1
|
Wang Q, Liao J, He Z, Su Y, Lin D, Xu L, Xu H, Lin J. LHX6 Affects Erlotinib Resistance and Migration of EGFR-Mutant Non-Small-Cell Lung Cancer HCC827 Cells Through Suppressing Wnt/β-Catenin Signaling. Onco Targets Ther 2020; 13:10983-10994. [PMID: 33149613 PMCID: PMC7605383 DOI: 10.2147/ott.s258896] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 10/08/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND miR-214 has been reported to contribute to erlotinib resistance in non-small-cell lung cancer (NSCLC) through targeting LHX6; however, the molecular mechanisms underlying the involvement of LHX6 in mediating the resistance to EGFR-TKIs in erlotinib-resistant NSCLC HCC827 (HCC827/ER) cells remain unknown. This study aimed to investigate the mechanisms responsible for the contribution of LHX6 to EGFR-TKIs resistance in HCC827/ER cells. MATERIALS AND METHODS HCC827/ER cells were generated by erlotinib treatment at a dose-escalation scheme. LHX6 knockout or overexpression was modeled in HCC827 and HCC827/ER cells, and then erlotinib IC50 values were measured. The cell migration ability was evaluated using a transwell migration assay, and the TCF/LEF luciferase activity was assessed with a TCF/LEF reporter luciferase assay. LHX6, β-catenin and Cyclin D1 expression was quantified using qPCR and Western blotting assays. In addition, the LHX6 expression was detected in lung cancer and peri-cancer specimens using immunohistochemical staining, and the associations of LHX expression with the clinicopathological characteristics of lung cancer were evaluated. RESULTS Lower LHX6 expression was detected in HCC827/ER cells than in HCC827 cells (P < 0.0001), while higher β-catenin expression was seen in HCC827/ER cells than in HCC827 cells (P < 0.001). LHX6 knockout increased erlotinib resistance and cell migration ability in HCC827 cells, and LHX6 overexpression inhibited erlotinib resistance and cell migration ability in HCC827/ER cells. In addition, LHX6 mediated erlotinib resistance and cell migration ability in HCC827/ER cells via the Wnt/β-catenin pathway. Immunohistochemical staining showed lower LHX6 expression in lung cancer specimens relative to peri-cancer specimens, and there were no associations of LHX6 expression with pathologic stage, gender, age or tumor size in lung cancer patients (P > 0.05). CONCLUSION LHX6 down-regulation may induce EGFR-TKIs resistance and increase the migration ability of HCC827/ER cells via activation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Thoracic Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| | - Jinrong Liao
- Department of Radiobiology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| | - Zhiyong He
- Department of Thoracic Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
- Fujian Provincial Key Laboratory of Translation Cancer Medicine, Fuzhou350014, People’s Republic of China
| | - Ying Su
- Department of Radiobiology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| | - Dong Lin
- Department of Thoracic Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| | - Ling Xu
- Department of Thoracic Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| | - Haipeng Xu
- Department of Thoracic Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| | - Jinghui Lin
- Department of Thoracic Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou350014, People’s Republic of China
| |
Collapse
|
2
|
|
3
|
WANG XI, HE CHAO, HU XIAOTONG. LIM homeobox transcription factors, a novel subfamily which plays an important role in cancer (Review). Oncol Rep 2014; 31:1975-85. [DOI: 10.3892/or.2014.3112] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 03/13/2014] [Indexed: 11/06/2022] Open
|
4
|
Nadal N, Chapiro E, Flandrin-Gresta P, Thouvenin S, Vasselon C, Beldjord K, Fenneteau O, Bernard O, Campos L, Nguyen-Khac F. LHX2 deregulation by juxtaposition with the IGH locus in a pediatric case of chronic myeloid leukemia in B-cell lymphoid blast crisis. Leuk Res 2012; 36:e195-8. [PMID: 22727509 DOI: 10.1016/j.leukres.2012.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 05/07/2012] [Accepted: 05/21/2012] [Indexed: 11/30/2022]
MESH Headings
- B-Lymphocytes/pathology
- Blast Crisis/genetics
- Blast Crisis/pathology
- Child
- Chromosomes, Human, Pair 14/genetics
- Chromosomes, Human, Pair 9/genetics
- Gene Expression Regulation, Leukemic
- Genes, Immunoglobulin Heavy Chain/genetics
- Genetic Loci/genetics
- Humans
- LIM-Homeodomain Proteins/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Male
- Transcription Factors/genetics
- Translocation, Genetic/genetics
- Translocation, Genetic/physiology
Collapse
|
5
|
Hung TM, Hu RH, Ho CM, Chiu YL, Lee JL, Jeng YM, Shih DTB, Lee PH. Downregulation of alpha-fetoprotein expression by LHX4: a critical role in hepatocarcinogenesis. Carcinogenesis 2011; 32:1815-23. [PMID: 21965270 DOI: 10.1093/carcin/bgr219] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
LHX4 is a member of the LIM-homeobox family and plays a critical role in pituitary development and differentiation. Several lines of evidences have reported their aberrant expression in cancers. However, the exact roles of LHX4 in carcinogenesis remain unclear. In this study, LHX4 expression was analyzed in tumor and paired non-tumor tissues obtained from patients with hepatocellular carcinoma (HCC) using western blotting and immunohistochemistry. LHX4 was found to be downregulated in tumor tissues and negatively correlated with differentiation grade and alpha-fetoprotein (AFP) levels in 66 HCC patients. To clarify the biological functions of LHX4, transient or stable transfectants overexpressing LHX4 were generated in human hepatoma cells (Huh7 and HepG2). LHX4 overexpression in Huh7 and HepG2 cells induced a more differentiated phenotype by reducing AFP expression. Using in silico analysis, the evolutionary conserved region within the AFP promoter containing LHX4-binding site was identified, implying that AFP is a putative target for LHX4. Moreover, ectopic LHX4 overexpression attenuated Huh7 and HepG2 proliferation. Importantly, the growth-inhibitory effect of LHX4 was reversed by replenishing AFP to the LHX4-overexpressing cells, providing a functional relevance between LHX4 and AFP. Finally, we analyzed expressions of LHX4 and AFP during normal liver development. Hepatic LHX4 expression increased in adult liver in a manner that parallel AFP repression. In conclusion, these data indicate that LHX4 may act as a potential tumor suppressor in hepatocarcinogenesis, suggesting that targeting LHX4 to downregulate AFP might have therapeutic implications.
Collapse
Affiliation(s)
- Tzu-Min Hung
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei 100, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
6
|
The SSX family of cancer-testis antigens as target proteins for tumor therapy. Clin Dev Immunol 2010; 2010:150591. [PMID: 20981248 PMCID: PMC2963798 DOI: 10.1155/2010/150591] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/01/2010] [Indexed: 01/01/2023]
Abstract
Cancer-testis antigens (CTAs) represent an expanding class of tumor-associated proteins defined on the basis of their tissue-restricted expression to testis or ovary germline cells and frequent ectopic expression in tumor tissue. The expression of CTA in MHC class I-deficient germline cells makes these proteins particularly attractive as immunotherapeutic targets because they serve as essentially tumor-specific antigens for MHC class I-restricted CD8+ T cells. Moreover, because CTAs are expressed in many types of cancer, any therapeutic developed to target these antigens might have efficacy for multiple cancer types. Of particular interest among CTAs is the synovial sarcoma X chromosome breakpoint (SSX) family of proteins, which includes ten highly homologous family members. Expression of SSX proteins in tumor tissues has been associated with advanced stages of disease and worse patient prognosis. Additionally, both humoral and cell-mediated immune responses to SSX proteins have been demonstrated in patients with tumors of varying histological origin, which indicates that natural immune responses can be spontaneously generated to these antigens in cancer patients. The current review will describe the history and identification of this family of proteins, as well as what is known of their function, expression in normal and malignant tissues, and immunogenicity.
Collapse
|
7
|
Li M, Lan X, Chen H, Zhang L, Jing Y, Ren G, Wei T, Wang X. The novel missense mutation of goat LHX4 gene. Small Rumin Res 2008. [DOI: 10.1016/j.smallrumres.2008.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Brandal P, Panagopoulos I, Bjerkehagen B, Gorunova L, Skjeldal S, Micci F, Heim S. Detection of a t(1;22)(q23;q12) translocation leading to an EWSR1-PBX1 fusion gene in a myoepithelioma. Genes Chromosomes Cancer 2008; 47:558-64. [PMID: 18383210 DOI: 10.1002/gcc.20559] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chromosome banding as well as molecular cytogenetic methods are of great help in the diagnosis of mesenchymal tumors. Myoepithelial neoplasms of soft tissue including myoepitheliomas, mixed tumors, and parachordomas are diagnoses that have been increasingly recognized the last few years. It is still debated which neoplasms should be included in these morphologically heterogeneous entities, and the boundaries between them are not clear-cut. The pathogenetic mechanisms behind myoepithelial tumors are unknown. Only five parachordomas and one mixed tumor have previously been karyotyped, and nothing is known about their molecular genetic characteristics. We present a mesenchymal tumor classified as a myoepithelioma that had a balanced translocation t(1;22)(q23;q12) as the sole karyotypic change. A novel EWSR1-PBX1 fusion gene consisting of exons 1-8 of the 5'-end of EWSR1 and exons 5-9 of the 3'-end of PBX1 was shown to result from the translocation. Both genes are known to be targeted also by other neoplasia-specific translocations, PBX1 in acute lymphoblastic leukemia and EWSR1 in several solid tumors, most of which are malignant. Based on the structure of the novel fusion gene detected, its transforming mechanism is thought to be the same as for other fusion genes involving EWSR1 or PBX1.
Collapse
Affiliation(s)
- Petter Brandal
- Department of Oncology, Division of Cancer Medicine and Radiotherapy, The Norwegian Radium Hospital, Rikshospitalet University Hospital, Montebello, Oslo, Norway.
| | | | | | | | | | | | | |
Collapse
|
9
|
Liu S, Luo H, Liu J, McNeilly AS, Cui S. Specificity protein 1 (Sp1) plays role in regulating LIM homeodomain transcription factor Lhx4 gene expression. Biochem Biophys Res Commun 2008; 366:36-41. [PMID: 18053794 DOI: 10.1016/j.bbrc.2007.11.082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 11/13/2007] [Indexed: 11/28/2022]
Abstract
Both Sp-family factor Specificity protein 1 (Sp1) and LIM homeodomain transcription factor Lhx4 are involved in regulating the development of pituitary gland and nervous system in mammals. Sp1 gene mutation results in death of mouse embryo around day 11 of gestation, and mouse anterior pituitary development is severely hypoplastic after Lhx4 mutation. While Sp1 interacts with the related Lhx3 gene it is unclear whether Sp1 and Lhx4 also interact to regulate their physiological functions. The present study demonstrates that Lhx4 promoter is TATA-less and GC-rich and these sequences are conserved in different species. We have shown using site-directed mutagenesis and the Dual-Glo Luciferase Assay System that within the -515 to +36bp basic activity regions of hLhx4 promoter the GC boxes were important for Sp1 regulation of the hLhx4 promoter. The electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) experiments confirmed that Sp1 interacted with Lhx4 by directly binding to GC boxes located in Lhx4 promoter. We conclude Sp1 directly regulates Lhx4 gene expression.
Collapse
Affiliation(s)
- Shuqiang Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Yuanmingyuan West Road No. 2, Beijing 100094, People's Republic of China
| | | | | | | | | |
Collapse
|
10
|
Fadlelmola FM, Zhou M, de Leeuw RJ, Dosanjh NS, Harmer K, Huntsman D, Lam WL, Banerjee D. Sub-megabase resolution tiling (SMRT) array-based comparative genomic hybridization profiling reveals novel gains and losses of chromosomal regions in Hodgkin Lymphoma and Anaplastic Large Cell Lymphoma cell lines. Mol Cancer 2008; 7:2. [PMID: 18179710 PMCID: PMC2254646 DOI: 10.1186/1476-4598-7-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2007] [Accepted: 01/07/2008] [Indexed: 01/09/2023] Open
Abstract
Background Hodgkin lymphoma (HL) and Anaplastic Large Cell Lymphoma (ALCL), are forms of malignant lymphoma defined by unique morphologic, immunophenotypic, genotypic, and clinical characteristics, but both overexpress CD30. We used sub-megabase resolution tiling (SMRT) array-based comparative genomic hybridization to screen HL-derived cell lines (KMH2 and L428) and ALCL cell lines (DEL and SR-786) in order to identify disease-associated gene copy number gains and losses. Results Significant copy number gains and losses were observed on several chromosomes in all four cell lines. Assessment of copy number alterations with 26,819 DNA segments identified an average of 20 genetic alterations. Of the recurrent minimally altered regions identified, 11 (55%) were within previously published regions of chromosomal alterations in HL and ALCL cell lines while 9 (45%) were novel alterations not previously reported. HL cell lines L428 and KMH2 shared gains in chromosome cytobands 2q23.1-q24.2, 7q32.2-q36.3, 9p21.3-p13.3, 12q13.13-q14.1, and losses in 13q12.13-q12.3, and 18q21.32-q23. ALCL cell lines SR-786 and DEL, showed gains in cytobands 5p15.32-p14.3, 20p12.3-q13.11, and 20q13.2-q13.32. Both pairs of HL and ALCL cell lines showed losses in 18q21.32-18q23. Conclusion This study is considered to be the first one describing HL and ALCL cell line genomes at sub-megabase resolution. This high-resolution analysis allowed us to propose novel candidate target genes that could potentially contribute to the pathogenesis of HL and ALCL. FISH was used to confirm the amplification of all three isoforms of the trypsin gene (PRSS1/PRSS2/PRSS3) in KMH2 and L428 (HL) and DEL (ALCL) cell lines. These are novel findings that have not been previously reported in the lymphoma literature, and opens up an entirely new area of research that has not been previously associated with lymphoma biology. The findings raise interesting possibilities about the role of signaling pathways triggered by membrane associated serine proteases in HL and aggressive NHL, similar to those described in epithelial tumors.
Collapse
Affiliation(s)
- Faisal M Fadlelmola
- Centre for Translational and Applied Genomics (CTAG), Department of Pathology and Laboratory Medicine, British Columbia Cancer Agency, Vancouver Cancer Centre, Vancouver, BC, V5Z 4E6, Canada.
| | | | | | | | | | | | | | | |
Collapse
|
11
|
de Bruijn DRH, Nap JP, van Kessel AG. The (epi)genetics of human synovial sarcoma. Genes Chromosomes Cancer 2007; 46:107-17. [PMID: 17117414 DOI: 10.1002/gcc.20399] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Human synovial sarcomas are aggressive soft tissue tumors with relatively high rates of recurrences and metastases. They display a variable response to common treatment protocols such as radiation and chemotherapy. For the development of novel diagnostic, prognostic, and therapeutic approaches, detailed information on the molecular mechanisms underlying the development of these tumors is of imperative importance. Fusion of the SS18 and (one of the) SSX genes is a molecular hallmark of human synovial sarcomas. The SS18 and SSX genes encode nuclear proteins that exhibit opposite transcription regulatory activities, likely through epigenetic mechanisms. The SS18 protein functions as a transcriptional coactivator and interacts directly with members of the epigenetic chromatin remodeling and modification machineries. In contrast, the SSX proteins function as transcriptional corepressors and are associated with several Polycomb group proteins. Since the domains involved in these apparently opposite transcription regulatory activities are retained in the SS18-SSX fusion proteins, we hypothesize that these fusion proteins function as "activator-repressors" of transcription. The implications of this model for human synovial sarcoma development and future treatment are discussed.
Collapse
Affiliation(s)
- Diederik R H de Bruijn
- Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen Center for Molecular Life Sciences, Nijmegen, The Netherlands
| | | | | |
Collapse
|
12
|
de Bruijn DRH, van Dijk AHA, Willemse MP, van Kessel AG. The C terminus of the synovial sarcoma-associated SSX proteins interacts with the LIM homeobox protein LHX4. Oncogene 2007; 27:653-62. [PMID: 17667940 DOI: 10.1038/sj.onc.1210688] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As a result of the synovial sarcoma-associated t(X;18) translocation, the SS18 gene on chromosome 18 is fused to either one of the three closely related SSX genes on the X chromosome. The SS18 protein is thought to act as a transcriptional co-activator, whereas the SSX proteins are thought to act as transcriptional corepressors. The main SSX-repression domain is located in its C terminus, a domain that is retained in the respective SS18-SSX fusion proteins. Both the SS18 and SSX proteins lack DNA-binding domains. Previously, we found that the SS18 and SS18-SSX fusion proteins may be tethered to DNA targets via the SS18-interacting protein AF10. Here, we set out to isolate proteins that interact with the SSX C-terminal repression domain using a yeast two-hybrid interaction trap. Of the positive clones isolated, two corresponded to the LIM homeobox protein LHX4, a DNA-binding protein that is involved in transcription regulation. An endogenous interaction was subsequently established in mammalian cells via colocalization and coimmunoprecipitation of the respective proteins. Interestingly, the LHX4 gene was previously found to be deregulated in various human leukemias. In addition, it was previously found that LIM homeobox proteins may bind to and activate the glycoprotein-alpha (CGA) promoter. Using LHX4 chromatin immunoprecipitation and CGA-promoter assays, we found that endogenous LHX4 binds to the CGA promoter and that LHX4-mediated CGA activation is enhanced by the SS18-SSX protein, but not by the SSX protein. Taken together, we conclude that this novel protein - protein interaction may have direct consequences for the (de)regulation of SSX and/or SS18-SSX target genes and, thus, for the development of human synovial sarcomas.
Collapse
Affiliation(s)
- D R H de Bruijn
- Department of Human Genetics, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
13
|
Jariwala U, Prescott J, Jia L, Barski A, Pregizer S, Cogan JP, Arasheben A, Tilley WD, Scher HI, Gerald WL, Buchanan G, Coetzee GA, Frenkel B. Identification of novel androgen receptor target genes in prostate cancer. Mol Cancer 2007; 6:39. [PMID: 17553165 PMCID: PMC1904239 DOI: 10.1186/1476-4598-6-39] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Accepted: 06/06/2007] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The androgen receptor (AR) plays critical roles in both androgen-dependent and castrate-resistant prostate cancer (PCa). However, little is known about AR target genes that mediate the receptor's roles in disease progression. RESULTS Using Chromatin Immunoprecipitation (ChIP) Display, we discovered 19 novel loci occupied by the AR in castrate resistant C4-2B PCa cells. Only four of the 19 AR-occupied regions were within 10-kb 5'-flanking regulatory sequences. Three were located up to 4-kb 3' of the nearest gene, eight were intragenic and four were in gene deserts. Whereas the AR occupied the same loci in C4-2B (castrate resistant) and LNCaP (androgen-dependent) PCa cells, differences between the two cell lines were observed in the response of nearby genes to androgens. Among the genes strongly stimulated by DHT in C4-2B cells--D-dopachrome tautomerase (DDT), Protein kinase C delta (PRKCD), Glutathione S- transferase theta 2 (GSTT2), Transient receptor potential cation channel subfamily V member 3 (TRPV3), and Pyrroline-5-carboxylate reductase 1 (PYCR1)--most were less strongly or hardly stimulated in LNCaP cells. Another AR target gene, ornithine aminotransferase (OAT), was AR-stimulated in a ligand-independent manner, since it was repressed by AR siRNA knockdown, but not stimulated by DHT. We also present evidence for in vivo AR-mediated regulation of several genes identified by ChIP Display. For example, PRKCD and PYCR1, which may contribute to PCa cell growth and survival, are expressed in PCa biopsies from primary tumors before and after ablation and in metastatic lesions in a manner consistent with AR-mediated stimulation. CONCLUSION AR genomic occupancy is similar between LNCaP and C4-2B cells and is not biased towards 5' gene flanking sequences. The AR transcriptionally regulates less than half the genes nearby AR-occupied regions, usually but not always, in a ligand-dependent manner. Most are stimulated and a few are repressed. In general, response is stronger in C4-2B compared to LNCaP cells. Some of the genes near AR-occupied regions appear to be regulated by the AR in vivo as evidenced by their expression levels in prostate cancer tumors of various stages. Several AR target genes discovered in the present study, for example PRKCD and PYCR1, may open avenues in PCa research and aid the development of new approaches for disease management.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Androgens
- Binding Sites
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/genetics
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/metabolism
- Chromosomes, Human/drug effects
- Chromosomes, Human/metabolism
- Dihydrotestosterone/pharmacology
- Extracellular Matrix Proteins/biosynthesis
- Extracellular Matrix Proteins/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Glutathione Transferase/biosynthesis
- Glutathione Transferase/genetics
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Male
- Mucin-6
- Mucins/biosynthesis
- Mucins/genetics
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasms, Hormone-Dependent/genetics
- Neoplasms, Hormone-Dependent/metabolism
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/genetics
- Oligonucleotide Array Sequence Analysis
- Ornithine-Oxo-Acid Transaminase/biosynthesis
- Ornithine-Oxo-Acid Transaminase/genetics
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Protein Kinase C-delta/biosynthesis
- Protein Kinase C-delta/genetics
- Pyrroline Carboxylate Reductases/biosynthesis
- Pyrroline Carboxylate Reductases/genetics
- Receptors, Androgen/genetics
- Receptors, Androgen/physiology
- TRPV Cation Channels/biosynthesis
- TRPV Cation Channels/genetics
- Transcription, Genetic
- delta-1-Pyrroline-5-Carboxylate Reductase
Collapse
Affiliation(s)
- Unnati Jariwala
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Jennifer Prescott
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Li Jia
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Artem Barski
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Steve Pregizer
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Jon P Cogan
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Armin Arasheben
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Wayne D Tilley
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide/Hanson Institute, Adelaide, Australia
| | - Howard I Scher
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Memorial Sloan-Kettering Cancer Center, Department of Medicine, Joan and Sanford I. Weill College of Medicine, New York, NY, USA
| | - William L Gerald
- Genitourinary Oncology Service, Division of Solid Tumor Oncology, Memorial Sloan-Kettering Cancer Center, Department of Medicine, Joan and Sanford I. Weill College of Medicine, New York, NY, USA
| | - Grant Buchanan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Dame Roma Mitchell Cancer Research Laboratories, School of Medicine, The University of Adelaide/Hanson Institute, Adelaide, Australia
| | - Gerhard A Coetzee
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Baruch Frenkel
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Orthopedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, USA
| |
Collapse
|
14
|
Mullen RD, Colvin SC, Hunter CS, Savage JJ, Walvoord EC, Bhangoo AP, Ten S, Weigel J, Pfäffle RW, Rhodes SJ. Roles of the LHX3 and LHX4 LIM-homeodomain factors in pituitary development. Mol Cell Endocrinol 2007; 265-266:190-5. [PMID: 17210222 PMCID: PMC1853274 DOI: 10.1016/j.mce.2006.12.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The LHX3 and LHX4 LIM-homeodomain transcription factors play essential roles in pituitary gland and nervous system development. Mutations in the genes encoding these regulatory proteins are associated with combined hormone deficiency diseases in humans and animal models. Patients with these diseases have complex syndromes involving short stature, and reproductive and metabolic disorders. Analyses of the features of these diseases and the biochemical properties of the LHX3 and LHX4 proteins will facilitate a better understanding of the molecular pathways that regulate the development of the specialized hormone-secreting cells of the mammalian anterior pituitary gland.
Collapse
Affiliation(s)
- Rachel D. Mullen
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Stephanie C. Colvin
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Biology, Indiana University-Purdue University, Indianapolis, IN
| | - Chad S. Hunter
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Biology, Indiana University-Purdue University, Indianapolis, IN
| | - Jesse J. Savage
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Biology, Indiana University-Purdue University, Indianapolis, IN
| | | | | | - Svetlana Ten
- Infants and Children’s Hospital of Brooklyn at Maimonides, Brooklyn, NY
| | | | | | - Simon J. Rhodes
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
15
|
Abstract
The human and mouse genomes each contain at least 12 genes encoding LIM homeodomain (LIM-HD) transcription factors. These gene regulatory proteins feature two LIM domains in their amino termini and a characteristic DNA binding homeodomain. Studies of mouse models and human patients have established that the LIM-HD factors are critical for the development of specialized cells in multiple tissue types, including the nervous system, skeletal muscle, the heart, the kidneys, and endocrine organs such as the pituitary gland and the pancreas. In this article, we review the roles of the LIM-HD proteins in mammalian development and their involvement in human diseases.
Collapse
Affiliation(s)
- Chad S Hunter
- Department of Biology and The Indiana University Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202-5132, USA
| | | |
Collapse
|