Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Virol ; 96(13): e0057722, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35730977

ABSTRACT

Despite effective antiretroviral therapy, HIV-1 persistence in latent reservoirs remains a major obstacle to a cure. We postulate that HIV-1 silencing factors suppress HIV-1 reactivation and that inhibition of these factors will increase HIV-1 reactivation. To identify HIV-1 silencing factors, we conducted a genome-wide CRISPR inhibition (CRISPRi) screen using four CRISPRi-ready, HIV-1-d6-GFP-infected Jurkat T cell clones with distinct integration sites. We sorted cells with increased green fluorescent protein (GFP) expression and captured single guide RNAs (sgRNAs) via targeted deep sequencing. We identified 18 HIV-1 silencing factors that were significantly enriched in HIV-1-d6-GFPhigh cells. Among them, SLTM (scaffold attachment factor B-like transcription modulator) is an epigenetic and transcriptional modulator having both DNA and RNA binding capacities not previously known to affect HIV-1 transcription. Knocking down SLTM by CRISPRi significantly increased HIV-1-d6-GFP expression (by 1.9- to 4.2-fold) in three HIV-1-d6-GFP-Jurkat T cell clones. Furthermore, SLTM knockdown increased the chromatin accessibility of HIV-1 and the gene in which HIV-1 is integrated but not the housekeeping gene POLR2A. To test whether SLTM inhibition can reactivate HIV-1 and further induce cell death of HIV-1-infected cells ex vivo, we established a small interfering RNA (siRNA) knockdown method that reduced SLTM expression in CD4+ T cells from 10 antiretroviral therapy (ART)-treated, virally suppressed, HIV-1-infected individuals ex vivo. Using limiting dilution culture, we found that SLTM knockdown significantly reduced the frequency of HIV-1-infected cells harboring inducible HIV-1 by 62.2% (0.56/106 versus 1.48/106 CD4+ T cells [P = 0.029]). Overall, our study indicates that SLTM inhibition reactivates HIV-1 in vitro and induces cell death of HIV-1-infected cells ex vivo. Our study identified SLTM as a novel therapeutic target. IMPORTANCE HIV-1-infected cells, which can survive drug treatment and immune cell killing, prevent an HIV-1 cure. Immune recognition of infected cells requires HIV-1 protein expression; however, HIV-1 protein expression is limited in infected cells after long-term therapy. The ways in which the HIV-1 provirus is blocked from producing protein are unknown. We identified a new host protein that regulates HIV-1 gene expression. We also provided a new method of studying HIV-1-host factor interactions in cells from infected individuals. These improvements may enable future strategies to reactivate HIV-1 in infected individuals so that infected cells can be killed by immune cells, drug treatment, or the virus itself.


Subject(s)
HIV Infections , HIV Seropositivity , HIV-1 , Virus Activation , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes , Chromatin/genetics , Chromatin/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Knockdown Techniques , HIV Infections/physiopathology , HIV Seropositivity/genetics , HIV-1/physiology , Humans , Jurkat Cells , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/metabolism , Virus Activation/genetics
2.
Cell Prolif ; 54(4): e13016, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33660290

ABSTRACT

OBJECTIVES: Mouse incisor mesenchymal stem cells (MSCs) have self-renewal ability and osteo/odontogenic differentiation potential. However, the mechanism controlling the continuous self-renewal and osteo/odontogenic differentiation of mouse incisor MSCs remains unclear. Special AT-rich sequence-binding protein 2 (SATB2) positively regulates craniofacial patterning, bone development and regeneration, whereas SATB2 deletion or mutation leads to craniomaxillofacial dysplasia and delayed tooth and root development, similar to bone morphogenetic protein (BMP) loss-of-function phenotypes. However, the detailed mechanism underlying the SATB2 role in odontogenic MSCs is poorly understood. The aim of this study was to investigate whether SATB2 can regulate self-renewal and osteo/odontogenic differentiation of odontogenic MSCs. MATERIALS AND METHODS: Satb2 expression was detected in the rapidly renewing mouse incisor mesenchyme by immunofluorescence staining, quantitative RT-PCR and Western blot analysis. Ad-Satb2 and Ad-siSatb2 were constructed to evaluate the effect of Satb2 on odontogenic MSCs self-renewal and osteo/odontogenic differentiation properties and the potential role of Satb2 with the osteogenic factor bone morphogenetic protein 9 (Bmp9) in vitro and in vivo. RESULTS: Satb2 was found to be expressed in mesenchymal cells and pre-odontoblasts/odontoblasts. We further discovered that Satb2 effectively enhances mouse incisor MSCs self-renewal. Satb2 acted synergistically with the potent osteogenic factor Bmp9 in inducing osteo/odontogenic differentiation of mouse incisor MSCs in vitro and in vivo. CONCLUSIONS: Satb2 promotes self-renewal and osteo/odontogenic differentiation of mouse incisor MSCs. Thus, Satb2 can cooperate with Bmp9 as a new efficacious bio-factor for osteogenic regeneration and tooth engineering.


Subject(s)
Cell Differentiation , Growth Differentiation Factor 2/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Mesenchymal Stem Cells/cytology , Odontoblasts/cytology , Transcription Factors/metabolism , Adenoviridae/genetics , Animals , Bone Regeneration , Cell Adhesion , Cell Line , Cell Proliferation , Cell Self Renewal , Genetic Vectors/genetics , Genetic Vectors/metabolism , Growth Differentiation Factor 2/genetics , Hydrogels/chemistry , Incisor/cytology , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Odontoblasts/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tissue Scaffolds/chemistry , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
3.
Nature ; 583(7817): 625-630, 2020 07.
Article in English | MEDLINE | ID: mdl-32669713

ABSTRACT

The recent discovery of N6-methyladenine (N6-mA) in mammalian genomes suggests that it may serve as an epigenetic regulatory mechanism1. However, the biological role of N6-mA and the molecular pathways that exert its function remain unclear. Here we show that N6-mA has a key role in changing the epigenetic landscape during cell fate transitions in early development. We found that N6-mA is upregulated during the development of mouse trophoblast stem cells, specifically at regions of stress-induced DNA double helix destabilization (SIDD)2-4. Regions of SIDD are conducive to topological stress-induced unpairing of the double helix and have critical roles in organizing large-scale chromatin structures3,5,6. We show that the presence of N6-mA reduces the in vitro interactions by more than 500-fold between SIDD and SATB1, a crucial chromatin organizer that interacts with SIDD regions. Deposition of N6-mA also antagonizes SATB1 function in vivo by preventing its binding to chromatin. Concordantly, N6-mA functions at the boundaries between euchromatin and heterochromatin to restrict the spread of euchromatin. Repression of SIDD-SATB1 interactions mediated by N6-mA is essential for gene regulation during trophoblast development in cell culture models and in vivo. Overall, our findings demonstrate an unexpected molecular mechanism for N6-mA function via SATB1, and reveal connections between DNA modification, DNA secondary structures and large chromatin domains in early embryonic development.


Subject(s)
Adenine/analogs & derivatives , DNA/chemistry , DNA/metabolism , Embryonic Development , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Adenine/metabolism , Animals , Base Pairing , Embryonic Development/genetics , Euchromatin/genetics , Euchromatin/metabolism , Female , Humans , Male , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Stem Cells/cytology , Stem Cells/metabolism , Thermodynamics , Trophoblasts/cytology
4.
Sci Rep ; 10(1): 8615, 2020 05 25.
Article in English | MEDLINE | ID: mdl-32451408

ABSTRACT

The Special AT-rich sequence binding protein 1 (SATB1) is a genome organizer protein that controls gene expression of numerous genes by regulating chromatin architecture and targeting chromatin-remodeling/-modifying enzymes onto specific chromatin regions. SATB1 is overexpressed in various tumors. In head and neck squamous cell carcinoma (HNSCC), SATB1 upregulation is correlated with TNM classification, metastasis, poor prognosis and reduced overall survival. In this paper, we comprehensively analyze cellular and molecular effects of SATB1 in a large set of primary cell lines from primary HNSCC or metastases, using RNAi-mediated knockdown in vitro and, therapeutically, in tumor xenograft mouse models in vivo. In a series of 15 cell lines, major differences in SATB1 levels are observed. In various 2-D and 3-D assays, growth inhibition upon efficient siRNA-mediated SATB1 knockdown depends on the cell line rather than initial SATB1 levels. Inhibitory effects are found to be based on cell cycle deceleration, apoptosis induction, decreased HER3 and Heregulin A&B expression, and effects on EMT genes. In vivo, systemic treatment of tumor xenograft-bearing mice with siRNAs formulated in polymeric nanoparticles inhibits tumor growth of two HNSCC xenograft models, resulting from therapeutic SATB1 reduction and concomitant decrease of proliferation and induction of apoptosis. In conclusion, SATB1 represents a promising target in HNSCC, affecting crucial cellular processes and molecular pathways.


Subject(s)
Head and Neck Neoplasms/pathology , Matrix Attachment Region Binding Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Line, Tumor , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Nanoparticles/chemistry , Neoplasm Staging , Neuregulin-1/genetics , Neuregulin-1/metabolism , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , RNA, Small Interfering/therapeutic use , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Transplantation, Heterologous , Up-Regulation
5.
Spine (Phila Pa 1976) ; 45(17): E1057-E1065, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32205703

ABSTRACT

STUDY DESIGN: Experimental analysis of the thoracic ligamentum flavum cell osteogenic differentiation process. OBJECTIVE: This study aimed to explore the role of miR-29a-5p and special AT-rich sequence-binding protein 2 (SATB2) in a pathological osteogenic process. SUMMARY OF BACKGROUND DATA: Thoracic ossification of the ligamentum flavum (TOLF) is an uncommon disease wherein ligaments within the spine undergo progressive ossification, resulting in stenosis of the spinal canal and myelopathy. MiR-29a-5p was found to be downregulated in ligament cells from ossified ligament tissue in a previous study. However, whether miR-29a-5p is involved in the process of TOLF has not been investigated. METHODS: The expression of miR-29a-5p in ligament tissues or in the context of TOLF osteogenic cell differentiation was measured via qRT-PCR. Alkaline phosphatase activity assay and Alizarin red staining were used to analyze cellular osteogenesis. The protein-level expression of SATB2, SIRT1, and Smad3 were measured via immunohistochemistry or western blotting. Dual luciferase reporter assays and western blotting were used to confirm that miR-29a targets SATB2. RESULTS: SATB2 was found to be upregulated and miR-29a-5p was downregulated in TOLF tissue. We additionally observed decreased miR-29a-5p expression during the process of TOLF osteogenic cell differentiation, and there was a marked reduction in the expression of key mediators of osteogenesis when miR-29a-5p was overexpressed. Consistent with this, when miR-29a-5p was inhibited this led to enhanced osteogenic cell differentiation of these cells. We further found miR-29a-5p to directly target and suppress the expression of SATB2. Knock-down of SATB2 was sufficient to reduce the ability of miR-29a-5p to inhibit osteogenesis, and this also led to decreased SIRT1 expression and Smad3 acetylation. CONCLUSION: Together our findings indicate that miR-29a-5p is able to prevent thoracic ligamentum flavum cell osteogenesis at least in part via targeting SATB2 and thereby suppressing the SIRT1/Smad3 deacetylation pathway. LEVEL OF EVIDENCE: N/A.


Subject(s)
Ligamentum Flavum/metabolism , Matrix Attachment Region Binding Proteins/biosynthesis , MicroRNAs/biosynthesis , Osteogenesis/physiology , Sirtuin 1/biosynthesis , Smad3 Protein/biosynthesis , Transcription Factors/biosynthesis , Acetylation , Adult , Aged , Cells, Cultured , Female , HEK293 Cells , Humans , Ligamentum Flavum/pathology , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Middle Aged , Signal Transduction/physiology , Sirtuin 1/antagonists & inhibitors , Smad3 Protein/antagonists & inhibitors , Thoracic Vertebrae/metabolism , Thoracic Vertebrae/pathology , Transcription Factors/antagonists & inhibitors
6.
Anticancer Agents Med Chem ; 19(14): 1728-1736, 2019.
Article in English | MEDLINE | ID: mdl-31284872

ABSTRACT

BACKGROUND: The 12-hydroxy-14-dehydroandrographolide (DP) is a predominant component of the traditional herbal medicine Andrographis paniculata (Burm. f.) Nees (Acanthaceae). Recent studies have shown that DP exhibits potent anti-cancer effects against oral and colon cancer cells. OBJECTIVE: This investigation examined the potential effects of DP against osteosarcoma cell. METHODS: A cell analyzer was used to measure cell viability. The cell growth and proliferation were performed by Flow cytometry and BrdU incorporation assay. The cell migration and invasion were determined by wound healing and transwell assay. The expression of EMT related proteins was examined by Western blot analysis. RESULTS: In this study, we found that DP treatment repressed osteosarcoma (OS) cell growth in a dose-dependent manner. DP treatment significantly inhibited OS cell proliferation by arresting the cell cycle at G2/M phase. In addition, DP treatment effectively inhibited the migration and invasion abilities of OS cells through wound healing and Transwell tests. Mechanistic studies revealed that DP treatment effectively rescued the epithelialmesenchymal transition (EMT), while forced expression of SATB2 in OS cells markedly reversed the pharmacological effect of DP on EMT. CONCLUSION: Our data demonstrated that DP repressed OS cell growth through inhibition of proliferation and cell cycle arrest; DP also inhibited metastatic capability of OS cells through a reversal of EMT by targeting SATB2. These findings demonstrate DP's potential as a therapeutic drug for OS treatment.


Subject(s)
Diterpenes/pharmacology , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Osteosarcoma/drug therapy , Transcription Factors/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Diterpenes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition/drug effects , Humans , Matrix Attachment Region Binding Proteins/metabolism , Molecular Structure , Osteosarcoma/metabolism , Osteosarcoma/pathology , Structure-Activity Relationship , Transcription Factors/metabolism , Wound Healing/drug effects
7.
Life Sci ; 222: 212-220, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30557545

ABSTRACT

AIMS: Fluvastatin reduces tumor proliferation and increased apoptotic activity in various cancers. Special AT-rich sequence binding protein 1 (SATB1) is a genome organizer that reprogrammes the gene transcription profiles of tumors to promote growth and metastasis. The antitumor effect and molecular mechanisms of fluvastatin on lung cancer is poorly understood. This study aimed to investigate the antitumor effect of fluvastatin on lung cancer and its possible mechanics. MAIN METHODS: Cell viability assay was used to examine the inhibition of fluvastatin on proliferation of H292 cells. In order to investigate the antitumor mechanics, SATB1 knock-down H292 cells was constructed by lentiviral transfection. RT-PCR and Western blot were performed to examine the effects of fluvastatin on expression of SATB1 and Wnt/ß-catenin signaling components. KEY FINDINGS: Fluvastatin significantly inhibited proliferation and invasion of H292 cells in a time- and dose-dependent manner and promoted the apoptosis (p < 0.05). The expression of SATB1 was down-regulated by fluvastatin in a dose-dependent manner. The proliferation and invasion of SATB1-shRNA cells was significantly suppressed, and the apoptosis was significantly enhanced (p < 0.05). We also show that the common target genes were regulated by SATB1 and Wnt/ß-catenin pathway simultaneously. There may be a functional link between SATB1 and Wnt/ß-catenin pathway. SIGNIFICANCE: We presented a possible mechanism of statins that fluvastatin significantly suppressed the in vitro tumor progression of H292 cells possibly by down-regulation of SATB1 via Wnt/ß-catenin pathway, which provided new therapeutic possibilities for more cancers driven by hyperexpression of SATB1 and Wnt/ß-catenin pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Down-Regulation/physiology , Fluvastatin/therapeutic use , Lung Neoplasms/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Phenotype , A549 Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Down-Regulation/drug effects , Fluvastatin/pharmacology , Humans , Lung Neoplasms/drug therapy , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Neoplasm Invasiveness/pathology
8.
Cell Death Dis ; 9(11): 1065, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30337520

ABSTRACT

Cancer-associated fibroblasts (CAFs), a dominant component of the pancreatic tumor microenvironment, are mainly considered as promotors of malignant progression, but the underlying molecular mechanism remains unclear. Here, we show that SDF-1 secreted by CAFs stimulates malignant progression and gemcitabine resistance in pancreatic cancer, partially owing to paracrine induction of SATB-1 in pancreatic cancer cells. CAF-secreted SDF-1 upregulated the expression of SATB-1 in pancreatic cancer cells, which contributed to the maintenance of CAF properties, forming a reciprocal feedback loop. SATB-1 was verified to be overexpressed in human pancreatic cancer tissues and cell lines by quantitative real-time PCR, western blot, and immunohistochemical staining, which correlated with tumor progression and clinical prognosis in pancreatic cancer patients. We found that SATB-1 knockdown inhibited proliferation, migration, and invasion in SW1990 and PANC-1 cells in vitro, whereas overexpression of SATB-1 in Capan-2 and BxPC-3 cells had the opposite effect. Immunofluorescence staining showed that conditioned medium from SW1990 cells expressing SATB-1 maintained the local supportive function of CAFs. Furthermore, downregulation of SATB-1 inhibited tumor growth in mouse xenograft models. In addition, we found that overexpression of SATB-1 in pancreatic cancer cells participated in the process of gemcitabine resistance. Finally, we investigated the clinical correlations between SDF-1 and SATB-1 in human pancreatic cancer specimens. In summary, these findings demonstrated that the SDF-1/CXCR4/SATB-1 axis may be a potential new target of clinical interventions for pancreatic cancer patients.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Carcinoma, Pancreatic Ductal/genetics , Chemokine CXCL12/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Matrix Attachment Region Binding Proteins/genetics , Pancreatic Neoplasms/genetics , Aged , Animals , Antimetabolites, Antineoplastic/therapeutic use , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL12/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Humans , Lymphatic Metastasis , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mice, Nude , Middle Aged , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Survival Analysis , Tumor Burden , Xenograft Model Antitumor Assays , Gemcitabine
9.
Bone ; 114: 137-143, 2018 09.
Article in English | MEDLINE | ID: mdl-29909059

ABSTRACT

Emerging evidence indicates that microRNAs (miRNAs, miRs) play diverse roles in the regulation of biological processes, including osteoblastic differentiation. In this study, we found that miR-383 is a critical regulator of osteoblastic differentiation. We showed that miR-383 was downregulated during osteoblastic differentiation of rat bone marrow mesenchymal stem cells (BMSCs). Overexpression of miR-383 suppressed osteoblastic differentiation of BMSCs by downregulating alkaline phosphatase (ALP), matrix mineralization, and mRNA and protein levels of RUNX2 and OCN, whereas a knockdown of miR-383 promoted osteoblastic differentiation in vitro. The results of in vivo analysis indicated that inhibition of miR-383 expression enhanced the efficacy of new bone formation in a rat calvarial defect model. Mechanistic experiments revealed that special AT-rich-sequence-binding protein 2 (Satb2) was a direct and functional target of miR-383. Knockdown of Satb2 had inhibitory effects resembling those of miR-383 on the osteoblast differentiation of rat BMSCs. In addition, the positive effect of miR-383 suppression on osteoblastic differentiation was apparently abrogated by Satb2 silencing. Collectively, these results indicate that miR-383 plays an inhibitory role in osteogenic differentiation of rat BMSCs and may act by targeting Satb2.


Subject(s)
Cell Differentiation/physiology , Matrix Attachment Region Binding Proteins/biosynthesis , Mesenchymal Stem Cells/metabolism , MicroRNAs/biosynthesis , Osteoblasts/metabolism , Osteogenesis/physiology , Transcription Factors/biosynthesis , Animals , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Transcription Factors/antagonists & inhibitors
10.
Cell Prolif ; 51(4): e12455, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29701273

ABSTRACT

OBJECTIVES: SATB2 has been shown to be markedly reduced in colorectal cancer (CRC) tissues relative to paired normal controls; however, the mechanism behind remains not well understood. To investigate why SATB2 was down-regulated in CRC, we attempted to analyse it from the angle of miRNA-mRNA modulation. MATERIALS AND METHODS: SATB2 expression was detected in CRC tissues using immunohistochemistry and verified using real-time PCR on mRNA level, followed by analysis of clinicopathological significance of its expression. Metastatic variation of CRC cells was evaluated both in vivo and in vitro. To find out the potential miRNA that directly regulate the SATB2, luciferase reporter assay was performed following the bioinformatic prediction. RESULTS: SATB2 was confirmed to be closely linked with the metastasis and shorter overall survival of CRC in our own cases. Silencing of SATB2 was shown to be able to promote the metastatic ability of CRC cells in vivo, enhancing the epithelial-mesenchymal transition (EMT). Mechanistically, miR-34c-5p was identified to be a novel miRNA that can directly modulate the SATB2. It turned out that the promoter of miR-34c-5p was methylated, which leads to the repression of miR-34c-5p in CRC. Treatment with 5-Aza-dC can reasonably and significantly restore the level of miR-34c-5p in CRC cells relative to control, thereby down-regulating the SATB2. CONCLUSIONS: Together, our study revealed that SATB2 targeted by methylated miR-34c-5p can suppress the metastasis, weakening the EMT in CRC.


Subject(s)
Cell Proliferation/physiology , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition/physiology , Matrix Attachment Region Binding Proteins/metabolism , MicroRNAs/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/mortality , DNA Methylation/drug effects , Decitabine , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Middle Aged , Prognosis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
11.
Eur Rev Med Pharmacol Sci ; 21(20): 4516-4528, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29131265

ABSTRACT

OBJECTIVE: To investigate the role of SATB2 in stem cell-like properties of osteosarcoma and identify new strategies to eliminate cancer stem cells of osteosarcoma. MATERIALS AND METHODS: Osteosarcoma cancer stem cells were derived by sarcosphere generation or chemo drug enrichment. SATB2 and pluripotency-associated gene expression in osteosarcoma CSCs were analyzed using qRT-PCR and Western blotting. The sphere formation assay, cell counting kit-8 assay and anti-chemotherapy proteins were used to measure the effects of altered SATB2, N-cadherin expression or metformin treatment in CSCs. Nude mice were injected with SATB2-deficient U2OS/MTX cells to assess the role of SATB2 in osteosarcoma growth and chemoresistance in vivo. Bioinformatics analyses were performed to identify SATB2 downstream target genes and immunochemistry to determine the correlation between SATB2 expression and patient outcome. Western blotting and luciferase reporter assays were used to examine the effects of N-cadherin and SATB2 inhibition on the NF-kB pathway. RESULTS: SATB2 was upregulated in osteosarcoma stem cells. Knockdown of SATB2 decreased sarcosphere formation, cell proliferation and stem cell-like gene expression in vitro, meanwhile reduced tumor growth and chemoresistance in vivo. High SATB2 expression in osteosarcoma patient samples was associated with poor clinical outcome. N-cadherin was one critical downstream target gene of SATB2 that mediated the stem cell-like phenotype. Reduction of SATB2 or N-cadherin resulted in NF-kB inactivation, which led to impaired osteosarcoma sphere formation and tumor cell proliferation. Metformin treatment of osteosarcoma cells enhanced the effects of chemotherapy via suppression of N-cadherin. CONCLUSIONS: SATB2 plays an important role in regulating osteosarcoma stem cell-like properties and tumor growth. The combination of conventional chemotherapy and metformin may be a promising therapeutic strategy for osteosarcoma patients.


Subject(s)
Bone Neoplasms/pathology , Cell Proliferation/drug effects , Matrix Attachment Region Binding Proteins/metabolism , Metformin/pharmacology , Osteosarcoma/pathology , Signal Transduction/drug effects , Transcription Factors/metabolism , Adolescent , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/mortality , Cadherins/metabolism , Female , Humans , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Nude , NF-kappa B/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Osteosarcoma/metabolism , Osteosarcoma/mortality , RNA Interference , RNA, Small Interfering/metabolism , Survival Rate , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Young Adult
12.
Stem Cell Res ; 19: 139-147, 2017 03.
Article in English | MEDLINE | ID: mdl-28167342

ABSTRACT

Breast tumors are heterogeneous and carry a small population of progenitor cells that can produce various subtypes of breast cancer. SATB2 (special AT-rich binding protein-2) is a newly identified transcription factor and epigenetic regulator. It is highly expressed in embryonic stem cells, but not in adult tissues, and regulates pluripotency-maintaining factors. However, the molecular mechanisms by which SATB2 induces transformation of human mammary epithelial cells (HMECs) leading to malignant phenotype are unknown. The main goal of this paper is to examine the molecular mechanisms by which SATB2 induces cellular transformation of HMECs into cells that are capable of self-renewal. SATB2-transformed HMECs gain the phenotype of breast progenitor cells by expressing markers of stem cells, pluripotency-maintaining factor, and epithelial to mesenchymal transition. SATB2 is highly expressed in human breast cancer cell lines, primary mammary tissues and cancer stem cells (CSCs), but not in HMECs and normal breast tissues. Chromatin Immunoprecipitation assays demonstrate that SATB2 can directly bind to promoters of Bcl-2, c-Myc, Nanog, Klf4, and XIAP, suggesting a role of SATB2 in regulation of pluripotency, cell survival and proliferation. Furthermore, inhibition of SATB2 by shRNA in breast cancer cell lines and CSCs attenuates cell proliferation and EMT phenotype. Our results suggest that SATB2 induces dedifferentiation/transformation of mature HMECs into progenitor-like cells.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/cytology , Matrix Attachment Region Binding Proteins/metabolism , Transcription Factors/metabolism , Cell Line , Cell Movement , Cell Proliferation , Chromatin Immunoprecipitation , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Kruppel-Like Factor 4 , Lentivirus/genetics , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
13.
BMC Cancer ; 17(1): 3, 2017 01 03.
Article in English | MEDLINE | ID: mdl-28049521

ABSTRACT

BACKGROUND: The Special AT-rich Sequence Binding Protein 1 (SATB1) regulates the expression of many genes by acting as a global chromatin organizer. While in many tumor entities SATB1 overexpression has been observed and connected to pro-tumorigenic processes, somewhat contradictory evidence exists in brain tumors with regard to SATB1 overexpression in glioblastoma and its association with poorer prognosis and tumor progression. On the functional side, initial data indicate that SATB1 may be involved in several tumor cell-relevant processes. METHODS: For the detailed analysis of the functional relevance and possible therapeutic potential of SATB1 inhibition, we employ transient siRNA-mediated knockdown and comprehensively analyze the cellular and molecular role of SATB1 in glioblastoma. RESULTS: In various cell lines with different SATB1 expression levels, a SATB1 gene dose-dependent inhibition of anchorage-dependent and -independent proliferation is observed. This is due to cell cycle-inhibitory and pro-apoptotic effects of SATB1 knockdown. Molecular analyses reveal SATB1 knockdown effects on multiple important (proto-) oncogenes, including Myc, Bcl-2, Pim-1, EGFR, ß-catenin and Survivin. Molecules involved in cell cycle, EMT and cell adhesion are affected as well. The putative therapeutic relevance of SATB1 inhibition is further supported in an in vivo tumor xenograft mouse model, where the treatment with polymeric nanoparticles containing SATB1-specific siRNAs exerts antitumor effects. CONCLUSION: Our results demonstrate that SATB1 may represent a promising target molecule in glioblastoma therapy whose inhibition or knockdown affects multiple crucial pathways.


Subject(s)
Brain Neoplasms/pathology , Brain/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Matrix Attachment Region Binding Proteins/metabolism , Animals , Apoptosis , Brain/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Cycle , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Nude , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
BMB Rep ; 49(10): 578-583, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27697109

ABSTRACT

Special AT-rich sequence binding protein 1 (SATB1) is a nuclear matrix-associated DNA-binding protein that functions as a chromatin organizer. SATB1 is highly expressed in aggressive breast cancer cells and promotes growth and metastasis by reprograming gene expression. Through genomewide cross-examination of gene expression and histone methylation, we identified SATB1 target genes for which expression is associated with altered epigenetic marks. Among the identified genes, long noncoding RNA urothelial carcinoma-associated 1 (UCA1) was upregulated by SATB1 depletion. Upregulation of UCA1 coincided with increased H3K4 trimethylation (H3K4me3) levels and decreased H3K27 trimethylation (H3K27me3) levels. Our study showed that SATB1 binds to the upstream region of UCA1 in vivo, and that its promoter activity increases with SATB1 depletion. Furthermore, simultaneous depletion of SATB1 and UCA1 potentiated suppression of tumor growth and cell survival. Thus, SATB1 repressed the expression of oncogenic UCA1, suppressing growth and survival of breast cancer cells. [BMB Reports 2016; 49(10): 578-583].


Subject(s)
Epigenesis, Genetic , Matrix Attachment Region Binding Proteins/metabolism , RNA, Long Noncoding/metabolism , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Flow Cytometry , Genes, Reporter , Histones/metabolism , Humans , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Methylation , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Up-Regulation
15.
Cell Rep ; 14(7): 1774-1786, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876172

ABSTRACT

Special AT-rich sequence-binding protein 1 (Satb1) governs genome-wide transcriptional programs. Using a conditional knockout mouse, we find that Satb1 is required for normal differentiation of conventional dendritic cells (DCs). Furthermore, Satb1 governs the differentiation of inflammatory DCs by regulating major histocompatibility complex class II (MHC II) expression through Notch1 signaling. Mechanistically, Satb1 binds to the Notch1 promoter, activating Notch expression and driving RBPJ occupancy of the H2-Ab1 promoter, which activates MHC II transcription. However, tumor-driven, unremitting expression of Satb1 in activated Zbtb46(+) inflammatory DCs that infiltrate ovarian tumors results in an immunosuppressive phenotype characterized by increased secretion of tumor-promoting Galectin-1 and IL-6. In vivo silencing of Satb1 in tumor-associated DCs reverses their tumorigenic activity and boosts protective immunity. Therefore, dynamic fluctuations in Satb1 expression govern the generation and immunostimulatory activity of steady-state and inflammatory DCs, but continuous Satb1 overexpression in differentiated DCs converts them into tolerogenic/pro-inflammatory cells that contribute to malignant progression.


Subject(s)
Dendritic Cells/immunology , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/immunology , Matrix Attachment Region Binding Proteins/immunology , Ovarian Neoplasms/immunology , Animals , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic , Dendritic Cells/pathology , Female , Galectin 1/genetics , Galectin 1/immunology , Histocompatibility Antigens Class II/genetics , Histones/genetics , Histones/immunology , Humans , Immune Tolerance , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Signal Transduction , Transcription Factors/genetics , Transcription Factors/immunology
16.
FEBS Lett ; 590(3): 396-407, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26785690

ABSTRACT

miRNAs play a number of roles in bone, including mediating the pathological effects of inflammation. Here, we found that miR-33a-5p expression was significantly increased after TNF-α treatment during BMP-2-induced osteogenic differentiation of hBMSCs. Luciferase reporter assays and western blotting demonstrated that special AT-rich sequence-binding protein 2 (SATB2) is a target of miR-33a-5p. Moreover, we show that BMP-2 induces SATB2 expression by interacting with SATB2 directly via the BMP-2-RUNX2 pathway. However, TNF-α first decreases SATB2 expression by inhibiting miR-33a-5p degradation. We thus conclude that miR-33a-5p plays a central role in this complex regulatory network. These findings will help to understand the regulatory role of miR-33a-5p in the inflammatory process.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Matrix Attachment Region Binding Proteins/metabolism , MicroRNAs/metabolism , Osteogenesis , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 2/genetics , Cell Proliferation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/agonists , Core Binding Factor Alpha 1 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Humans , Male , Matrix Attachment Region Binding Proteins/agonists , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Mice , MicroRNAs/antagonists & inhibitors , Recombinant Proteins/metabolism , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
17.
Tumour Biol ; 37(6): 7413-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26678884

ABSTRACT

Special AT-rich sequence-binding protein-1 (SATB1) is critical for genome organizer that reprograms chromatin organization and transcription profiles, and associated with tumor growth and metastasis in several cancer types. Many studies suggest that SATB1 overexpression is an indicator of poor prognosis in various cancers, such as breast cancer, malignant cutaneous melanoma, and liver cancer. However, their expression patterns and function values for adult T cell leukemia (ATL) are still largely unknown. The aim of this study is to examine the levels of SATB1 in ATL and to explore its function and mechanisms in Jurkat cell line. Here, we reported that SATB1 expressions were decreased in ATL cells (p < 0.001) compared with normal controls. Knockdown of SATB1 expression significantly enhanced invasion of Jurkat cell in vitro. Furthermore, knockdown of SATB1 gene enhances ß-catenin nuclear accumulation and transcriptional activity and thus may increase the invasiveness of Jurkat cell through the activation of Wnt/ß-catenin signaling pathway in vitro.


Subject(s)
Gene Expression Regulation, Neoplastic , Matrix Attachment Region Binding Proteins/physiology , Neoplasm Proteins/physiology , Wnt Signaling Pathway/physiology , Adult , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling , Humans , Jurkat Cells , Leukemia-Lymphoma, Adult T-Cell/blood , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia-Lymphoma, Adult T-Cell/pathology , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/biosynthesis , Matrix Attachment Region Binding Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , RNA, Small Interfering/genetics , Wnt Signaling Pathway/genetics
18.
Tumour Biol ; 36(11): 9073-81, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26084613

ABSTRACT

Recent studies suggest that SATB1 is a promising therapeutic target for prostate cancer. To develop novel SATB1-based therapeutic agents for prostate cancer, in this study, we aimed to construct ZD55-SATB1, an oncolytic adenovirus ZD55 carrying shRNA targeting SATB1, and investigate its effects on the inhibition of prostate cancer growth and metastasis. ZD55-SATB1 was constructed and used to infect human prostate cancer cell lines DU145 and LNCaP. The inhibitory effect of ZD55-SATB1 on SATB1 expression was evaluated by reverse transcription polymerase chain reaction (RT-PCR) and Western blot analysis. The cytotoxicity of ZD55-SATB1 was detected by MTT assay. Cell invasion was detected by Matrigel invasion assay. The in vivo antitumor activities of ZD55-SATB1 were evaluated in xenograft mouse model. We found that ZD55-SATB1 selectively replicated and significantly reduced SATB1 expression in DU145 and LNCaP cells. ZD55-SATB1 effectively inhibited the viability and invasion of DU145 and LNCaP cells in vitro and inhibited prostate cancer growth and metastasis in xenograft nude mice. In conclusion, replicative oncolytic adenovirus armed with SATB1 shRNA exhibits effective antitumor effect in human prostate cancer. Our study provides the basis for the development of ZD55-SATB1 for the treatment of prostate cancer.


Subject(s)
Cell Proliferation/genetics , Matrix Attachment Region Binding Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/biosynthesis , Mice , Oncolytic Viruses/genetics , Prostatic Neoplasms/pathology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
19.
FEBS Lett ; 589(12): 1359-68, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25896016

ABSTRACT

Liver fibrosis is a worldwide clinical issue. Activation of hepatic stellate cells (HSCs) is the central event during liver fibrosis. We investigated the role of SATB1 in HSC activation and liver fibrogenesis. The results show that SATB1 expression is reduced during HSC activation. Additionally, SATB1 inhibits HSC activation, proliferation, migration, and collagen synthesis. Furthermore, CTGF, a pro-fibrotic agent, is also significantly inhibited by SATB1 through the Ras/Raf-1/MEK/ERK/Ets-1 pathway. In vivo, SATB1 deactivates HSCs and attenuates fibrosis in TAA-induced fibrotic rat livers. These data indicate that SATB1 plays an important role in HSC activation and liver fibrosis.


Subject(s)
Disease Models, Animal , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Homeodomain Proteins/metabolism , Liver Cirrhosis/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Collagen/biosynthesis , Connective Tissue Growth Factor/antagonists & inhibitors , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Gene Silencing , Hepatic Stellate Cells/pathology , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Liver Cirrhosis/pathology , MAP Kinase Signaling System , Male , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Matrix Attachment Region Binding Proteins/genetics , Random Allocation , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
20.
Int J Med Sci ; 11(12): 1262-9, 2014.
Article in English | MEDLINE | ID: mdl-25317073

ABSTRACT

In a previous study, we found that the global genome organizer Special AT-rich binding protein 1 (SATB1) is highly expressed in mesenchymal-derived human osteosarcoma U2OS cells and that the knock-down of SATB1 results in the inhibition of cell proliferation. The present study was aimed at investigating the effect of silencing SATB1 on cell migration, invasion, apoptosis and resistance to the chemotherapeutic drug arsenic trioxide. Cell migration and invasion were detected by wound-healing assays and trans-well invasion assays, respectively. Cell apoptosis was analyzed by an in situ Cell Death Detection POD Kit, based on terminal deoxynucleotydyl transferase mediated dUTP nick-end labeling (TUNEL) staining and mRNAs were analyzed by real time qRT-PCR. We found that cell migration and invasion were inhibited and that the proportion of apoptotic cells and sensitivities to the chemotherapeutic drug arsenic trioxide were enhanced by knockdown of SATB1 in U2OS cells. Furthermore, mRNA of ABCC1 and ABCG2 were decreased strikingly after SATB1 silencing. It was concluded that the elevated expression of SATB1 in U2OS cells contributes to maintenance of the malignant phenotype and resistance to chemotherapeutic drugs ATO, suggesting that silencing SATB1 in the cells might improve the effects of arsenic trioxides in the treatment of osteosarcoma in which SATB1 is over-expressed and that ABCC1 and ABCG2 were involved in SATB1 mediated resistance of U2OS cells to ATO.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Bone Neoplasms/therapy , Matrix Attachment Region Binding Proteins/antagonists & inhibitors , Osteosarcoma/therapy , Oxides/pharmacology , Apoptosis/genetics , Arsenic Trioxide , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression , Gene Knockdown Techniques , Humans , Matrix Attachment Region Binding Proteins/genetics , Neoplasm Invasiveness/genetics , Osteosarcoma/genetics , Osteosarcoma/pathology , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...