA could alter the course of colorectal cancer progression. Our results showed that siRNA knockdown of RPL9 suppresses colorectal cancer (CRC) cell growth and long-term colony formation through an increase in sub-G1 cell population and a strong induction of apoptotic cell death. To obtain insights into the molecular changes in response to RPL9 knockdown, global changes in gene expression were examined using RNA sequencing. It revealed that RPL9-specific knockdown led to dysregulation of 918 genes in HCT116 and 3178 genes in HT29 cells. Among these, 296 genes showed same directional regulation (128 upregulated and 168 downregulated genes) and were considered as a common RPL9 knockdown signature. Particularly, we found through a network analysis that Id-1, which is functionally associated with activation of NF-κB and cell survival, was commonly downregulated. Subsequent western blot analysis affirmed that RPL9 silencing induced the decrease in the levels of Id-1 and phosphorylated IκBα in both HCT116 and HT29 cells. Also, the same condition decreased the levels of PARP-1 and pro-caspase-3, accelerating apoptosis. Furthermore, inhibition of RPL9 expression significantly suppressed the growth of human CRC xenografts in nude mice. These findings indicate that the function of RPL9 is correlated with Id-1/NF-κB signaling axis and suggest that targeting RPL9 could be an attractive option for molecular therapy of colorectal cancer.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Ribosomal Proteins/antagonists & inhibitors , Animals , Apoptosis , Blotting, Western , Cell Cycle , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Immunoenzyme Techniques , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
J Exp Clin Cancer Res ; 35: 57, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029190

ABSTRACT

BACKGROUND: Gastric cancer remains a major cause of mortality and morbidity worldwide. In recent years, gene-based therapeutic strategies were confirmed promising in cancer inhibition and attracted great attention. RNA interference (RNAi) is a powerful tool for gene therapy and has been widely employed to aid in treatment for various diseases, especially cancers. However, effective delivery of small interfering RNA (siRNA) to target cells in vivo remains a challenge for that it is prone to degradation and only lasts a few days in rapidly dividing cells. METHODS: Due to its biocompatibility and well-established safety profile, collagen represents a favourable matrix for in-site drug delivery. In the study, collagen hydrogel was used as carriers to test the feasibility of localized and sustained delivery of Id1-targeted siRNA for in vivo gastric cancer inhibition. To enhance the siRNA delivery, cationic polyethylenimine (PEI) was further emplored for scallold modification. The efficacy of siRNA delivery and cancer inhibition were evaluated with multimodality of mehods in vitro and in vivo. RESULTS: Our results showed that addition of polyethylenimine (PEI) to collagen can facilitate entry of Id1-siRNA into target cells, prolong the silencing effect, and further inhibit tumor growth both in vitro and in vivo. CONCLUSION: This collagen-based delivery system may facilitate the pathogenesis elucidation and design of effective therapies against gastric cancer.


Subject(s)
Cell Cycle/drug effects , Genetic Therapy/methods , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , Stomach Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrogels/administration & dosage , Hydrogels/chemistry , Mice , Mice, Nude , Polyethyleneimine/chemistry , RNA, Small Interfering/pharmacology , Stomach Neoplasms/genetics , Xenograft Model Antitumor Assays
11.
J Cell Sci ; 128(12): 2340-50, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25948585

ABSTRACT

Rett syndrome (RTT) is a progressive neurological disorder caused by mutations in the X-linked protein methyl-CpG-binding protein 2 (MeCP2). The endogenous function of MeCP2 during neural differentiation is still unclear. Here, we report that mecp2 is required for brain development in zebrafish. Mecp2 was broadly expressed initially in embryos and enriched later in the brain. Either morpholino knockdown or genetic depletion of mecp2 inhibited neuronal differentiation, whereas its overexpression promoted neuronal differentiation, suggesting an essential role of mecp2 in directing neural precursors into differentiated neurons. Mechanistically, her2 (the zebrafish ortholog of mammalian Hes5) was upregulated in mecp2 morphants in an Id1-dependent manner. Moreover, knockdown of either her2 or id1 fully rescued neuronal differentiation in mecp2 morphants. These results suggest that Mecp2 plays an important role in neural cell development by suppressing the Id1-Her2 axis, and provide new evidence that embryonic neural defects contribute to the later motor and cognitive dysfunctions in RTT.


Subject(s)
Cell Differentiation , Embryo, Nonmammalian/cytology , Genes, erbB-2 , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Methyl-CpG-Binding Protein 2/metabolism , Neurons/cytology , Zebrafish/metabolism , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Base Sequence , Blotting, Western , Brain/cytology , Brain/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Nonmammalian/metabolism , Immunoenzyme Techniques , Immunoprecipitation , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Methyl-CpG-Binding Protein 2/genetics , Mice , Molecular Sequence Data , Neurogenesis/physiology , Neurons/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
12.
Biochimie ; 112: 139-50, 2015 May.
Article in English | MEDLINE | ID: mdl-25778840

ABSTRACT

The four known ID proteins (ID1-4, Inhibitor of Differentiation) share a homologous helix loop helix (HLH) domain and act as dominant negative regulators of basic-HLH transcription factors. ID proteins also interact with many non-bHLH proteins in complex networks. The expression of ID proteins is increasingly observed in many cancers. Whereas ID-1, ID-2 and ID-3, are generally considered as tumor promoters, ID4 on the contrary has emerged as a tumor suppressor. In this study we demonstrate that ID4 heterodimerizes with ID-1, -2 and -3 and promote bHLH DNA binding, essentially acting as an inhibitor of inhibitors of differentiation proteins. Interaction of ID4 was observed with ID1, ID2 and ID3 that was dependent on intact HLH domain of ID4. Interaction with bHLH protein E47 required almost 3 fold higher concentration of ID4 as compared to ID1. Furthermore, inhibition of E47 DNA binding by ID1 was restored by ID4 in an EMSA binding assay. ID4 and ID1 were also colocalized in prostate cancer cell line LNCaP. The alpha helix forming alanine stretch N-terminal, unique to HLH ID4 domain was required for optimum interaction. Ectopic expression of ID4 in DU145 prostate cancer line promoted E47 dependent expression of CDKNI p21. Thus counteracting the biological activities of ID-1, -2 and -3 by forming inactive heterodimers appears to be a novel mechanism of action of ID4. These results could have far reaching consequences in developing strategies to target ID proteins for cancer therapy and understanding biologically relevant ID-interactions.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Inhibitor of Differentiation Protein 1 , Inhibitor of Differentiation Protein 2 , Inhibitor of Differentiation Proteins , Neoplasm Proteins , Prostatic Neoplasms , Transcription, Genetic , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Protein 2/antagonists & inhibitors , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/antagonists & inhibitors , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
13.
Oncogene ; 34(15): 1968-78, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-24882581

ABSTRACT

The previous studies in this lab discovered that microRNA-885-3p (miR-885-3p) was regulated by a sulfated polysaccharide that bound to bone morphogenetic protein receptor, type IA (BMPR1A) to inhibit angiogenesis. However, its specific role and its mechanism of action in tumor cells have not been elucidated. We show that miR-885-3p markedly suppresses angiogenesis in vitro and in vivo. MiR-885-3p inhibits Smad1/5/8 phosphorylation and downregulates DNA-binding protein inhibitor ID-1 (Id1), a proangiogenic factor, by targeting BMPR1A, leading to impaired angiogenesis. Overexpression or silencing of BMPR1A affects angiogenesis in a Smad/Id1-dependent manner. We further show that miR-885-3p impairs the growth of HT-29 colon cancer cell xenografts in nude mice by suppressing angiogenesis through disruption of BMPR1A and Smad/Id1 signaling. These results support a novel role for miR-885-3p in tumor angiogenesis by targeting BMPR1A, which regulates a proangiogenic factor, and provide new evidence that targeting miRNAs might be an effective therapeutic strategy for improving colon cancer treatment.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Colonic Neoplasms/blood supply , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , MicroRNAs/metabolism , Smad Proteins/antagonists & inhibitors , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/therapy , Female , HEK293 Cells , HT29 Cells , Heterografts , Humans , Inhibitor of Differentiation Protein 1/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/administration & dosage , MicroRNAs/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Phosphorylation , Signal Transduction , Smad Proteins/metabolism , Transfection
14.
Blood Cells Mol Dis ; 53(4): 204-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25065484

ABSTRACT

Hepcidin transcription is strongly down-regulated under hypoxic conditions, however whether hypoxia inhibits hepcidin directly or indirectly is still unknown. We investigated the time course of hypoxia-mediated hepcidin down-regulation in vivo in healthy volunteers exposed to hypobaric hypoxia at high altitude and, based on the hypothesis that circulating factors are implicated in hepcidin inhibition, we analyzed the effect of sera of these volunteers exposed to normoxia and hypoxia on hepcidin expression in Huh-7 cell lines. Hypoxia led to a significant hepcidin down-regulation in vivo that was almost complete within 72h of exposure and followed erythropoietin induction. This delay in hepcidin down-regulation suggests the existence of soluble factor/s regulating hepcidin production. We then stimulated HuH-7 cells with normoxic and hypoxic sera to analyze the effects of sera on hepcidin regulation. Hypoxic sera had a significant inhibitory effect on hepcidin promoter activity assessed by a luciferase assay, although the amount of such decrease was not as relevant as that observed in vivo. Cellular mRNA analysis showed that a number of volunteers' sera inhibited hepcidin expression, concurrently with ID1 inhibition, suggesting that inhibitory factor(s) may act through the SMAD-pathway.


Subject(s)
Biological Factors/pharmacology , Hepatocytes/drug effects , Hepcidins/genetics , Hypoxia/blood , RNA, Messenger/genetics , Adult , Altitude , Biological Factors/blood , Cell Line , Erythropoietin/genetics , Erythropoietin/metabolism , Female , Gene Expression Regulation , Genes, Reporter , Hepatocytes/cytology , Hepatocytes/metabolism , Hepcidins/antagonists & inhibitors , Hepcidins/metabolism , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Luciferases/genetics , Luciferases/metabolism , Male , Promoter Regions, Genetic , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins/genetics , Smad Proteins/metabolism
15.
Biochim Biophys Acta ; 1844(9): 1675-83, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24981796

ABSTRACT

The inhibitors of DNA binding Id1-4 are helix-loop-helix (HLH) proteins that exert their biological function by interacting with members of the basic-HLH (bHLH) transcription-factor family. The HLH domains of the Id and bHLH proteins allow both self- and hetero-association. Due to their abnormal expression in cancer cells, the Id proteins are potential protein targets for cancer treatment. Suitable Id-protein inactivators should promote self-association and/or prevent hetero-association. In this work we evaluated the ability of the Id-protein HLH domain to recognize itself in form of short sequences extracted from the helical and loop regions. We performed a peptide scan of the Id1 HLH domain 64-106 based on three-residue overlapping octapeptides. Interaction of each octapeptide with the natively folded Id1 HLH domain was investigated by CD and fluorescence spectroscopy. The results from both techniques showed that the helix-based but not the loop-based octapeptides interacted with the Id1 HLH domain in the low-micromolar range. In contrast, a nitrotyrosine-containing analog of the Id1 HLH region, which was unable to reproduce the native-like conformation, quenched only the 2-amino-benzoyl-(Abz)-labeled loop-based octapeptides. This opposite self-recognition pattern suggests that the short helix-based and loop-based sequences should be able to distinguish different folding states of the Id1 HLH domain. This feature may be biologically relevant, as the Id proteins are predicted to behave as intrinsically disordered proteins, being in equilibrium between rapidly exchanging monomeric conformations and structurally better-defined homo-/heterodimers displaying the parallel four-helix bundle.


Subject(s)
Helix-Loop-Helix Motifs , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Oligopeptides/chemistry , Amino Acid Sequence , Circular Dichroism , High-Throughput Screening Assays , Humans , Inhibitor of Differentiation Protein 1/chemistry , Kinetics , Ligands , Molecular Sequence Data , Oligopeptides/chemical synthesis , Protein Binding , Protein Structure, Tertiary , Spectrometry, Fluorescence , Staining and Labeling , Tyrosine/analogs & derivatives , Tyrosine/chemistry , ortho-Aminobenzoates/chemistry
16.
J Pept Sci ; 19(11): 676-83, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24123584

ABSTRACT

Id proteins, inhibitors of DNA binding proteins, have highly conserved dimerization motif known as the helix-loop-helix (HLH) domain that acts as a negative regulator of basic HLH (bHLH) transcription factors. In signaling pathways, Id proteins play an important role in cellular development, proliferation, and differentiation. The mechanism of Id proteins is to antagonize bHLH proteins, thereby preventing them from binding to DNA and inhibiting transcription of cellular differentiation-associated genes in cancer. Recently, we reported an inhibitor of Id1, peptide 3C, which showed good affinity to Id1 protein and exhibited inhibitory effects in cancer cells. In this study, Ala (A)-substituted analogs of peptide 3C were synthesized by SPPS, purified by RP-HPLC, and characterized by MALDI-TOF MS. Binding of each peptide to Id1 or Id1-HLH (the HLH domain of Id1) was monitored by surface plasmon resonance (SPR)-based biosensor. Biological effect of each peptide in MCF-7 breast cancer cells was analyzed by MTT cell viability assay. The secondary structure of substituted analogs of peptide 3C was investigated by circular dichroism (CD) spectroscopy. SPR results revealed that A-substituted analogs of peptide 3C showed weaker binding to Id1 than that of peptide 3C, indicating that the six amino acid residues in the N-terminal of peptide 3C were all essential for binding to Id1 and the importance of amino acid residue was I(2) > Q(6) > Y(1) > G(4) > L(5) > E(3). In addition, substitution of E(3) in peptide 3C with D, Q, and R did not improve the binding potency of peptide 3C. MTT assay demonstrated that neither A-substituted nor position 3-substituted analogs of peptide 3C showed increased antiproliferative effect in MCF-7 cancer cells. CD results indicated that peptide 3C exhibited the highest content of α-helical structure (39.37%), suggesting that the α-helical structure may contribute to its binding potency for Id1 and Id1-HLH. SAR results provided important information for the development of peptidic inhibitors of Id1 as anticancer agents and demonstrated peptide 3C as a promising lead for further modifications.


Subject(s)
Antineoplastic Agents/pharmacology , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , MyoD Protein/chemistry , Peptide Fragments/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , MCF-7 Cells , Peptide Fragments/chemical synthesis , Protein Binding , Protein Structure, Secondary , Solid-Phase Synthesis Techniques , Structure-Activity Relationship
17.
Cancer Res ; 73(5): 1559-69, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23243024

ABSTRACT

Glioblastoma is the most common form of primary adult brain tumors. A majority of glioblastomas grow invasively into distant brain tissue, leading to tumor recurrence, which is ultimately incurable. It is, therefore, essential to discover master regulators that control glioblastoma invasiveness and target them therapeutically. We show here that the transcriptional regulator Id-1 plays a critical role in modulating the invasiveness of glioblastoma cell lines and primary glioblastoma cells. Id-1 expression levels positively correlate with glioma cell invasiveness in culture and with histopathologic grades in patient biopsies. Id-1 knockdown dramatically reduces glioblastoma cell invasion that is accompanied by profound morphologic changes and robust reduction in expression levels of "mesenchymal" markers, as well as inhibition of self-renewal potential and downregulation of glioma stem cell markers. Importantly, genetic knockdown of Id-1 leads to a significant increase in survival in an orthotopic model of human glioblastoma. Furthermore, we show that a nontoxic compound, cannabidiol, significantly downregulates Id-1 gene expression and associated glioma cell invasiveness and self-renewal. In addition, cannabidiol significantly inhibits the invasion of glioblastoma cells through an organotypic brain slice and glioma progression in vivo. Our results suggest that Id-1 regulates multiple tumor-promoting pathways in glioblastoma and that drugs targeting Id-1 represent a novel and promising strategy for improving the therapy and outcome of patients with glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Inhibitor of Differentiation Protein 1/physiology , Neoplasm Invasiveness/genetics , Animals , Brain Neoplasms/pathology , Cannabidiol/pharmacology , Cell Line, Tumor , Female , Glioblastoma/pathology , Humans , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/metabolism , Mice , Mice, Nude , Neurospora , RNA Interference , Transplantation, Heterologous , Up-Regulation
18.
J Mol Model ; 18(11): 4865-84, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22714536

ABSTRACT

Among the many abnormally expressed proteins in ovarian cancer, the prominent cancer in women, ID1 (inhibitors of DNA binding protein 1) is a potential one among other several targets. Interaction of ID1 with ETS-1 (transcriptional activator of p16(INK4a)) suppresses the transcription of p16(INK4a) and causes abnormal cell proliferation. A peptide aptamer (ID1/3-PA7) has been designed to prevent this interaction and thereby leading to the transcription of p16(INK4a). However, the structural basis behind the molecular interaction of ID1 with ETS-1 (agonist) and ID1/3-PA7 (antagonist) is poorly understood. In order to understand this structural recognition and their interaction mechanism, in silico methods were used. From this interaction analysis, the residues of ETS-1 involved in interaction with the p16(INK4a) promoter were found to be targeted by ID1. Subsequently, ETS-1 binding residues of ID1 were found to be targeted by its aptamer- ID1/3-PA7. These results suggest that both ETS-1 and ID1/3-PA7 binds at the same region harbored by the residues-H97, D100, R103, D104, L107, A144, C145, D149, D150 and C154 of ID1. All these observations correlate with the experimental reports, suggesting that the identified residues might play a crucial role in promulgating the oncogenic effects of ID1. In silico alanine scanning mutagenesis also confirms the role of identified hot spot residues in p16(INK4a) regulation. Finally, the molecular dynamic simulation studies reveal the prolonged stability of the aforementioned interacting complexes. The obtained results throw light on the structure and residues of ID1 involved in transcriptional regulation of p16(INK4a).


Subject(s)
Aptamers, Peptide/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/chemistry , Proto-Oncogene Protein c-ets-1/metabolism , Aptamers, Peptide/pharmacology , Binding Sites , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , Drug Design , Female , Humans , Inhibitor of Differentiation Protein 1/agonists , Inhibitor of Differentiation Protein 1/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Ovarian Neoplasms/drug therapy , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Protein c-ets-1/chemistry , Transcription, Genetic
19.
Mol Med Rep ; 5(4): 1075-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22245935

ABSTRACT

Inhibitor of differentiation or DNA binding (Id1) is a member of the helix-loop-helix transcription factor family that is overexpressed in various types of cancer, including gastric carcinoma. Previous studies showed that Id1 is a prognostic marker in patients with gastric cancer. However, the role of Id1 in the proliferation of human gastric cancer cells has yet to be clarified. In the present study, we downregulated the Id1 gene in SGC-7901 gastric cancer cells by RNA interference, and we also constructed a recombinant plasmid-expressing Id1 to investigate its effects on the proliferation of SGC-7901 cells. Results showed that the downregulation of Id1 inhibited proliferation of SGC-7901 cells, while the upregulation of Id1 had no effect on SGC-7901 cell proliferation. The potential mechanism was also investigated. The changes of certain proteins associated with cell proliferation, apoptosis and the cell cycle were detected by western blotting. Furthermore, we demonstrated a positive correlation between Id1 and phospho-Akt expression in SGC-7901 cells.


Subject(s)
Down-Regulation , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Cell Division , Cell Line, Tumor , Cell Proliferation/drug effects , G2 Phase , Humans , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Signal Transduction , Stomach Neoplasms/metabolism
20.
Diabetes ; 60(10): 2506-14, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21940780

ABSTRACT

OBJECTIVE: The molecular mechanisms responsible for pancreatic ß-cell dysfunction in type 2 diabetes remain unresolved. Increased expression of the helix-loop-helix protein Id1 has been found in islets of diabetic mice and in vitro models of ß-cell dysfunction. Here, we investigated the role of Id1 in insulin secretion and glucose homeostasis. RESEARCH DESIGN AND METHODS: Id1 knockout (Id1(-/-)) and wild-type mice were fed a chow or high-fat diet. Glucose tolerance, insulin tolerance, ß-cell mass, insulin secretion, and islet gene expression were assessed. Small interfering RNA (siRNA) was used to silence Id1 in MIN6 cells, and responses to chronic palmitate treatment were assessed. RESULTS: Id1(-/-) mice exhibited an improved response to glucose challenge and were almost completely protected against glucose intolerance induced by high-fat diet. This was associated with increased insulin levels and enhanced insulin release from isolated islets, whereas energy intake, body weight, fat pad weight, ß-cell mass, and insulin action were unchanged. Islets from Id1(-/-) mice displayed reduced stress gene expression and were protected against high-fat diet-induced downregulation of ß-cell gene expression (pancreatic duodenal homeobox-1, Beta2, Glut2, pyruvate carboxylase, and Gpr40). In MIN6 cells, siRNA-mediated inhibition of Id1 enhanced insulin secretion after chronic palmitate treatment and protected against palmitate-mediated loss of ß-cell gene expression. CONCLUSIONS: These findings implicate Id1 as a negative regulator of insulin secretion. Id1 expression plays an essential role in the etiology of glucose intolerance, insulin secretory dysfunction, and ß-cell dedifferentiation under conditions of increased lipid supply.


Subject(s)
Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Insulin-Secreting Cells/metabolism , Animals , Blood Glucose , Cell Line , Gene Expression Regulation/physiology , Inhibitor of Differentiation Protein 1/antagonists & inhibitors , Inhibitor of Differentiation Protein 1/genetics , Insulin/blood , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Knockout , Palmitic Acid/pharmacology , RNA, Small Interfering/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL