Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
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
2.
Exp Mol Med ; 48: e255, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27561949

ABSTRACT

Cancer stem cells are a subpopulation of cancer cells characterized by self-renewal ability, tumorigenesis and drug resistance. The aim of this study was to investigate the role of HMGA1, a chromatin remodeling factor abundantly expressed in many different cancers, in the regulation of cancer stem cells in ovarian cancer. Spheroid-forming cancer stem cells were isolated from A2780, SKOV3 and PA1 ovarian cancer cells by three-dimensional spheroid culture. Elevated expression of HMGA1 was observed in spheroid cells along with increased expression of stemness-related genes, such as SOX2, KLF4, ALDH, ABCB1 and ABCG2. Furthermore, spheroid A2780 cells, compared with adherent cells, showed higher resistance to chemotherapeutic agents such as paclitaxel and doxorubicin. HMGA1 knockdown in spheroid cells reduced the proliferative advantage and spheroid-forming efficiency of the cells and the expression of stemness-related genes. HMGA1 overexpression in adherent A2780 cells increased cancer stem cell properties, including proliferation, spheroid-forming efficiency and the expression of stemness-related genes. In addition, HMGA1 regulated ABCG2 promoter activity through HMGA1-binding sites. Knockdown of HMGA1 in spheroid cells reduced resistance to chemotherapeutic agents, whereas the overexpression of HMGA1 in adherent ovarian cancer cells increased resistance to chemotherapeutic agents in vitro. Furthermore, HMGA1-overexpressing A2780 cells showed a significant survival advantage after chemotherapeutic agent treatment in a xenograft tumorigenicity assay. Together, our results provide novel insights regarding the critical role of HMGA1 in the regulation of the cancer stem cell characteristics of ovarian cancer cells, thus suggesting that HMGA1 may be an important target in the development of therapeutics for ovarian cancer patients.


Subject(s)
Cell Proliferation , Drug Resistance, Neoplasm , HMGA1a Protein/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , HMGA1a Protein/analysis , HMGA1a Protein/genetics , Humans , Kruppel-Like Factor 4 , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Spheroids, Cellular , Tumor Cells, Cultured
3.
Acta Neurochir (Wien) ; 156(3): 451-61; discussion 461, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24452592

ABSTRACT

BACKGROUND: The postoperative biological behavior of nonfunctioning pituitary adenomas (NFPAs) is variable. Some residual NFPAs are stable long-term, others grow, and some recur despite complete removal. The usual histological markers of tumor aggressiveness are often similar between recurring, regrowing, and stable tumors, and therefore are not reliable as prognostic parameters. In this study, the clinical utility of proliferation indices (labeling index, Li) based on immunohistochemistry targeted at antigens Ki-67 and High-mobility group A1 (HMGA-1) for prediction of NFPA prognosis was investigated. METHODS: Fifty patients with NFPAs were investigated. In each patient, Ki-67 and HMGA-1 Li were evaluated. Based on postoperative magnetic resonance images, patients were classified as tumor-free (18 patients), or harboring a residual tumor (32 patients). The latter group was further subdivided into groups with stable tumor remnants (11 patients) or progressive tumor remnants (21 patients). RESULTS: The median follow-up period was 8 years. No significant relationship between HMGA-1 Li and residual tumor growth was found. Growing residual tumors showed a trend towards higher Ki-67 Li compared with stable ones (p = 0.104). All tumor remnants with Ki-67 Li above 2.2% were growing. The relationship between residual tumor growth and Ki-67 Li exceeding the cutoff value of 2.2% was significant (p = 0.01 in univariate, p = 0.044 in multivariate analysis). CONCLUSIONS: The prognostic significance of the HMGA-1 antigen was not confirmed. In contrast, the Ki-67 Li provides useful and valuable information for the postoperative management of NFPAs. In residual adenomas with a Ki-67 Li above 2.2%, regrowth should be expected, and these tumors may require shorter intervals of follow-up magnetic resonance imaging (MRI) and/or early adjuvant therapy. Future larger studies are needed to confirm the results of this study.


Subject(s)
Adenoma/chemistry , HMGA1a Protein/analysis , Ki-67 Antigen/analysis , Neoplasm Recurrence, Local/chemistry , Neoplasm, Residual/chemistry , Pituitary Neoplasms/chemistry , Adenoma/pathology , Adenoma/physiopathology , Adenoma/surgery , Adult , Aged , Disease-Free Survival , Female , Follow-Up Studies , Humans , Immunohistochemistry , Magnetic Resonance Imaging , Male , Middle Aged , Mitotic Index , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/physiopathology , Neoplasm, Residual/pathology , Neoplasm, Residual/physiopathology , Pituitary Neoplasms/pathology , Pituitary Neoplasms/physiopathology , Pituitary Neoplasms/surgery , Prognosis
4.
Nature ; 480(7376): 254-8, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037311

ABSTRACT

A full description of the human proteome relies on the challenging task of detecting mature and changing forms of protein molecules in the body. Large-scale proteome analysis has routinely involved digesting intact proteins followed by inferred protein identification using mass spectrometry. This 'bottom-up' process affords a high number of identifications (not always unique to a single gene). However, complications arise from incomplete or ambiguous characterization of alternative splice forms, diverse modifications (for example, acetylation and methylation) and endogenous protein cleavages, especially when combinations of these create complex patterns of intact protein isoforms and species. 'Top-down' interrogation of whole proteins can overcome these problems for individual proteins, but has not been achieved on a proteome scale owing to the lack of intact protein fractionation methods that are well integrated with tandem mass spectrometry. Here we show, using a new four-dimensional separation system, identification of 1,043 gene products from human cells that are dispersed into more than 3,000 protein species created by post-translational modification (PTM), RNA splicing and proteolysis. The overall system produced greater than 20-fold increases in both separation power and proteome coverage, enabling the identification of proteins up to 105 kDa and those with up to 11 transmembrane helices. Many previously undetected isoforms of endogenous human proteins were mapped, including changes in multiply modified species in response to accelerated cellular ageing (senescence) induced by DNA damage. Integrated with the latest version of the Swiss-Prot database, the data provide precise correlations to individual genes and proof-of-concept for large-scale interrogation of whole protein molecules. The technology promises to improve the link between proteomics data and complex phenotypes in basic biology and disease research.


Subject(s)
Protein Isoforms/analysis , Protein Isoforms/chemistry , Proteome/analysis , Proteome/chemistry , Proteomics/methods , Alternative Splicing , Cell Line , Cellular Senescence/genetics , DNA Damage , Databases, Protein , HMGA1a Protein/analysis , HMGA1b Protein/analysis , HeLa Cells , Humans , Phenotype , Protein Processing, Post-Translational , Proteolysis , Proteomics/instrumentation
5.
Nucleic Acids Res ; 39(16): 7124-33, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21596776

ABSTRACT

Binding of proteins to DNA is usually considered 1D with one protein bound to one DNA molecule. In principle, proteins with multiple DNA binding domains could also bind to and thereby cross-link different DNA molecules. We have investigated this possibility using high-mobility group A1 (HMGA1) proteins, which are architectural elements of chromatin and are involved in the regulation of multiple DNA-dependent processes. Using direct stochastic optical reconstruction microscopy (dSTORM), we could show that overexpression of HMGA1a-eGFP in Cos-7 cells leads to chromatin aggregation. To investigate if HMGA1a is directly responsible for this chromatin compaction we developed a DNA cross-linking assay. We were able to show for the first time that HMGA1a can cross-link DNA directly. Detailed analysis using point mutated proteins revealed a novel DNA cross-linking domain. Electron microscopy indicates that HMGA1 proteins are able to create DNA loops and supercoils in linearized DNA confirming the cross-linking ability of HMGA1a. This capacity has profound implications for the spatial organization of DNA in the cell nucleus and suggests cross-linking activities for additional nuclear proteins.


Subject(s)
DNA/chemistry , HMGA1a Protein/chemistry , AT-Hook Motifs , Amino Acid Sequence , Amino Acids, Basic/chemistry , Animals , COS Cells , Chlorocebus aethiops , Chromatin/chemistry , DNA/ultrastructure , HMGA1a Protein/analysis , HMGA1a Protein/genetics , Molecular Sequence Data , Mutation
6.
Neurochem Int ; 56(6-7): 736-9, 2010.
Article in English | MEDLINE | ID: mdl-20347905

ABSTRACT

The high-mobility group A protein 1a (HMGA1a) is a well-documented DNA-binding protein acting as an architectural transcription regulator. Recently, HMGA1a protein has been identified as a hypoxia-inducible RNA-binding trans-acting factor for aberrant splicing of presenilin-2 (PS2) pre-mRNA observed in the brains of sporadic Alzheimer's disease. Interestingly, this aberrant splicing of PS2 was also observed in the brains of bipolar disorder and schizophrenia. Many downstream genes under the control of HMGA1a could be associated with schizophrenia. On the other hand, many gene transcripts are aberrantly spliced in schizophrenia. Therefore, we examined the expression at the mRNA and protein levels of this DNA- and RNA-binding factor HMGA1a in the lymphoblastoid cell lines obtained from 16 schizophrenia patients with age-matched controls. We observed markedly higher HMGA1a mRNA and the increased HMGA1a protein in the nuclear fractions of schizophrenia patients. In contrast, there were no significant differences in the expression levels of HMGA1b, which is an alternatively spliced isoform of HMGA1a. The present study is the first to report a significant upregulation of HMGA1a in schizophrenia, suggesting its potential roles in both transcription and splicing of target genes linked with schizophrenia.


Subject(s)
HMGA1a Protein/genetics , Lymphocytes/chemistry , Schizophrenia/genetics , Alternative Splicing/genetics , Cell Line, Transformed , Cell Nucleus/chemistry , Gene Expression , HMGA1a Protein/analysis , HMGA1b Protein/genetics , Humans , Immunoblotting , Lymphocytes/ultrastructure , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
7.
Cancer Invest ; 28(4): 340-9, 2010 May.
Article in English | MEDLINE | ID: mdl-19916743

ABSTRACT

It is well known that HMGA1 group of non-histone chromosomal proteins are up-regulated in several human cancers. We studied the HMGA1 expression in squamous cell carcinoma of skin in mice followed by the treatment with Cisplatin, which is often used in combination therapies of cancers. A short course of Cisplatin treatment led to apoptotic cell death and downregulation (by 40%) of HMGA1. However, extended treatment of Cisplatin caused necrotic cell death; concomitantly HMGA1 expression decreased by 90%. Present results indicate a strong association of HMGA1 with Cisplatin-linked tumor regression. Therefore, HMGA1 could be a potential target in designing therapeutic strategies against cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Cisplatin/pharmacology , HMGA1a Protein/physiology , HMGA1b Protein/physiology , Skin Neoplasms/drug therapy , Animals , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/pathology , Female , HMGA1a Protein/analysis , HMGA1a Protein/genetics , HMGA1b Protein/analysis , HMGA1b Protein/genetics , Mice , Skin Neoplasms/chemistry , Skin Neoplasms/pathology
8.
Proc Natl Acad Sci U S A ; 105(5): 1692-7, 2008 Feb 05.
Article in English | MEDLINE | ID: mdl-18234858

ABSTRACT

In all eukaryotic cells, origins of DNA replication are characterized by the binding of the origin recognition complex (ORC). How ORC is positioned to sites where replication initiates is unknown, because metazoan ORC binds DNA without apparent sequence specificity. Thus, additional factors might be involved in ORC positioning. Our experiments indicate that a family member of the high-mobility group proteins, HMGA1a, can specifically target ORC to DNA. Coimmunoprecipitations and imaging studies demonstrate that HMGA1a interacts with different ORC subunits in vitro and in vivo. This interaction occurs mainly in AT-rich heterochromatic regions to which HMGA1a localizes. Fusion proteins of HMGA1a and the DNA-binding domain of the viral factor EBNA1 or the prokaryotic tetracycline repressor, TetR, can recruit ORC to cognate operator sites forming functional origins of DNA replication. When HMGA1a is targeted to plasmid DNA, the prereplicative complex is assembled during G(1) and the amount of ORC correlates with the local concentration of HMGA1a. Nascent-strand abundance assays demonstrate that DNA replication initiates at or near HMGA1a-rich sites. Our experiments indicate that chromatin proteins can target ORC to DNA, suggesting they might specify origins of DNA replication in metazoan cells.


Subject(s)
DNA Replication , HMGA1a Protein/metabolism , Origin Recognition Complex/metabolism , Replication Origin , Binding Sites , Cell Line , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , HMGA1a Protein/analysis , HMGA1a Protein/genetics , Humans , Immunoprecipitation , Origin Recognition Complex/analysis , Plasmids/chemistry , Plasmids/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
9.
J Periodontal Res ; 43(1): 76-83, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18067550

ABSTRACT

BACKGROUND AND OBJECTIVE: High-mobility-group box 1 functions as a late-phase inflammatory mediator. It can be released extracellularly by macrophages and necrotic cells through lipopolysaccharide and tumor necrosis factor-alpha. The objective of this study was to clarify the source of high-mobility-group box 1 in chronic periodontitis tissues and tumor necrosis factor-alpha-stimulated gingival epithelial cells, and subsequently elucidate its inducible inflammatory pathway. MATERIAL AND METHODS: Chronic periodontitis and healthy gingival sections were stained for high-mobility-group box 1 by immunohistochemistry and immunofluorescence. The amounts of high-mobility-group box 1 released into the gingival crevicular fluid and supernatants from gingival epithelial cells stimulated by tumor necrosis factor-alpha were examined by western blot. The phosphorylation of mitogen-activated protein kinases (MAPKs) in gingival epithelial cells was also examined. RESULTS: High-mobility-group box 1 was detected in the cytoplasm and nucleus of gingival epithelial cells with periodontitis. Western blotting revealed a significant increase in high-mobility-group box 1 expression in the gingival crevicular fluid from periodontitis patients. High-mobility-group box 1 production in gingival epithelial cells was increased following stimulation with tumor necrosis factor-alpha. The molecular dialogue between tumor necrosis factor-alpha and gingival epithelial cells involved modulation of the activities of p38MAPK, Jun N-terminal kinase and p44/42. Interestingly, only phosphorylation of p38MAPK contributed to more than half of the signaling initiated by tumor necrosis factor-alpha-elicited high-mobility-group box 1 release. CONCLUSION: High-mobility-group box 1 is continuously released from the gingival epithelial cells modulated by tumor necrosis factor-alpha. These findings imply that high-mobility-group box 1 expression and possibly p38MAPK constitute important features in periodontitis.


Subject(s)
Epithelial Cells/drug effects , Gingiva/drug effects , HMGA1a Protein/metabolism , Periodontitis/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Case-Control Studies , Cell Survival , Epithelial Cells/metabolism , Female , Gingiva/cytology , Gingival Crevicular Fluid/chemistry , HMGA1a Protein/analysis , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/analysis , Mitogen-Activated Protein Kinase Kinases/metabolism , Rats , p38 Mitogen-Activated Protein Kinases/analysis , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Diagn Mol Pathol ; 16(4): 243-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18043289

ABSTRACT

Malignant ectomesenchymoma (MEM) represents a heterogeneous group of tumors, most likely originating from pluripotent primitive neural crest cells. In this report, we present an 8-month-old infant boy with an MEM on the left scrotum. Retrospective review of the incision biopsy showed the presence of a few ganglion cells in an otherwise classic embryonal rhabdomyosarcoma (RMS), whereas in the resection specimen after chemotherapy the combined RMS and ganglioneuroma components were very obvious. Cytogenetic analysis of the residual lesion showed an abnormal karyotype, 49, XY, +2, -6, +11, +20, +mar, with a hyperploidy in a subset of cells. By fluorescence in situ hybridization analysis, the marker chromosome was identified as originating from chromosome 6, and the tumor cells were negative for PAX3/PAX7 disrupting translocations specific for alveolar RMS. Gains of chromosomes 2, 11, and 20, found in the current case, are a common finding in embryonal RMS. These gains probably reflect the myogenic differentiation of MEM and support the genetic link between these 2 neoplasms. In addition to the conventional cytogenetics, array comparative genomic hybridization analysis was performed on the primary and residual tumors. The genomic profiles of both specimens were basically the same including the presence of 2 distinctive chromosome 6p21.32-p21.2 and 6p11.2 amplification regions in the primary tumor, which vanished in the postchemotherapy specimen. The pretreatment biopsy exhibited strong expression of HMGA1 and HMGA2 proteins in immunohistochemistry, with the shift toward the loss of expression of both genes in the posttreatment tumoral tissue. This finding supports the oncogenic properties of the HMGA family of proteins and their role in the process of malignant transformation.


Subject(s)
Gene Expression Profiling , Genital Neoplasms, Male/diagnosis , Mesenchymoma/diagnosis , Rhabdomyosarcoma, Embryonal/diagnosis , Scrotum , Chromosome Aberrations , Diagnosis, Differential , Genital Neoplasms, Male/pathology , HMGA1a Protein/analysis , HMGA2 Protein/analysis , Humans , Infant , Male , Mesenchymoma/pathology , Oligonucleotide Array Sequence Analysis , PAX3 Transcription Factor , PAX7 Transcription Factor/analysis , Paired Box Transcription Factors/analysis , Rhabdomyosarcoma, Embryonal/pathology , Scrotum/pathology
11.
J Proteome Res ; 6(6): 2304-14, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17455969

ABSTRACT

High-mobility group (HMG) A1 proteins including HMGA1a and HMGA1b are chromosomal proteins that function in a variety of cellular processes such as cell growth, transcription regulation, neoplastic transformation, and progression. Overexpression of HMGA1 proteins has been associated with almost every type of cancer cells. Post-translational modifications (PTMs) of HMGA1 proteins in different types of human cancer cell lines have been extensively explored over the past decade. Here, we extended the identification of PTMs of HMGA1 proteins to human breast tumor tissue specimens with different carcinoma progression stages (metastatic and primary cancer) as well as the paired adjacent normal breast tissues. In this regard, we employed tandem mass spectrometry to examine the nature and sites of PTMs of HMGA1 proteins isolated from cancerous/normal human breast tissues. Novel PTMs of HMGA1a protein, that is, monomethylation at Lys30 and Lys54 as well as monophosphorylation at Ser43 and Ser48, were detected in cancer tissues. In these cancer tissues, we also found C-terminal constitutive phosphorylation in HMGA1a and HMGA1b as well as mono- and dimethylation of Arg25 in HMGA1a, which were previously found to be present in these proteins isolated from human cancer cell lines. Furthermore, a more complex spectrum of PTMs on HMGA1 proteins was correlated with a more aggressive malignancy in human breast cancer tissues.


Subject(s)
Breast Neoplasms/chemistry , HMGA1a Protein/analysis , HMGA1b Protein/analysis , Protein Processing, Post-Translational , Amino Acid Sequence , Breast/chemistry , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , HMGA1a Protein/metabolism , HMGA1b Protein/metabolism , Humans , Lysine/analysis , Lysine/metabolism , Methylation , Molecular Sequence Data , Serine/analysis , Serine/metabolism , Tandem Mass Spectrometry
12.
J Clin Invest ; 117(3): 693-702, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17290307

ABSTRACT

High-mobility group A1 (HMGA1) overexpression and gene rearrangement are frequent events in human cancer, but the molecular basis of HMGA1 oncogenic activity remains unclear. Here we describe a mechanism through which HMGA1 inhibits p53-mediated apoptosis by counteracting the p53 proapoptotic activator homeodomain-interacting protein kinase 2 (HIPK2). We found that HMGA1 overexpression promoted HIPK2 relocalization in the cytoplasm and inhibition of p53 apoptotic function, while HIPK2 overexpression reestablished HIPK2 nuclear localization and sensitivity to apoptosis. HIPK2 depletion by RNA interference suppressed the antiapoptotic effect of HMGA1, which indicates that HIPK2 is the target required for HMGA1 to repress the apoptotic activity of p53. Consistent with this process, a strong correlation among HMGA1 overexpression, HIPK2 cytoplasmic localization, and low spontaneous apoptosis index (comparable to that observed in mutant p53-carrying tumors) was observed in WT p53-expressing human breast carcinomas. Hence, cytoplasmic relocalization of HIPK2 induced by HMGA1 overexpression is a mechanism of inactivation of p53 apoptotic function that we believe to be novel.


Subject(s)
Apoptosis/genetics , Carrier Proteins/metabolism , Gene Expression Regulation , HMGA1a Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Breast Neoplasms/chemistry , Breast Neoplasms/metabolism , Carcinoma/chemistry , Carcinoma/metabolism , Carrier Proteins/analysis , Carrier Proteins/antagonists & inhibitors , Cell Nucleus/chemistry , Cytoplasm/chemistry , Cytoplasm/metabolism , HMGA1a Protein/analysis , Humans , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , RNA Interference , Tumor Cells, Cultured
13.
J Pathol ; 209(2): 206-12, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16521118

ABSTRACT

High mobility group A (HMGA) proteins play an important role in the regulation of transcription, differentiation, and neoplastic transformation. In this work, the expression of HMGA 1 and 2 in 152 lung carcinomas of mainly non-small-cell histological type has been studied by immunohistochemistry in order to evaluate their feasibility as lung cancer markers. In 17 lung cancer cases, the related bronchial epithelial changes were also studied for HMGA1 and 2 expression. RNA expression of HMGA1a and b isoforms and of HMGA2 was determined by real-time semi-quantitative RT-PCR in 23 lung carcinomas. High expression of HMGA1 and HMGA2 at both mRNA and protein levels was detected in lung carcinomas, compared with normal lung tissue. Nuclear immunostaining for HMGA1 and 2 proteins also occurred in hyperplastic, metaplastic, and dysplastic bronchial epithelium. Increased nuclear expression of HMGA1 and 2 correlated with poor survival (for adenocarcinomas, HMGA1, p=0.006; HMGA2, p=0.05). While the expression of HMGA2 was significantly associated with cell proliferation (p=0.008), HMGA1 expression did not show any association with proliferation or apoptotic index. Sequencing of HMGA2 transcripts from tumours with very high expression showed a normal full-length transcript. As HMGA proteins were expressed in about 90% of lung carcinomas and their expression was inversely associated with survival, they may provide useful markers for lung cancer diagnosis and prognosis.


Subject(s)
Carcinoma/chemistry , HMGA Proteins/analysis , Lung Neoplasms/chemistry , Neoplasm Proteins/analysis , Aged , Apoptosis/physiology , Carcinoma/pathology , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/physiology , Female , HMGA1a Protein/analysis , HMGA1b Protein/analysis , HMGA2 Protein/analysis , Humans , Immunohistochemistry/methods , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction/methods , Tissue Array Analysis/methods
14.
Pathol Res Pract ; 201(4): 333-9, 2005.
Article in English | MEDLINE | ID: mdl-15991841

ABSTRACT

We report a case of pleomorphic adenoma (benign mixed tumor) of the breast, which is an extremely rare location for this tumor. Examination of a 55-year-old woman unexpectedly revealed a mass measuring 0.8 cm in diameter in the subareolar region of the right breast. Excisional biopsy was performed, and the tumor histologically showed pleomorphic adenoma composed of duct epithelial cells, myoepithelial cells, and a myxochondroid matrix. Immunohistochemically, duct epithelial cells were positive for the estrogen receptor, but negative for the progesterone receptor. The nuclei of the spindle and myoepithelial cells were immunoreactive for HMGI-C and HMGI(Y) proteins, indicating a histogenesis similar to pleomorphic adenoma of the salivary glands. Interphase fluorescence in situ hybridization performed on paraffin-embedded tissue sections with 12q15 probes and a 6p21 probe demonstrated no chromosomal rearrangement. Sixty-nine cases of this type of tumor arising in the breast have been described previously. Using imaging procedures, the tumor has occasionally been misdiagnosed as malignant clinically and even pathologically in frozen section diagnosis. Careful diagnosis based on paraffin sections is required to avoid unnecessary aggressive surgery, and pathologists should include pleomorphic adenoma in the differential diagnosis of a demarcated, juxtaareolar, small hard mass.


Subject(s)
Adenoma, Pleomorphic/pathology , Breast Neoplasms/pathology , Adenoma, Pleomorphic/chemistry , Adenoma, Pleomorphic/surgery , Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/surgery , Female , HMGA1a Protein/analysis , HMGA2 Protein/analysis , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Middle Aged , Receptors, Estrogen/analysis
15.
J AOAC Int ; 88(2): 577-84, 2005.
Article in English | MEDLINE | ID: mdl-15859086

ABSTRACT

With the development of transgenic crops, regulations to label the genetically modified organisms (GMOs) and their derived products have been issued in many countries. Polymerase chain reaction (PCR) methods are thought to be reliable and useful techniques for qualitative and quantitative detection of GMOs. These methods are generally needed to amplify the transgene and compare the amplified results with that of a corresponding reference gene to get the reliable results. Specific primers were developed for the rapeseed (Brassica napus), high-mobility-group protein I/Y(HMG-I/Y) single-copy gene and PCR cycling conditions suitable for the use of this sequence as an endogenous reference gene in both qualitative and quantitative PCR assays. Both methods were assayed with 15 different rapeseed varieties, and identical amplified products were obtained with all of them. No amplification was observed when templates were the DNA samples from the other species of Brassica genus or other species, such as broccoli, stem mustard, cauliflower, Chinese cabbage, cabbage, sprouts, Arabidopsis thaliana, carrot, tobacco, soybean, mung bean, tomato, pepper, eggplant, plum, wheat, maize, barley, rice, lupine, and sunflower. This system was specific for rapeseed. Limits of detection and quantitation in qualitative and quantitative PCR systems were about 13 pg DNA (about 10 haploid genomes) and about 1.3 pg DNA (about 1 haploid genome), respectively. To further test the feasibility of this HMG-I/Y gene as an endogenous reference gene, samples containing transgenic rapeseed GT73 with the inserted glyphosate oxidoreductase (GOX) gene were quantitated. These demonstrated that the endogenous PCR detection systems were applicable to the qualitative and quantitative detection of transgenic rapeseed.


Subject(s)
Brassica rapa/chemistry , Brassica rapa/genetics , Glycine/analogs & derivatives , HMGA1a Protein/analysis , HMGA1a Protein/genetics , Plants, Genetically Modified/chemistry , Plants, Genetically Modified/genetics , Blotting, Southern , DNA Primers , DNA, Plant/chemistry , DNA, Plant/genetics , Glycine/metabolism , Molecular Sequence Data , Plants/genetics , Reference Standards , Reverse Transcriptase Polymerase Chain Reaction , Glyphosate
16.
Nihon Rinsho ; 61(4): 677-88, 2003 Apr.
Article in Japanese | MEDLINE | ID: mdl-12718095

ABSTRACT

The aberrant splicing isoform(PS2V), generated by exon 5 skipping of the Presenilin-2(PS2) gene transcript, is a diagnostic feature of sporadic Alzheimer's disease(sAD). We found PS2V is hypoxia-inducible in SK-N-SH cells and purified a responsible trans-acting factor based on its binding to an exon 5 fragment. The factor was identified as the High Mobility Group protein A1a: (HMGA1a). HMGA1a bound to a specific sequence on exon5, located upstream of the 5'-splice site. HMGA1a expression was induced by hypoxia. Over-expression of HMGA1a generated PS2V, through interference with U1snRNP binding to the 5'-splice site and caused exon5 skipping. HMGA1a levels were increased in brain from sAD patients. We propose a novel mechanism of sAD that involves HMGA1a-induced aberrant splicing of PS2 pre-mRNA in the absence of any mutations.


Subject(s)
Alzheimer Disease/genetics , HMGA1a Protein/genetics , Membrane Proteins/genetics , Exons/genetics , HMGA1a Protein/analysis , Humans , Presenilin-2 , RNA Splicing
SELECTION OF CITATIONS
SEARCH DETAIL
...