Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Blood ; 137(17): 2360-2372, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33150374

ABSTRACT

Clonal evolution drives tumor progression, dissemination, and relapse in multiple myeloma (MM), with most patients dying of relapsed disease. This multistage process requires tumor cells to enter the circulation, extravasate, and colonize distant bone marrow (BM) sites. Here, we developed a fluorescent or DNA-barcode clone-tracking system on MM PrEDiCT (progression through evolution and dissemination of clonal tumor cells) xenograft mouse model to study clonal behavior within the BM microenvironment. We showed that only the few clones that successfully adapt to the BM microenvironment can enter the circulation and colonize distant BM sites. RNA sequencing of primary and distant-site MM tumor cells revealed a progression signature sequentially activated along human MM progression and significantly associated with overall survival when evaluated against patient data sets. A total of 28 genes were then computationally predicted to be master regulators (MRs) of MM progression. HMGA1 and PA2G4 were validated in vivo using CRISPR-Cas9 in the PrEDiCT model and were shown to be significantly depleted in distant BM sites, indicating their role in MM progression and dissemination. Loss of HMGA1 and PA2G4 also compromised the proliferation, migration, and adhesion abilities of MM cells in vitro. Overall, our model successfully recapitulates key characteristics of human MM disease progression and identified potential new therapeutic targets for MM.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic , HMGA1a Protein/metabolism , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , CRISPR-Cas Systems , Cell Adhesion , Cell Movement , Cell Proliferation , Clonal Evolution , Disease Progression , Female , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , Humans , Mice , Mice, SCID , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Prognosis , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Survival Rate , Tumor Cells, Cultured
2.
Sci Rep ; 10(1): 18850, 2020 11 02.
Article in English | MEDLINE | ID: mdl-33139812

ABSTRACT

The mammalian high mobility group protein AT-hook 2 (HMGA2) is a multi-functional DNA-binding protein that plays important roles in tumorigenesis and adipogenesis. Previous results showed that HMGA2 is a potential therapeutic target of anticancer and anti-obesity drugs by inhibiting its DNA-binding activities. Here we report the development of a miniaturized, automated AlphaScreen ultra-high-throughput screening assay to identify inhibitors targeting HMGA2-DNA interactions. After screening the LOPAC1280 compound library, we identified several compounds that strongly inhibit HMGA2-DNA interactions including suramin, a century-old, negatively charged antiparasitic drug. Our results show that the inhibition is likely through suramin binding to the "AT-hook" DNA-binding motifs and therefore preventing HMGA2 from binding to the minor groove of AT-rich DNA sequences. Since HMGA1 proteins also carry multiple "AT-hook" DNA-binding motifs, suramin is expected to inhibit HMGA1-DNA interactions as well. Biochemical and biophysical studies show that charge-charge interactions and hydrogen bonding between the suramin sulfonated groups and Arg/Lys residues play critical roles in the binding of suramin to the "AT-hook" DNA-binding motifs. Furthermore, our results suggest that HMGA2 may be one of suramin's cellular targets.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , HMGA1a Protein/antagonists & inhibitors , HMGA2 Protein/antagonists & inhibitors , Suramin/chemistry , Adipogenesis/drug effects , Amino Acid Motifs/drug effects , Base Sequence/drug effects , Binding Sites/drug effects , Carcinogenesis/drug effects , DNA/drug effects , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HMGA1a Protein/chemistry , HMGA1a Protein/genetics , HMGA2 Protein/chemistry , HMGA2 Protein/genetics , High-Throughput Screening Assays , Humans , Suramin/isolation & purification , Suramin/pharmacology
3.
BMC Urol ; 19(1): 53, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31196036

ABSTRACT

BACKGROUND: Let-7 is one of the earliest discovered microRNAs(miRNAs) and has been reported to be down-regulated in multiple malignant tumors. The effects and molecular mechanisms of let-7i in bladder cancer are still unclear. This study was to investigate the effects and potential mechanisms of let-7i on bladder cancer cells. METHODS: Total RNA was extracted from bladder cancer cell lines. The expression levels of let-7i and HMGA1 were examined by quantitative real-time PCR. Cell viability was detected using the CCK-8 and colony formation assays, while transwell and wound healing assays were used to evaluate migration ability. Luciferase reporter assay and western blot were used to confirm the target gene of let-7i. RESULTS: Compared with the SV-40 immortalized human uroepithelial cell line (SV-HUC-1), bladder cancer cell lines T24 and 5637 had low levels of let-7i expression, but high levels of high mobility group protein A1 (HMGA1) expression. Transfection of cell lines T24 and 5637 with let-7i mimic suppressed cell proliferation and migration. Luciferase reporter assay confirmed HMGA1 may be one of the target genes of let-7i-5p. Protein and mRNA expression of HMGA1 was significantly downregulated in let-7i mimic transfected cell lines T24 and 5637. CONCLUSIONS: Up-regulation of let-7i suppressed proliferation and migration of the human bladder cancer cell lines T24 and 5637 by targeting HMGA1. These findings suggest that let-7i might be considered as a novel therapeutic target for bladder cancer.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , HMGA1a Protein/biosynthesis , MicroRNAs/biosynthesis , Urinary Bladder Neoplasms/metabolism , Cell Line, Transformed , Cell Line, Tumor , HMGA1a Protein/antagonists & inhibitors , Humans , Urinary Bladder Neoplasms/pathology
4.
Eur Rev Med Pharmacol Sci ; 23(9): 3771-3778, 2019 May.
Article in English | MEDLINE | ID: mdl-31115003

ABSTRACT

OBJECTIVE: To investigate the role of micro ribonucleic acid (miR)-195 in acquired resistance to 5-fluorouracil (5-FU) in gastric cancer and its potential mechanism. MATERIALS AND METHODS: The drug resistance of AGS/5-FU and SGC-7901/5-FU cells compared with their parental cells was verified via methyl thiazolyl tetrazolium (MTT) assay, and the expression levels of miR-195 and high-mobility group protein A1 (HMGA1) in AGS/5-FU and SGC-7901/5-FU cells were detected via quantitative Reverse Transcription-Polymerase Chain Reaction (qRT-PCR) and Western blotting. MiR-195 mimic and miR-195 inhibitor were transfected into AGS/5-FU and AGS cells, respectively, the changes in HMGA1 expression were detected via qRT-PCR and Western blotting, and the sensitivity of cells to 5-FU after transfection was detected via MTT assay. After the wild-type and mutant-type luciferase reporter plasmids of HMGA1 were co-transfected with miR-195 mimic or miR-195 NC into cells, the luciferase activity was analyzed using the dual-luciferase reporter system. Finally, the rescue experiment was performed to confirm whether the changes in HMGA1 expression promote the formation of drug resistance in gastric cancer. RESULTS: Both AGS/5-FU and SGC-7901/5-FU cells were significantly resistant to 5-FU compared with their parental cells, and miR-195 was down-regulated in AGS/5-FU and SGC-7901/5-FU cells, while HMGA1 was up-regulated in AGS and SGC-7901 cells. The transfection with miR-195 mimic could suppress the expression level of HMGA1 in AGS/5-FU cells, while the transfection with miR-195 inhibitor could up-regulate the expression level of HMGA1 in AGS cells. Moreover, miR-195 could bind to HMGA1 3'-untranslated region (3'UTR) in a targeted way, thereby inhibiting its expression. It was confirmed via a rescue experiment that the changes in HMGA1 expression promoted the formation of drug resistance in gastric cancer. CONCLUSIONS: The down-regulation of miR-195 induces the resistance to 5-FU in gastric cancer through promoting the expression of HMGA1.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , HMGA1a Protein/metabolism , MicroRNAs/metabolism , Stomach Neoplasms/pathology , 3' Untranslated Regions , Antagomirs/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/metabolism , Stomach Neoplasms/genetics
5.
Cell Biochem Funct ; 35(7): 464-471, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29023799

ABSTRACT

High Mobility Group AT-hook 1 (HMGA1) was identified as a target of miR-214 in human cervical and colorectal cancers (CaCx and CRC) in a previous study. While the expression of miR-214 remains suppressed, HMGA1 behaves as a potent oncogene and plays crucial roles in several aberrant signalling pathways by interacting with intermediates like RELA, CTNNB1, STAT3, and TP53 in CaCx and CRC. Hypothetically, miR-214 should be able to regulate the stabilization of some of these intermediates through the regulation of HMGA1. This was assessed by ectopically expressing miR-214 or complementarily, by inhibiting the expression of HMGA1. In promoter luciferase assays, miR-214 inhibited NF-κB and Wnt activities but elevated TP53 activity in cancer cells. Further, miR-214 suppressed the expression of HMGA1, RELA, CTNNB1, and STAT3 while elevating TP53 levels, similar to when small interfering RNA (siRNA) against HMGA1 was used, as revealed by Western blotting. It is suggested that poor expression of miR-214, commonly reported in CaCx and CRC tissues, may not only result in the sustained expression of HMGA1 but also that of RELA, CTNNB1, and STAT3, and a congruent suppression of TP53 during cancer initiation/progression. These several states are, however, reversed when miR-214 is reintroduced and could explain the tumour suppressive functions observed in earlier studies. Further studies are, however, required to reveal how microRNA-mediated regulation of HMGA1 expression may affect individual signalling pathways in CaCx and CRC. Current results reveal that miR-214 is not only able to regulate the expression of its direct target, HMGA1, but also that of a few signalling intermediates like TP53, RELA, CTNNB1, and STAT3, with which HMGA1 interacts. These intermediates play crucial roles in signalling pathways commonly deregulated in human CaCx and CRC. Hence, it is proposed that miR-214 might act as a tumour suppressor by regulating several aberrant signalling pathways through HMGA1. This knowledge has the potential to help design novel therapeutic strategies in CaCx and CRC.


Subject(s)
MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Tumor Suppressor Protein p53/metabolism , beta Catenin/metabolism , Antagomirs/metabolism , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Down-Regulation , Female , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , HMGA1a Protein/metabolism , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Wnt Signaling Pathway
6.
J Biomol Struct Dyn ; 35(4): 689-703, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26923360

ABSTRACT

High-mobility group A1 (HMGA1) is a non-histone chromosomal protein, which is known as 'architectural' transcription factor that facilitates the assembly of 'enhanceosome.' Because of its elevated expression in a number of human malignancies, with barely minimal levels in healthy adults, HMGA1 is considered as potential 'tumor marker.' Therefore, we looked at the inhibition of hmga1 using anti-gene strategy, as an attractive therapeutic approach. This was achieved by two triplex forming oligonucleotides (TFOs), TFO1 and TFO2 targeted to the promoter of hmga1 at positions, -284--304 and -2800--2826, respectively. The stability of two DNA triplexes was characterized using a variety of biophysical and thermodynamics techniques and was confirmed by gel retardation assay using γ-32P [ATP]. The efficacy of TFOs on HMGA1 expression was evaluated in HeLa cells using MTT assay, Flow cytometry, Western blot, and RT-PCR. Results revealed that DNA Triplex1 formed by TFO1 is more stable and stronger than the corresponding Triplex2. Although both TFOs downregulated hmga1 expression at mRNA and protein levels and caused apoptotic cell death in HeLa cell line, TFO1 demonstrated a greater effect at low concentration which corroborates well with the stability data. Thus, TFO-mediated inhibition of hmga1 expression can be a promising strategy for the development of novel anti-cancer therapeutics.


Subject(s)
Apoptosis/genetics , DNA/genetics , HMGA1a Protein/antagonists & inhibitors , Oligonucleotides/genetics , Promoter Regions, Genetic/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Adult , Base Sequence , Female , HMGA1a Protein/genetics , Humans , Tumor Cells, Cultured
7.
Mol Med Rep ; 15(2): 534-542, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28000891

ABSTRACT

Lung cancer is the most common cause of cancer­associated mortality worldwide, and the number of cases is increasing annually. Several studies have shown that microRNAs (miRNAs) control proliferation, differentiation, and apoptosis in various cell types, and increasing evidence indicates the presence of aberrant miRNA expression profiles and unique miRNA signaling pathways in several types of cancer. The present study aimed to identify miRNAs, which correlated specifically with the progression of lung cancer through the analysis of 57,100 transcripts and 1,341 small RNA expression profiles in 26 lung adenocarcinoma cell lines using next­generation sequencing. The most marked negative correlation was found between the expression of hsa­miR­26a­1 and messenger RNA (mRNA), and a list of mRNAs, which exhibited negative correlation with hsa­miR­26a­1 were investigated. The most marked negative correlation was observed between the expression levels of hsa­miR­26a­1 and high mobility group A1 (HMGA1). Using a lung adenocarcinoma cell line, the present study analyzed the effect of the overexpression of miR­26a on the expression of HMGA1 and found that miR­26a repressed the expression of HMGA1 by reducing the mRNA levels of HMGA1. Furthermore, it was demonstrated that the overexpression of miR­26a in a lung adenocarcinoma cell line repressed cell migration, invasion and growth by targeting HMGA1. Taken together, the present study showed a significant negative correlation between the expression of miR­26a and HMGA1 in 26 lung adenocarcinoma cell lines, and provided evidence that the suppression of miR­26a supports the progression of cancer by stimulating the expression of HMGA1.


Subject(s)
HMGA1a Protein/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions , Base Sequence , Cell Line, Tumor , Cell Movement , Cell Proliferation , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , PTEN Phosphohydrolase/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sequence Alignment , Transcriptome
8.
Oncotarget ; 7(43): 70247-70263, 2016 Oct 25.
Article in English | MEDLINE | ID: mdl-27602961

ABSTRACT

Interleukin (IL)-24, a novel tumor suppressor/cytokine exhibits antitumor activity against a broad-spectrum of human cancer cells. In a recent study, we showed that IL-24 inhibited AKT in lung cancer cells. However, the molecular mechanism of AKT inhibition by IL-24 remains elusive.The high mobility group (HMG) A1 a member of the non-histone chromosomal proteins and commonly referred to as architectural transcription factor, regulates transcription of various genes involved in cell growth and survival. Overexpression of HMGA1 has been shown to be associated with tumor progression and metastasis in several cancers, including human lung cancer. A recent study demonstrated that HMGA1 activates AKT function by reducing the activity of the protein phosphatase, phosphatase 2A subunit B (PPP2R2A) via the oncogenic micro (mi) RNA-222. Based on this report we hypothesized that IL-24-mediated AKT inhibition involved the HMGA1/miR-222 axis.To test our hypothesis, in the present study we used a H1299 lung cancer cell line that expressed exogenous human IL-24 when induced with doxycycline (DOX). Induction of IL-24 expression in the tumor cells markedly reduced HMGA1 mRNA and protein levels. Using a mechanistic approach, we found that IL-24 reduced miR-222-3p and -5p levels, as determined by qRT-PCR. Associated with HMGA1 and miR-222 inhibition was a marked increase in PPP2R2A, with a concomitant decrease in phosphorylated AKTT308/S473 expression. SiRNA-mediated knockdown of HMGA1 in combination with IL-24 significantly reduced AKT T308/S473 protein expression and greatly reduced cell migration and invasion compared with individual treatments. Further combination of IL-24 and a miR-222-3p inhibitor significantly increased PPP2R2A expression.Our results demonstrate for the first time that IL-24 inhibits AKT via regulating the HMGA1/miR-222 signaling node in human lung cancer cells and acts as an effective tumor suppressor. Thus, a therapy combining IL-24 with HMGA1 siRNA or miR-222-3p inhibitor should present effective treatment of lung cancer.


Subject(s)
HMGA1a Protein/antagonists & inhibitors , Interleukins/physiology , Lung Neoplasms/therapy , MicroRNAs/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Cell Line, Tumor , Cell Movement , Doxycycline/pharmacology , HMGA1a Protein/analysis , HMGA1a Protein/physiology , Humans , MicroRNAs/physiology , Neoplasm Invasiveness , Proto-Oncogene Proteins c-akt/physiology , Signal Transduction/physiology
9.
Br J Cancer ; 115(6): 741-51, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27537384

ABSTRACT

BACKGROUND: MicroRNA-214 (miR-214) has been shown to act as a tumour suppressor in human cervical and colorectal cancer cells. The aim of this study was to experimentally validate high mobility group AT-hook 1 as a novel target for miR-214-mediated suppression of growth and motility. METHODS: HMGA1 and miR-214 expression levels were estimated in cervical and colorectal clinical specimens using qPCR. HMGA1 3' untranslated region luciferase assays were performed to validate HMGA1 as a target of miR-214. Effect of altering the expression of miR-214 or HMGA1 on proliferation, migration and invasion of human cervical and colorectal cancer cells was investigated. RESULTS: miR-214 expression was poor while that of HMGA1 was high in cervical and colorectal cancer tissues. miR-214-re-expression or HMGA1 downregulation inhibited proliferation, migration and invasion of cancer cells while miR-214 inhibition had opposite effects. miR-214 was demonstrated to bind to the wild-type 3' untranslated region of HMGA1 but not with its mutant. CONCLUSIONS: Low expression of miR-214 concurrent with elevated levels of HMGA1 may contribute to cervical and colorectal cancer progression. miR-214-mediated regulation of HMGA1 is a novel mechanism for its tumour-suppressive actions in human cervical and colorectal cancer cells and opens up avenues for novel therapeutic strategies for these two cancers.


Subject(s)
Carcinoma/genetics , Colorectal Neoplasms/genetics , HMGA1a Protein/physiology , MicroRNAs/genetics , Neoplasm Proteins/physiology , RNA, Neoplasm/genetics , Uterine Cervical Neoplasms/genetics , 3' Untranslated Regions/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/biosynthesis , HMGA1a Protein/genetics , Humans , Male , Neoplasm Invasiveness , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Uterine Cervical Neoplasms/pathology
10.
Biochemistry (Mosc) ; 79(6): 538-44, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25100012

ABSTRACT

Emerging data have shown that microRNAs (miRNAs) have important functions in the processes of cardiac hypertrophy and heart failure that occur during the postnatal period. Cardiac overexpression of miR-195 results in pathological cardiac growth and heart failure in transgenic mice. In the present study, we analyzed the roles of miR-195 in cardiomyocyte hypertrophy and found that miR-195 was greatly upregulated during isoprenaline-induced cardiomyocyte hypertrophy. By using mRNA microarray and molecular approach, we identified a novel putative target of miR-195 called high-mobility group A1 (HMGA1). Total mRNA microarray showed that HMGA1 was downregulated in primary cardiomyocytes that overexpressed miR-195. Using luciferase activity assay, we demonstrated that miR-195 interacts with the 3'-untranslated region of HMGA1 mRNA. Moreover, we showed that miR-195 in primary cardiomyocytes downregulates the expression of HMGA1 at the protein level. Taken together, our data demonstrated that miR-195 can negatively regulate a new target, HMGA1, which is involved in cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , HMGA1a Protein/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , 3' Untranslated Regions , Animals , Base Sequence , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cells, Cultured , Down-Regulation , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , Isoproterenol/toxicity , Mice , Mice, Transgenic , MicroRNAs/chemistry , Myocytes, Cardiac/cytology , RNA Interference , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Sequence Alignment , Up-Regulation
11.
Expert Opin Ther Targets ; 18(5): 541-53, 2014 May.
Article in English | MEDLINE | ID: mdl-24684280

ABSTRACT

INTRODUCTION: Emerging evidence demonstrates that the high mobility group A1 (HMGA1) chromatin remodeling protein is a key molecular switch required by cancer cells for tumor progression and a poorly differentiated, stem-like state. Because the HMGA1 gene and proteins are expressed at high levels in all aggressive tumors studied to date, research is needed to determine how to 'turn off' this master regulatory switch in cancer. AREAS COVERED: In this review, we describe prior studies that underscore the central role of HMGA1 in refractory cancers and we discuss approaches to target HMGA1 in cancer therapy. EXPERT OPINION: Given the widespread overexpression of HMGA1 in diverse, aggressive tumors, further research to develop technology to target HMGA1 holds immense promise as potent anticancer therapy. Previous work in preclinical models indicates that delivery of short hairpin RNA or interfering RNA molecules to 'switch off' HMGA1 expression dramatically impairs cancer cell growth and tumor progression. The advent of nanoparticle technology to systemically deliver DNA or RNA molecules to tumors brings this approach even closer to clinical applications, although further efforts are needed to translate these advances into therapies for cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , HMGA1a Protein/antagonists & inhibitors , HMGA1b Protein/antagonists & inhibitors , Molecular Targeted Therapy/methods , Neoplasm Proteins/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Differentiation , Chromatin/ultrastructure , Clinical Trials as Topic , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Progression , Drug Screening Assays, Antitumor , Flavonoids/pharmacology , Flavonoids/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Genetic Therapy , HMGA1a Protein/genetics , HMGA1a Protein/physiology , HMGA1b Protein/genetics , HMGA1b Protein/physiology , Humans , Mice , Nanoparticles/administration & dosage , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms/drug therapy , Oxazines/pharmacology , Oxazines/therapeutic use , Piperidines/pharmacology , Piperidines/therapeutic use , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Stem Cells/cytology , Stem Cells/drug effects
12.
Biochem Biophys Res Commun ; 434(1): 70-4, 2013 Apr 26.
Article in English | MEDLINE | ID: mdl-23545254

ABSTRACT

The high mobility group A1 gene (HMGA1) has been previously implicated in breast carcinogenesis, and is considered an attractive target for therapeutic intervention because its expression is virtually absent in normal adult tissue. Other studies have shown that knockdown of HMGA1 reduces the tumorigenic potential of breast cancer cells in vitro. Therefore, we sought to determine if silencing HMGA1 can affect breast cancer development and metastatic progression in vivo. We silenced HMGA1 expression in the human breast cancer cell line MDA-MB-231 using an RNA interference vector, and observed a significant reduction in anchorage-independent growth and tumorsphere formation, which respectively indicate loss of tumorigenesis and self-renewal ability. Moreover, silencing HMGA1 significantly impaired xenograft growth in immunodeficient mice, and while control cells metastasized extensively to the lungs and lymph nodes, HMGA1-silenced cells generated only a few small metastases. Thus, our results show that interfering with HMGA1 expression reduces the tumorigenic and metastatic potential of breast cancer cells in vivo, and lend further support to investigations into targeting HMGA1 as a potential treatment for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/therapy , Gene Knockdown Techniques , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Spheroids, Cellular/pathology , Transplantation, Heterologous/pathology
13.
Histol Histopathol ; 27(5): 567-79, 2012 05.
Article in English | MEDLINE | ID: mdl-22419021

ABSTRACT

The High Mobility Group A1 (HMGA1, formerly HMG-I/Y) gene is highly expressed during embryogenesis and in virtually all aggressive human cancers studied to date, although its role in these settings is only beginning to emerge. Moreover, high levels of expression portend a poor prognosis in some tumors. Increasing evidence suggests that the HMGA1 protein functions as a master regulator with a critical role in normal development and tumor progression in diverse malignancies. These proteins contain AT-hook DNA binding domains that mediate binding to AT-rich regions of chromatin. After binding to DNA, HMGA1 alters DNA structure, and orchestrates the assembly of a transcriptional complex or "enhanceosome" to regulate gene expression. Previous studies indicate that HMGA1 participates in regulating fundamental cellular processes, including transcription, cell cycle progression, embryonic development, neoplastic transformation, differentiation, senescence, viral integration, and DNA repair by virtue of its ability to interact with other proteins, bind to DNA, and modulate gene expression. Recent studies also link HMGA1 expression to poor differentiation status and a refractory, stem cell-like state in aggressive cancers. Together, these findings suggest that HMGA1 could serve as a useful biomarker and therapeutic target in advanced malignancies. Here, we focus on prior studies implicating HMGA1 in the pathogenesis of refractory human tumors arising from diverse tissues and its potential role as a biomarker. We also review previous attempts to target HMGA1 pathways in cancer. Further study of HMGA1 promises to have a major impact on our ability to understand and treat cancer.


Subject(s)
Biomarkers, Tumor/metabolism , HMGA1a Protein/metabolism , Neoplasms/metabolism , Animals , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Female , Gene Expression , Gene Targeting , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/chemistry , HMGA1a Protein/genetics , Humans , Male , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/therapy
14.
Biochemistry ; 50(38): 8107-16, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21854010

ABSTRACT

The molecular mechanism for the displacement of HMGA1 proteins from DNA is integral to disrupting their cellular function, which is linked to many metastatic cancers. Chemical shift and NOESY NMR experiments provide structural evidence for the displacement of an AT hook peptide (DNA binding motif of HMGA1 proteins) by both monomeric and dimeric distamycin. However, the displaced AT hook alters distamycin binding by weakening the distamycin:DNA complex, while slowing monomeric distamycin dissociation when AT hook is in excess. The central role of the AT hook was evaluated by monitoring full-length HMGA1a protein binding using fluorescence anisotropy. HMGA1a was effectively displaced by distamycin, but the cooperative binding exhibited by distamycin was eliminated by displaced HMGA1a. Additionally, these studies indicate that HMGA1a is displaced from the DNA by 1 equiv of distamycin, suggesting the ability to develop therapeutics that take advantage of the positively cooperative nature of HMGA1a binding.


Subject(s)
Distamycins/pharmacology , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/chemistry , AT-Hook Motifs , Amino Acid Sequence , Base Sequence , Binding Sites , Binding, Competitive , DNA/chemistry , DNA/genetics , DNA/metabolism , Dimerization , Distamycins/chemistry , Distamycins/metabolism , Fluorescence Polarization , HMGA1a Protein/metabolism , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Nucleic Acid Conformation , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/metabolism , Protein Structure, Quaternary , Static Electricity
15.
Mol Cancer Res ; 7(11): 1803-12, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19903768

ABSTRACT

Although lung cancer is the leading cause of cancer death worldwide, the precise molecular mechanisms that give rise to lung cancer are incompletely understood. Here, we show that HMGA1 is an important oncogene that drives transformation in undifferentiated, large-cell carcinoma. First, we show that the HMGA1 gene is overexpressed in lung cancer cell lines and primary human lung tumors. Forced overexpression of HMGA1 induces a transformed phenotype with anchorage-independent cell growth in cultured lung cells derived from normal tissue. Conversely, inhibiting HMGA1 expression blocks anchorage-independent cell growth in the H1299 metastatic, undifferentiated, large-cell human lung carcinoma cells. We also show that the matrix metalloproteinase-2 (MMP-2) gene is a downstream target upregulated by HMGA1 in large-cell carcinoma cells. In chromatin immunoprecipitation experiments, HMGA1 binds directly to the MMP-2 promoter in vivo in large-cell lung cancer cells, but not in squamous cell carcinoma cells. In large-cell carcinoma cell lines, there is a significant, positive correlation between HMGA1 and MMP-2 mRNA. Moreover, interfering with MMP-2 expression blocks anchorage-independent cell growth in H1299 large-cell carcinoma cells, indicating that the HMGA1-MMP-2 pathway is required for this transformation phenotype in these cells. Blocking MMP-2 expression also inhibits migration and invasion in the H1299 large-cell carcinoma cells. Our findings suggest an important role for MMP-2 in transformation mediated by HMGA1 in large-cell, undifferentiated lung carcinoma and support the development of strategies to target this pathway in selected tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Transformation, Neoplastic/metabolism , HMGA1a Protein/metabolism , Lung Neoplasms/metabolism , Matrix Metalloproteinase 2/biosynthesis , Carcinoma, Large Cell/enzymology , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/metabolism , Carcinoma, Large Cell/pathology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Gene Expression , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/biosynthesis , HMGA1a Protein/genetics , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection , Up-Regulation
16.
Clin Cancer Res ; 14(5): 1470-7, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18316571

ABSTRACT

PURPOSE: HMGA1 proteins are architectural transcription factors that are overexpressed by pancreatic adenocarcinomas. We previously have shown that RNA interference targeting the HMGA1 gene may represent a potential chemosensitizing strategy in pancreatic adenocarcinoma cells. In this study, we tested the hypothesis that HMGA1 promotes chemoresistance to gemcitabine in pancreatic cancer cells. EXPERIMENTAL DESIGN AND RESULTS: Stable short hairpin RNA-mediated HMGA1 silencing in BxPC3 and MiaPaCa2 cells promoted chemosensitivity to gemcitabine, with reductions in gemcitabine IC(50) and increases in gemcitabine-induced apoptosis and caspase-3 activation. In contrast, forced HMGA1 overexpression in MiaPaCa2 cells promoted chemoresistance to gemcitabine, with increases in gemcitabine IC(50) and reductions in gemcitabine-induced apoptosis and caspase-3 activation. Dominant negative Akt abrogated HMGA1 overexpression-induced increases in chemoresistance to gemcitabine. Finally, HMGA1 silencing promoted chemosensitivity to gemcitabine in vivo in a nude mouse xenograft model of pancreatic adenocarcinoma. CONCLUSION: Our findings suggest that HMGA1 promotes chemoresistance to gemcitabine through an Akt-dependent mechanism. Targeted therapies directed at HMGA1 represent a potential strategy for ameliorating chemoresistance in pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , HMGA1a Protein/metabolism , Pancreatic Neoplasms/drug therapy , Adenocarcinoma/secondary , Animals , Apoptosis/drug effects , Blotting, Western , Carcinoma, Pancreatic Ductal/secondary , Caspase 3/metabolism , Cells, Cultured , Deoxycytidine/therapeutic use , Enzyme Activation/drug effects , HMGA1a Protein/antagonists & inhibitors , HMGA1a Protein/genetics , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction/drug effects , Tumor Cells, Cultured , Gemcitabine
17.
Cancer Gene Ther ; 11(9): 633-41, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15272314

ABSTRACT

Pancreatic carcinoma is one of the most aggressive tumors, and, being refractory to conventional therapies, is an excellent target for new therapeutic approaches. Based on our previous finding of high HMGA1 expression in pancreatic cancer cells compared to normal pancreatic tissue, we evaluated whether suppression of HMGA1 protein expression could be a treatment option for patients affected by pancreatic cancer. Here we report that HMGA1 proteins are overexpressed in pancreatic carcinoma cell lines, and their downregulation through an adenovirus carrying the HMGA1 gene in an antisense orientation (Ad Yas-GFP) results in the death of three human pancreatic carcinoma cell lines (PANC1, Hs766T and PSN1). Pretreatment of PANC1 and PSN1 cells with Ad Yas-GFP suppressed and reduced, respectively, their ability to form xenograft tumors in nude mice. To further verify the role of HMGA1 in pancreatic tumorigenesis, we used a HMGA1 antisense phosphorothioate oligodeoxynucleotide (ODN); its addition induced a decrease in HMGA1 protein levels and a significant reduction of the proliferation rate of PANC1-, Hs766T- and PSN1-treated cells. Therefore, suppression of HMGA1 protein synthesis by an HMGA1 antisense approach seems to be a feasible treatment strategy in pancreatic carcinomas.


Subject(s)
Apoptosis , Cell Proliferation/drug effects , HMGA1a Protein/antagonists & inhibitors , Models, Animal , Pancreatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Down-Regulation , Green Fluorescent Proteins/metabolism , HMGA1a Protein/genetics , HMGA1a Protein/metabolism , Humans , Male , Mice , Mice, Nude , Oligonucleotides, Antisense/pharmacology , Pancreatic Neoplasms/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...