51
|
In search of second-generation HIV integrase inhibitors: targeting integration beyond strand transfer. Future Med Chem 2009; 1:1259-74. [DOI: 10.4155/fmc.09.86] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Highly active antiretroviral therapy combines antiviral drugs targeting different steps in the HIV replication cycle in order to reduce viral loads in patients to undetectable levels. Since HIV readily develops resistance and can therefore escape the action of existing drugs, novel drugs with novel mechanisms of action must be developed. The integration of the viral genome into the human genome is an essential and critical replication step that is catalyzed by the viral integrase with the help of cellular cofactors. Although HIV-1 integrase has been studied for more than two decades, the first integrase inhibitor, raltegravir, was only recently approved for clinical use. A second compound, elvitegravir, is currently in advanced clinical trials. Both drugs interfere with the strand-transfer reaction of integrase. Due to the complexity and multistep nature of the integration reaction, several other functions of integrase can be exploited for drug discovery. In this review, we will describe these alternative strategies to inhibit integration. They have recently attracted considerable interest for the development of second-generation integrase inhibitors.
Collapse
|
52
|
Induction of the small heat shock protein alphaB-crystallin by genotoxic stress is mediated by p53 and p73. Breast Cancer Res Treat 2009; 122:159-68. [PMID: 19777343 DOI: 10.1007/s10549-009-0542-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Accepted: 09/02/2009] [Indexed: 12/14/2022]
Abstract
The small heat shock protein alphaB-crystallin is a molecular chaperone that is induced by stress and protects cells by inhibiting protein aggregation and apoptosis. To identify novel transcriptional regulators of the alphaB-crystallin gene, we examined the alphaB-crystallin promoter for conserved transcription factor DNA-binding elements and identified a putative response element for the p53 tumor suppressor protein. Ectopic expression of wild-type p53 induced alphaB-crystallin mRNA and protein with delayed kinetics compared to p21. Additionally, the induction of alphaB-crystallin by genotoxic stress was inhibited by siRNAs targeting p53. Although the p53-dependent transactivation of an alphaB-crystallin promoter luciferase reporter required the putative p53RE, chromatin immunoprecipitation failed to detect p53 binding to the alphaB-crystallin promoter. These results suggested an indirect mechanism of transactivation involving p53 family members p63 or p73. DeltaNp73 was dramatically induced by p53 in a TAp73-dependent manner, and silencing p73 suppressed the transcriptional activation of alphaB-crystallin by p53. Moreover, ectopic expression of DeltaNp73alpha (but not other p73 isoforms) increased alphaB-crystallin mRNA levels in the absence of p53. Collectively, our results link the molecular chaperone alphaB-crystallin to the cellular genotoxic stress response via a novel mechanism of transcriptional regulation by p53 and p73.
Collapse
|
53
|
Mediavilla-Varela M, Pacheco FJ, Almaguel F, Perez J, Sahakian E, Daniels TR, Leoh LS, Padilla A, Wall NR, Lilly MB, De Leon M, Casiano CA. Docetaxel-induced prostate cancer cell death involves concomitant activation of caspase and lysosomal pathways and is attenuated by LEDGF/p75. Mol Cancer 2009; 8:68. [PMID: 19715609 PMCID: PMC2741463 DOI: 10.1186/1476-4598-8-68] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 08/28/2009] [Indexed: 11/23/2022] Open
Abstract
Background Hormone-refractory prostate cancer (HRPC) is characterized by poor response to chemotherapy and high mortality, particularly among African American men when compared to other racial/ethnic groups. It is generally accepted that docetaxel, the standard of care for chemotherapy of HRPC, primarily exerts tumor cell death by inducing mitotic catastrophe and caspase-dependent apoptosis following inhibition of microtubule depolymerization. However, there is a gap in our knowledge of mechanistic events underlying docetaxel-induced caspase-independent cell death, and the genes that antagonize this process. This knowledge is important for circumventing HRPC chemoresistance and reducing disparities in prostate cancer mortality. Results We investigated mechanistic events associated with docetaxel-induced death in HRPC cell lines using various approaches that distinguish caspase-dependent from caspase-independent cell death. Docetaxel induced both mitotic catastrophe and caspase-dependent apoptosis at various concentrations. However, caspase activity was not essential for docetaxel-induced cytotoxicity since cell death associated with lysosomal membrane permeabilization still occurred in the presence of caspase inhibitors. Partial inhibition of docetaxel-induced cytotoxicity was observed after inhibition of cathepsin B, but not inhibition of cathepsins D and L, suggesting that docetaxel induces caspase-independent, lysosomal cell death. Simultaneous inhibition of caspases and cathepsin B dramatically reduced docetaxel-induced cell death. Ectopic expression of lens epithelium-derived growth factor p75 (LEDGF/p75), a stress survival autoantigen and transcription co-activator, attenuated docetaxel-induced lysosomal destabilization and cell death. Interestingly, LEDGF/p75 overexpression did not protect cells against DTX-induced mitotic catastrophe, and against apoptosis induced by tumor necrosis factor related apoptosis inducing ligand (TRAIL), suggesting selectivity in its pro-survival activity. Conclusion These results underscore the ability of docetaxel to induce concomitantly caspase-dependent and independent death pathways in prostate cancer cells. The results also point to LEDGF/p75 as a potential contributor to cellular resistance to docetaxel-induced lysosomal destabilization and cell death, and an attractive candidate for molecular targeting in HRPC.
Collapse
Affiliation(s)
- Melanie Mediavilla-Varela
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Raz-Prag D, Zeng Y, Sieving PA, Bush RA. Photoreceptor protection by adeno-associated virus-mediated LEDGF expression in the RCS rat model of retinal degeneration: probing the mechanism. Invest Ophthalmol Vis Sci 2009; 50:3897-906. [PMID: 19324854 PMCID: PMC2744960 DOI: 10.1167/iovs.08-3153] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Lens epithelium-derived growth factor (LEDGF) is upregulated in response to stress and enhances the survival of neurons in the retina and optic nerve, as well as a wide range of other cells, such as fibroblasts and keratinocytes. Photoreceptor protection was investigated in the RCS rat retinal degeneration model after Ledgf delivery with an adeno-associated virus (AAV) and the mechanism of protection explored. METHODS Thirty-six RCS and nine P23H rats had bilateral subretinal injections of AAV-Ledgf in one eye and buffer in the contralateral eye as the control. Retinal function was evaluated 8 weeks later by the electroretinogram and compared with photoreceptor cell layer count. LEDGF mRNA and protein levels and mRNA levels of known stress-related factors were compared in treated and control retinas to explore the mechanism of LEDGF protection. Nine RCS rats were treated with adenovirus-heat shock protein 27 (Ad-HSP27) and examined for protection. RESULTS Significant photoreceptor protection was evident functionally and morphologically in 65% to 100% of the RCS rats treated at early ages of up to 7 weeks. Cell protection was more prominent in the superior retinal hemisphere which has a slower natural degeneration rate in untreated eyes. Although many of the heat shock proteins and other stress-related genes showed significant elevation in the AAV-Ledgf-treated eyes, all increases were approximately twofold or less. Transduction of retinal cells with Ad-HSP27 also resulted in photoreceptor protection. AAV-Ledgf elicited no photoreceptor functional protection in P23H rhodopsin transgenic rat retina. CONCLUSIONS Chronic LEDGF treatment via AAV-Ledgf administration gave successful protection of photoreceptors in the RCS rat retina and retarded cell death by about 2 weeks. Induction of heat shock proteins also gave photoreceptor protection. However, compelling evidence was not found that LEDGF protection was associated with upregulation of heat shock proteins.
Collapse
Affiliation(s)
- Dorit Raz-Prag
- Section for Translational Research in Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders
| | - Yong Zeng
- Section for Translational Research in Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders
| | - Paul A. Sieving
- Section for Translational Research in Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders
- Office of the Director, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Ronald A. Bush
- Section for Translational Research in Retinal and Macular Degeneration, National Institute on Deafness and Other Communication Disorders
| |
Collapse
|
55
|
Thys W, Busschots K, McNeely M, Voet A, Christ F, Debyser Z. LEDGF/p75 and transportin-SR2 are cellular cofactors of HIV integrase and novel targets for antiviral therapy. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/17584310.3.2.171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The HIV replication cycle is an elaborate interplay between the viral machinery and cellular proteins. In this review we propose that protein–protein interactions between cellular proteins and HIV integrase are new targets for future antiviral therapy. We focus on the early steps of HIV replication, namely viral entry, uncoating, reverse transcription, trafficking, nuclear import and integration, and the host cell proteins involved herein. We then discuss the feasibility of developing small-molecule protein–protein interaction inhibitors as antiviral agents. Next, we review the HIV integrase cofactors described in the literature highlighting two validated cofactors, lens epithelium-derived growth factor/p75 and transportin-SR2, which are discussed in detail. Finally, a speculative viewpoint is given on small-molecule protein–protein interaction inhibitors as future HIV inhibitors.
Collapse
Affiliation(s)
- Wannes Thys
- Molecular Medicine, KU Leuven Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| | - Katrien Busschots
- Molecular Medicine, KU Leuven Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| | - Melissa McNeely
- Molecular Medicine, KU Leuven Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| | - Arnout Voet
- Molecular Medicine, KU Leuven Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| | - Frauke Christ
- Molecular Medicine, KU Leuven Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| | - Zeger Debyser
- Molecular Medicine, KU Leuven Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| |
Collapse
|
56
|
Llano M, Morrison J, Poeschla EM. Virological and cellular roles of the transcriptional coactivator LEDGF/p75. Curr Top Microbiol Immunol 2009; 339:125-46. [PMID: 20012527 PMCID: PMC3093762 DOI: 10.1007/978-3-642-02175-6_7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The chromatin-associated cellular proteins LEDGF/p75 and LEDGF/p52 have been implicated in transcriptional regulation, cell survival and autoimmunity. LEDGF/p75 also appears to act as a chromatin-docking factor or receptor for HIV-1 and other lentiviruses and to play a role in leukemogenesis. For both the viral and cellular roles of this protein, a key feature is its ability to act as a molecular adaptor and tether proteins to the chromatin fiber. This chapter reviews the emerging roles of LEDGF/p75 and LEDGF/p52 in diverse cellular processes and disease states.
Collapse
Affiliation(s)
- Manual Llano
- Biological Sciences Department, University of Texas, El Paso TX
| | - James Morrison
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester MN
| | - Eric M. Poeschla
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester MN
- Division of Infectious Diseases, Mayo Clinic College of Medicine, Rochester MN
| |
Collapse
|
57
|
Hare S, Shun MC, Gupta SS, Valkov E, Engelman A, Cherepanov P. A novel co-crystal structure affords the design of gain-of-function lentiviral integrase mutants in the presence of modified PSIP1/LEDGF/p75. PLoS Pathog 2009; 5:e1000259. [PMID: 19132083 PMCID: PMC2606027 DOI: 10.1371/journal.ppat.1000259] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Accepted: 12/08/2008] [Indexed: 01/03/2023] Open
Abstract
Lens epithelium derived growth factor (LEDGF), also known as PC4 and SFRS1 interacting protein 1 (PSIP1) and transcriptional co-activator p75, is the cellular binding partner of lentiviral integrase (IN) proteins. LEDGF accounts for the characteristic propensity of Lentivirus to integrate within active transcription units and is required for efficient viral replication. We now present a crystal structure containing the N-terminal and catalytic core domains (NTD and CCD) of HIV-2 IN in complex with the IN binding domain (IBD) of LEDGF. The structure extends the known IN-LEDGF interface, elucidating primarily charge-charge interactions between the NTD of IN and the IBD. A constellation of acidic residues on the NTD is characteristic of lentiviral INs, and mutations of the positively charged residues on the IBD severely affect interaction with all lentiviral INs tested. We show that the novel NTD-IBD contacts are critical for stimulation of concerted lentiviral DNA integration by LEDGF in vitro and for its function during the early steps of HIV-1 replication. Furthermore, the new structural details enabled us to engineer a mutant of HIV-1 IN that primarily functions only when presented with a complementary LEDGF mutant. These findings provide structural basis for the high affinity lentiviral IN-LEDGF interaction and pave the way for development of LEDGF-based targeting technologies for gene therapy.
Collapse
Affiliation(s)
- Stephen Hare
- Division of Medicine, Imperial College London, St. Mary's Campus, London, United Kingdom
| | - Ming-Chieh Shun
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Saumya Shree Gupta
- Division of Medicine, Imperial College London, St. Mary's Campus, London, United Kingdom
| | - Eugene Valkov
- Division of Medicine, Imperial College London, St. Mary's Campus, London, United Kingdom
| | - Alan Engelman
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Peter Cherepanov
- Division of Medicine, Imperial College London, St. Mary's Campus, London, United Kingdom
| |
Collapse
|
58
|
Brown-Bryan TA, Leoh LS, Ganapathy V, Pacheco FJ, Mediavilla-Varela M, Filippova M, Linkhart TA, Gijsbers R, Debyser Z, Casiano CA. Alternative splicing and caspase-mediated cleavage generate antagonistic variants of the stress oncoprotein LEDGF/p75. Mol Cancer Res 2008; 6:1293-307. [PMID: 18708362 PMCID: PMC2790462 DOI: 10.1158/1541-7786.mcr-08-0125] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
There is increasing evidence that an augmented state of cellular oxidative stress modulates the expression of stress genes implicated in diseases associated with health disparities such as certain cancers and diabetes. Lens epithelium-derived growth factor p75 (LEDGF/p75), also known as DFS70 autoantigen, is emerging as a survival oncoprotein that promotes resistance to oxidative stress-induced cell death and chemotherapy. We previously showed that LEDGF/p75 is targeted by autoantibodies in prostate cancer patients and is overexpressed in prostate tumors, and that its stress survival activity is abrogated during apoptosis. LEDGF/p75 has a COOH-terminally truncated splice variant, p52, whose role in stress survival and apoptosis has not been thoroughly investigated. We observed unbalanced expression of these proteins in a panel of tumor cell lines, with LEDGF/p75 generally expressed at higher levels. During apoptosis, caspase-3 cleaved p52 to generate a p38 fragment that lacked the NH(2)-terminal PWWP domain and failed to transactivate the Hsp27 promoter in reporter assays. However, p38 retained chromatin association properties and repressed the transactivation potential of LEDGF/p75. Overexpression of p52 or its variants with truncated PWWP domains in several tumor cell lines induced apoptosis, an activity that was linked to the presence of an intron-derived COOH-terminal sequence. These results implicate the PWWP domain of p52 in transcription function but not in chromatin association and proapoptotic activities. Consistent with their unbalanced expression in tumor cells, LEDGF/p75 and p52 seem to play antagonistic roles in the cellular stress response and could serve as targets for novel antitumor therapies.
Collapse
Affiliation(s)
- Terry A. Brown-Bryan
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, California
| | - Lai S. Leoh
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, California
| | - Vidya Ganapathy
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, California
| | - Fabio J. Pacheco
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Department of Biological Sciences, Centro Universitário Adventista de São Paulo, São Paulo, Brazil
| | - Melanie Mediavilla-Varela
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, California
| | - Maria Filippova
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Division of Biochemistry, Loma Linda University School of Medicine, Loma Linda, California
| | - Thomas A. Linkhart
- Muskuloskeletal Disease Center, Jerry L. Pettis Memorial Veterans Affairs Medical Center, Loma Linda, California
| | - Rik Gijsbers
- Molecular Medicine, Katholieke Universiteit Leuven, B-3000, Leuven, Belgium
| | - Zeger Debyser
- Molecular Medicine, Katholieke Universiteit Leuven, B-3000, Leuven, Belgium
| | - Carlos A. Casiano
- Center for Health Disparities and Molecular Medicine, Loma Linda University School of Medicine, Loma Linda, California
- Division of Microbiology and Molecular Genetics, Loma Linda University School of Medicine, Loma Linda, California
- Department of Medicine, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
59
|
Hou Y, McGuinness DE, Prongay AJ, Feld B, Ingravallo P, Ogert RA, Lunn CA, Howe JA. Screening for antiviral inhibitors of the HIV integrase-LEDGF/p75 interaction using the AlphaScreen luminescent proximity assay. ACTA ACUST UNITED AC 2008; 13:406-14. [PMID: 18480474 DOI: 10.1177/1087057108317060] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Small-molecule inhibitors of HIV integrase (HIV IN) have emerged as a promising new class of antivirals for the treatment of HIV/AIDS. The compounds currently approved or in clinical development specifically target HIV DNA integration and were identified using strand-transfer assays targeting the HIV IN/viral DNA complex. The authors have developed a second biochemical assay for identification of HIV integrase inhibitors, targeting the interaction between HIV IN and the cellular cofactor LEDGF/p75. They developed a luminescent proximity assay (AlphaScreen) designed to measure the association of the 80-amino-acid integrase binding domain of LEDGF/p75 with the 163-amino-acid catalytic core domain of HIV IN. This assay proved to be quite robust (with a Z' factor of 0.84 in screening libraries arrayed as orthogonal mixtures) and successfully identified several compounds specific for this protein-protein interaction.
Collapse
Affiliation(s)
- Yan Hou
- Department of Virology, Schering-Plough Research Institute, Kenilworth, New Jersey 07033, USA
| | | | | | | | | | | | | | | |
Collapse
|
60
|
Abstract
HIV integrates a DNA copy of its genome into a host cell chromosome in each replication cycle. The essential DNA cleaving and joining chemistry of integration is known, but there is less understanding of the process as it occurs in a cell, where two complex and dynamic macromolecular entities are joined: the viral pre-integration complex and chromatin. Among implicated cellular factors, much recent attention has coalesced around LEDGF/p75, a nuclear protein that may act as a chromatin docking factor or receptor for lentiviral pre-integration complexes. LEDGF/p75 tethers HIV integrase to chromatin, protects it from degradation, and strongly influences the genome-wide pattern of HIV integration. Depleting the protein from cells and/or over-expressing its integrase-binding domain blocks viral replication. Current goals are to establish the underlying mechanisms and to determine whether this knowledge can be exploited for antiviral therapy or for targeting lentiviral vector integration in human gene therapy.
Collapse
Affiliation(s)
- E M Poeschla
- Guggenheim 18, Mayo Clinic College of Medicine, 200 First Street SW, Rochester 55905, USA.
| |
Collapse
|
61
|
Engelman A, Cherepanov P. The lentiviral integrase binding protein LEDGF/p75 and HIV-1 replication. PLoS Pathog 2008; 4:e1000046. [PMID: 18369482 PMCID: PMC2275779 DOI: 10.1371/journal.ppat.1000046] [Citation(s) in RCA: 179] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2007] [Indexed: 01/10/2023] Open
Abstract
Retroviral replication proceeds through a stable proviral DNA intermediate, and numerous host cell factors have been implicated in its formation. In particular, recent results have highlighted an important role for the integrase-interactor lens epithelium-derived growth factor (LEDGF)/p75 in lentiviral integration. Cells engineered to over-express fragments of LEDGF/p75 containing its integrase-binding domain but lacking determinants essential for chromatin association are refractory to HIV-1 infection. Furthermore, both the levels of HIV-1 integration and the genomic distribution of the resultant proviruses are significantly perturbed in cells devoid of endogenous LEDGF/p75 protein. A strong bias towards integration along transcription units is a characteristic feature of lentiviruses. In the absence of LEDGF/p75, HIV-1 in large part loses that preference, displaying concomitant integration surges in the vicinities of CpG islands and gene promoter regions, elements naturally targeted by other types of retroviruses. Together, these findings highlight that LEDGF/p75 is an important albeit not strictly essential cofactor of lentiviral DNA integration, and solidify a role for chromatin-associated LEDGF/p75 as a receptor for lentiviral preintegration complexes. By now one of the best characterized virus-host interactions, the integrase-LEDGF/p75 interface opens a range of opportunities for lentiviral vector targeting for gene therapy applications as well as for the development of novel classes of antiretroviral drugs.
Collapse
Affiliation(s)
- Alan Engelman
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Division of AIDS, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Peter Cherepanov
- Division of Medicine, Imperial College London, St. Mary's Campus, London, United Kingdom
| |
Collapse
|
62
|
Shin JH, Piao CS, Lim CM, Lee JK. LEDGF binding to stress response element increases alphaB-crystallin expression in astrocytes with oxidative stress. Neurosci Lett 2008; 435:131-6. [PMID: 18343576 DOI: 10.1016/j.neulet.2008.02.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Revised: 02/11/2008] [Accepted: 02/12/2008] [Indexed: 01/17/2023]
Abstract
AlphaB-crystallin, known as a vertebrate lens protein, is a member of the small heat shock proteins (sHSP). AlphaB-crystallin is abundantly expressed in the vertebrate lens and striated muscles and it is also expressed constitutively in other tissues including the central nervous system (CNS). In our previous report, we showed alphaB-crystallin induction in activated astrocytes, which are enriched in the penumbra after transient focal cerebral ischemia. We also reported that alphaB-crystallin is significantly induced in astrocytes in the CA3 region of the hippocampus following KA-induced seizure. Here, we report that the expression of alphaB-crystallin is upregulated in H2O2-treated primary astrocyte cultures, which was prepared from newborn male Sprague-Dawley rats and that the proximal 408 bp of the alphaB-crystallin promoter harboring stress response element (STRE) is responsible for this induction. This effect of H2O2 was found to be virtually abolished by introducing mutations into STRE, and these mutations also impaired increased lens epithelial derived growth factor (LEDGF) binding to STRE after H2O2 treatment. Moreover, LEDGF was induced in primary astrocyte cultures after H2O2 treatment and alphaB-crystallin induction was significantly suppressed by transfecting small interfering RNA (siRNA) targeting LEDGF. Together these results indicate that the H2O2-induced upregulations of alphaB-crystallin in astrocytes are mediated by the LEDGF-STRE interaction on alphaB-crystallin promoter.
Collapse
Affiliation(s)
- Joo-Hyun Shin
- Department of Anatomy and Center for Advanced Medical Education (BK21 project), Inha University School of Medicine, Inchon, Republic of Korea
| | | | | | | |
Collapse
|
63
|
Kubo E, Fatma N, Akagi Y, Beier DR, Singh SP, Singh DP. TAT-mediated PRDX6 protein transduction protects against eye lens epithelial cell death and delays lens opacity. Am J Physiol Cell Physiol 2008; 294:C842-55. [PMID: 18184874 DOI: 10.1152/ajpcell.00540.2007] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A diminished level of endogenous antioxidant in cells/tissues is associated with reduced resistance to oxidative stress. Peroxiredoxin 6 (PRDX6), a protective molecule, regulates gene expression/function by controlling reactive oxygen species (ROS) levels. Using PRDX6 protein linked to TAT, the transduction domain from human immunodeficiency virus type 1 TAT protein, we demonstrated that PRDX6 was transduced into lens epithelial cells derived from rat or mouse lenses. The protein was biologically active, negatively regulating apoptosis and delaying progression of cataractogenesis by attenuating deleterious signaling. Lens epithelial cells from cataractous lenses bore elevated levels of ROS and were susceptible to oxidative stress. These cells harbored increased levels of active transforming growth factor (TGF)-beta 1 and of alpha-smooth muscle actin and beta ig-h3, markers for cataractogenesis. Importantly, cataractous lenses showed a 10-fold reduction in PRDX6 expression, whereas TGF-beta1 mRNA and protein levels were elevated. The changes were reversed, and cataractogenesis was delayed when PRDX6 was supplied. Results suggest that delivery of PRDX6 can postpone cataractogenesis, and this should be an effective approach to delaying cataracts and other degenerative diseases that are associated with increased ROS.
Collapse
Affiliation(s)
- Eri Kubo
- Department of Ophthalmology, University of Fukui, Fukui, Japan
| | | | | | | | | | | |
Collapse
|
64
|
Abstract
Recent studies demonstrated a number of links between chromatin structure, gene expression, extracellular signaling and cellular differentiation during lens development. Lens progenitor cells originate from a pool of common progenitor cells, the pre-placodal region (PPR) which is formed from a combination of extracellular signaling between the neural plate, naïve ectoderm and mesendoderm. A specific commitment to the lens program over alternate choices such as the formation of olfactory epithelium or the anterior pituitary is manifested by the formation of a thickened surface ectoderm, the lens placode. Mouse lens progenitor cells are characterized by the expression of a complement of lens lineage-specific transcription factors including Pax6, Six3 and Sox2, controlled by FGF and BMP signaling, followed later by c-Maf, Mab21like1, Prox1 and FoxE3. Proliferation of lens progenitors together with their morphogenetic movements results in the formation of the lens vesicle. This transient structure, comprised of lens precursor cells, is polarized with its anterior cells retaining their epithelial morphology and proliferative capacity, whereas the posterior lens precursor cells initiate terminal differentiation forming the primary lens fibers. Lens differentiation is marked by expression and accumulation of crystallins and other structural proteins. The transcriptional control of crystallin genes is characterized by the reiterative use of transcription factors required for the establishment of lens precursors in combination with more ubiquitously expressed factors (e.g. AP-1, AP-2alpha, CREB and USF) and recruitment of histone acetyltransferases (HATs) CBP and p300, and chromatin remodeling complexes SWI/SNF and ISWI. These studies have poised the study of lens development at the forefront of efforts to understand the connections between development, cell signaling, gene transcription and chromatin remodeling.
Collapse
Affiliation(s)
- Ales Cvekl
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | | |
Collapse
|
65
|
Abstract
Retroviral replication hinges on the formation of the provirus, the integrated product of the linear DNA that is made during reverse transcription. Integration is catalyzed by the viral recombinase integrase, yet a number of studies indicate that other viral or cellular proteins play important cofactor roles during HIV-1 integration. Some of these factors bind directly to integrase, whereas others gain access to the integration machinery by binding to the DNA or other viral proteins. This article reviews recent advances on the roles of cellular proteins in HIV-1 integration. As a number of studies have highlighted a particularly important role for the integrase interactor lens epithelium-derived growth factor (LEDGF), much of the focus will be on its mechanism of action and the potential to develop inhibitors of this crucial virus–host interaction.
Collapse
Affiliation(s)
- Alan Engelman
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute and Division of AIDS, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
66
|
Bartholomeeusen K, De Rijck J, Busschots K, Desender L, Gijsbers R, Emiliani S, Benarous R, Debyser Z, Christ F. Differential interaction of HIV-1 integrase and JPO2 with the C terminus of LEDGF/p75. J Mol Biol 2007; 372:407-21. [PMID: 17669426 DOI: 10.1016/j.jmb.2007.06.090] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 06/27/2007] [Accepted: 06/29/2007] [Indexed: 11/28/2022]
Abstract
The transcriptional co-activator lens epithelium-derived growth factor (LEDGF) has been shown to protect cells against environmental stress. The protein has been implicated in auto-immunity and cancer, and is present in cells as the p52 or p75 splice variant. Recently, LEDGF/p75, but not p52, was identified as the prominent interaction partner of human immunodeficiency virus type 1 (HIV-1) integrase. This interaction of HIV-1 integrase with the C-terminal integrase-binding domain of LEDGF/p75 is crucial for HIV-1 replication. To gain insight into the cell biology of LEDGF/p75, we were interested in identifying cellular binding partners of its C-terminal domain. By yeast-two-hybrid screening with a CEMC7 cDNA-library, we were able to identify JPO2 as a binding partner of the C-terminal part of LEDGF/p75. The specific interaction between JPO2 and LEDGF/p75 was verified by pull-down, AlphaScreen, and co-immunoprecipitation. Competition assays using recombinant proteins show a mutually exclusive binding of either JPO2 or HIV-1 integrase to LEDGF/p75. However, differing mechanisms of binding were suggested by continuing interaction of JPO2 with some LEDGF/p75 mutants (I365A, D366A, F406A) that are totally defective for interaction with HIV-1 integrase. This finding is of significance for the development of specific inhibitors targeting only the interaction between LEDGF/p75 and HIV-1 integrase, without disturbing interaction with other cellular factors. Over-expression of JPO2 resulted in a modest but reproducible inhibition of HIV-1 replication, consistent with competition between integrase and JPO2 for binding to LEDGF/p75. Furthermore, JPO2 over-expression activated transcription from the HIV-1 LTR.
Collapse
|
67
|
Hayouka Z, Rosenbluh J, Levin A, Loya S, Lebendiker M, Veprintsev D, Kotler M, Hizi A, Loyter A, Friedler A. Inhibiting HIV-1 integrase by shifting its oligomerization equilibrium. Proc Natl Acad Sci U S A 2007; 104:8316-21. [PMID: 17488811 PMCID: PMC1895947 DOI: 10.1073/pnas.0700781104] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Indexed: 11/18/2022] Open
Abstract
Proteins are involved in various equilibria that play a major role in their activity or regulation. The design of molecules that shift such equilibria is of great therapeutic potential. This fact was demonstrated in the cases of allosteric inhibitors, which shift the equilibrium between active and inactive (R and T) states, and chemical chaperones, which shift folding equilibrium of proteins. Here, we expand these concepts and propose the shifting of oligomerization equilibrium of proteins as a general methodology for drug design. We present a strategy for inhibiting proteins by "shiftides": ligands that specifically bind to an inactive oligomeric state of a disease-related protein and modulate its activity by shifting the oligomerization equilibrium of the protein toward it. We demonstrate the feasibility of our approach for the inhibition of the HIV-1 integrase (IN) protein by using peptides derived from its cellular-binding protein, LEDGF/p75, which specifically inhibit IN activity by a noncompetitive mechanism. The peptides inhibit the DNA-binding of IN by shifting the IN oligomerization equilibrium from the active dimer toward the inactive tetramer, which is unable to catalyze the first integration step of 3' end processing. The LEDGF/p75-derived peptides inhibit the enzymatic activity of IN in vitro and consequently block HIV-1 replication in cells because of the lack of integration. These peptides are promising anti-HIV lead compounds that modulate oligomerization of IN via a previously uncharacterized mechanism, which bears advantages over the conventional interface dimerization inhibitors.
Collapse
Affiliation(s)
| | | | - Aviad Levin
- Department of Biological Chemistry
- Department of Pathology, Hebrew University–Hadassah Medical School, Jerusalem 91120, Israel
| | - Shoshana Loya
- Department of Cell and Developmental Biology, The Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; and
| | - Mario Lebendiker
- Protein Purification Unit, Hebrew University of Jerusalem, Givat Ram, Jerusalem 91904, Israel
| | - Dmitry Veprintsev
- Centre for Protein Engineering, Medical Research Council Centre, Hills Road, Cambridge CB2 2QH, United Kingdom
| | - Moshe Kotler
- Department of Pathology, Hebrew University–Hadassah Medical School, Jerusalem 91120, Israel
| | - Amnon Hizi
- Department of Cell and Developmental Biology, The Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; and
| | | | | |
Collapse
|
68
|
Casiano CA, Mediavilla-Varela M, Tan EM. Tumor-associated antigen arrays for the serological diagnosis of cancer. Mol Cell Proteomics 2006; 5:1745-59. [PMID: 16733262 PMCID: PMC2790463 DOI: 10.1074/mcp.r600010-mcp200] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The recognition that human tumors stimulate the production of autoantibodies against autologous cellular proteins called tumor-associated antigens (TAAs) has opened the door to the possibility that autoantibodies could be exploited as serological tools for the early diagnosis and management of cancer. Cancer-associated autoantibodies are often driven by intracellular proteins that are mutated, modified, or aberrantly expressed in tumor cells and hence are regarded as immunological reporters that could help uncover molecular events underlying tumorigenesis. Emerging evidence suggests that each type of cancer might trigger unique autoantibody signatures that reflect the nature of the malignant process in the affected organ. The advent of novel genomic, proteomic, and high throughput approaches has accelerated interest in the serum autoantibody repertoire in human cancers for the discovery of candidate TAAs. The use of individual anti-TAA autoantibodies as diagnostic or prognostic tools has been tempered by their low frequency and heterogeneity in most human cancers. However, TAA arrays comprising several antigens significantly increase this frequency and hold great promise for the early detection of cancer, monitoring cancer progression, guiding individualized therapeutic interventions, and identification of novel therapeutic targets. Our recent studies suggest that the implementation of TAA arrays in screening programs for the diagnosis of prostate cancer and other cancers should be preceded by the optimization of their sensitivity and specificity through the careful selection of the most favorable combinations of TAAs.
Collapse
Affiliation(s)
- Carlos A Casiano
- Center for Health Disparities Research and Department of Biochemistry and Microbiology, Loma Linda University School of Medicine, California 92350, USA.
| | | | | |
Collapse
|
69
|
Chin MS, Caruso RC, Detrick B, Hooks JJ. Autoantibodies to p75/LEDGF, a cell survival factor, found in patients with atypical retinal degeneration. J Autoimmun 2006; 27:17-27. [PMID: 16757148 DOI: 10.1016/j.jaut.2006.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 03/13/2006] [Accepted: 04/24/2006] [Indexed: 11/21/2022]
Abstract
We have identified a group of patients with an atypical retinal degeneration having anti-retinal autoantibodies in their sera. This select population is characterized by a progressively severe loss of vision associated with a decrease in photoreceptor function, abnormal pigmentation of the retinal pigment epithelium and a negative family history of retinal dystrophy. Immunohistochemical analysis on mouse retinal tissues with sera from this group of patients demonstrated high titers of anti-retinal antibodies (320 to 1,280). Anti-retinal reactivity at these levels was not detected in sera from normal individuals, or from patients with uveitis or known genetic retinal degenerations. One antigen that was identified from a retina cDNA library with sera from a patient with atypical retinal degeneration was lens epithelium-derived growth factor (LEDGF). Western blot analysis revealed that sera from all three patients demonstrated reactivity to p75/LEDGF, a survival factor that protects cells from oxidative, thermal and UV damage. In conclusion, we have found a novel group of patients with a retinal degeneration of non-paraneoplastic, non-familial origin demonstrating immunoreactivity to an autoantigen, p75/LEDGF, heretofore not associated with this disease. Finally, identification of specific anti-retinal antibodies may have applications in the diagnosis and management of retinal degeneration.
Collapse
Affiliation(s)
- Marian S Chin
- Immunology and Virology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 10N248, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
70
|
Sugiura K, Muro Y, Nishizawa Y, Okamoto M, Shinohara T, Tomita Y, Usukura J. LEDGF/DFS70, a major autoantigen of atopic dermatitis, is a component of keratohyalin granules. J Invest Dermatol 2006; 127:75-80. [PMID: 16858421 DOI: 10.1038/sj.jid.5700487] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lens epithelium-derived growth factor/dense fine speckles 70 kDa protein (LEDGF/DFS70) is a transcriptional cofactor, a transcriptional activator, survival factor, and HIV-1 transporter. It is also a major autoantigen in patients with atopic dermatitis (AD), because autoantibodies to this protein are found in approximately 30% of AD patients. To better understand the role of autoantibodies and autoantigens in the pathogenesis of AD, we examined the distribution of LEDGF/DFS70 in the epidermis of normal human skin by light and electron microscopic immunocytochemistry. Increased amounts of LEDGF/DFS70 were located in the nuclei of cells in the basal layer, whereas the cytoplasm of cells in the granular layer stained for LEDGF/DFS70 by light microscopy. Using immunoelectron microscopy, we observed the accumulation of LEDGF/DFS70 in keratohyalin granules (KGs) in the cytoplasm of cells in the granular layer. In addition, Ig heavy chain-binding protein/glucose-regulated protein, 78-kDa (Bip/GRP78), a stress sensing protein in the endoplasmic reticulum, colocalized with LEDGF/DFS70 in the KGs. These results suggest that LEDGF/DFS70 is predominantly located in the nucleus of the basal epidermal cells and translocates into the cytoplasm during differentiation. Once in the cytoplasm, LEDGF/DFS70 accumulates in the KGs in the granular layer. Finally, LEDGF/DFS70, a "nuclear" autoantigen in AD, may play a functional role in the KGs.
Collapse
Affiliation(s)
- Kazumitsu Sugiura
- Department of Dermatology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
71
|
Maertens GN, Cherepanov P, Engelman A. Transcriptional co-activator p75 binds and tethers the Myc-interacting protein JPO2 to chromatin. J Cell Sci 2006; 119:2563-71. [PMID: 16735438 DOI: 10.1242/jcs.02995] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Transcriptional co-activator p75 is implicated in human cancer, autoimmunity and replication of human immunodeficiency virus type 1 (HIV-1) as a dominant integrase-interacting protein. Although characterized as chromatin associated, the normal biological role(s) of p75 remains fairly unclear. To gain insight into p75 function, we have characterized its cellular binding partners and report that JPO2, a recently identified Myc-binding protein, associates with p75 in vitro and in vivo. The pseudo HEAT repeat analogous topology (PHAT) domain of p75, which mediates its interaction with integrase, also mediates the interaction with JPO2, and recombinant integrase protein competes with JPO2 protein for binding to p75 in vitro. JPO2 binds p75 through a 61-residue (amino acids 58-119) region that is distinct from its Myc-interacting domain. In cells, JPO2 and p75 co-localize throughout the cell cycle, and both proteins concentrate on condensed chromosomes during mitosis. Strikingly, the association of JPO2 with chromatin strictly depends upon p75, similar to that of ectopically expressed integrase. Also similar to its effect on integrase, p75 stabilizes intracellular steady-state levels of JPO2 protein. Our results suggest a role for p75 in the Myc regulatory network, and indicate that p75 is a general adaptor protein tethering divergent factors to chromatin through its versatile integrase-binding domain.
Collapse
Affiliation(s)
- Goedele N Maertens
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
72
|
Turlure F, Maertens G, Rahman S, Cherepanov P, Engelman A. A tripartite DNA-binding element, comprised of the nuclear localization signal and two AT-hook motifs, mediates the association of LEDGF/p75 with chromatin in vivo. Nucleic Acids Res 2006; 34:1653-65. [PMID: 16549878 PMCID: PMC1405818 DOI: 10.1093/nar/gkl052] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Lens epithelium-derived growth factor p75 (LEDGF/p75) is a DNA-binding, transcriptional co-activator that participates in HIV-1 integration site targeting. Using complementary approaches, we determined the mechanisms of LEDGF/p75 DNA-binding in vitro and chromatin-association in living cells. The binding of highly-purified, recombinant protein was assayed by surface plasmon resonance (SPR) and electrophoretic mobility gel shift. Neither assay revealed evidence for sequence-specific DNA-binding. Residues 146-197 spanning the nuclear localization signal (NLS) and two AT-hook motifs mediated non-specific DNA-binding, and DNA-binding deficient mutants retained the ability to efficiently stimulate HIV-1 integrase activity in vitro. Chromatin-association was assessed by visualizing the localization of EGFP fusion proteins in interphase and mitotic cells. Although a conserved N-terminal PWWP domain was not required for binding to condensed mitotic chromosomes, its deletion subtly affected the nucleoplasmic distribution of the protein during interphase. A dual AT-hook mutant associated normally with chromatin, yet when the mutations were combined with NLS changes or deletion of the PWWP domain, chromatin-binding function was lost. As the PWWP domain did not readily bind free DNA in vitro, our results indicate that chromatin-association is primarily affected through DNA-binding, with the PWWP domain likely contributing a protein interaction to the overall affinity of LEDGF/p75 for human chromatin.
Collapse
Affiliation(s)
| | - Goedele Maertens
- Department of Pathology, Harvard Medical SchoolBoston, MA 02115, USA
| | | | - Peter Cherepanov
- Department of Pathology, Harvard Medical SchoolBoston, MA 02115, USA
| | - Alan Engelman
- Department of Pathology, Harvard Medical SchoolBoston, MA 02115, USA
- To whom correspondence should be addressed. Tel: +1 617 632 4361; Fax: +1 617 632 3113;
| |
Collapse
|
73
|
Takamura Y, Fatma N, Kubo E, Singh DP. Regulation of heavy subunit chain of gamma-glutamylcysteine synthetase by tumor necrosis factor-alpha in lens epithelial cells: role of LEDGF/p75. Am J Physiol Cell Physiol 2006; 290:C554-66. [PMID: 16403949 DOI: 10.1152/ajpcell.00398.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
TNF-alpha induces oxidative stress by generating reactive oxygen species (ROS). This molecule elevates the expression of gamma-glutamylcysteine synthetase heavy subunit (gamma-GCS-HS). Lens epithelium-derived growth factor (LEDGF)/p75, a transcriptional protein, is inducible by oxidative stress and protects cells from various stresses by upregulating stress-responsive genes. This paper presents evidence that TNF-alpha elevates the expression of LEDGF and that LEDGF is one of the transactivators of gamma-GCS-HS gene. An analysis of the gamma-GCS-HS promoter sequence (-819 to +518 nt) revealed the presence of putative sites for LEDGF binding. Gel mobility assay confirmed the binding of LEDGF to the heat shock element (nGAAn) and the stress response element (A/TGGGGA/T) present in gamma-GCS-HS promoter. Transactivation experiments showed activation of gamma-GCS-HS promoter in cells overexpressing LEDGF or treated with a sublethal dose of TNF-alpha (20 ng/ml). Downregulation of gamma-GCS-HS promoter activity in cells transfected with LEDGF small interfering RNA validated the finding. Notably, cells treated with TNF-alpha (20 ng/ml) for 24 h had an increased abundance of LEDGF and gamma-GCS-HS mRNA and protein. In contrast, cells treated with TNF-alpha for longer periods or with higher concentrations of TNF-alpha showed reduced expression of LEDGF and gamma-GCS-HS and increased cellular death with higher ROS levels. Cells overexpressing LEDGF revealed elevated GSH levels (10-15%), a condition that may potentially eliminate the insult to cells induced by TNF-alpha. Thus TNF-alpha regulation of LEDGF may be physiologically important, as elevated expression of LEDGF increases the expression of endogenous gamma-GCS-HS gene, the catalytic subunit of the regulating enzyme in GSH biosynthesis that may constitute a protective mechanism in limiting oxidative stress induced by inflammatory cytokines.
Collapse
Affiliation(s)
- Yoshihiro Takamura
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198-5840, USA
| | | | | | | |
Collapse
|
74
|
Vandekerckhove L, Christ F, Van Maele B, De Rijck J, Gijsbers R, Van den Haute C, Witvrouw M, Debyser Z. Transient and stable knockdown of the integrase cofactor LEDGF/p75 reveals its role in the replication cycle of human immunodeficiency virus. J Virol 2006; 80:1886-96. [PMID: 16439544 PMCID: PMC1367129 DOI: 10.1128/jvi.80.4.1886-1896.2006] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
After identifying the interaction between the transcriptional coactivator lens epithelium-derived growth factor (LEDGF/p75) and the human immunodeficiency virus type 1 (HIV-1) integrase (IN), we have now investigated the role of LEDGF/p75 during HIV replication. Transient small interfering RNA-mediated knockdown of LEDGF/p75 in HeLaP4 cells resulted in a three- to fivefold inhibition of HIV-1 (strain NL4.3) replication. Quantitative PCR was used to pinpoint the replication block to the integration step. Next, polyclonal and monoclonal HeLaP4-derived cell lines were selected with a stable knockdown of LEDGF/p75 mediated by a lentiviral vector (lentivector) encoding a short hairpin RNA (shRNA) targeting this protein. Cell lines stably transduced with a lentivector encoding an unrelated hairpin or a double-mismatch hairpin served as controls. Again, a two- to fourfold reduction of HIV-1 replication was observed. The extent of LEDGF/p75 knockdown closely correlated with the reduction of HIV-1 replication. After the back-complementation of LEDGF/p75 in the poly- and monoclonal knockdown cell lines using an shRNA-resistant expression plasmid, viral replication was restored to nearly wild-type levels. The Q168A mutation in integrase has been shown to interfere with the interaction with LEDGF/p75 without reducing the enzymatic activity. Transduction by HIV-1-derived lentivectors carrying the Q168A IN mutant was severely hampered, pointing again to a requirement for LEDGF/p75. Altogether, our data validate LEDGF/p75 as an important cellular cofactor for HIV integration and as a potential target for antiviral drug development.
Collapse
Affiliation(s)
- Linos Vandekerckhove
- Molecular Medicine, K.U.Leuven and IRC KULAK, Kapucijnenvoer 33, B-3000 Leuven, Flanders, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Reigstad LJ, Martinez A, Varhaug JE, Lillehaug JR. Nuclear localisation of endogenous SUMO-1-modified PDGF-C in human thyroid tissue and cell lines. Exp Cell Res 2006; 312:782-95. [PMID: 16443219 DOI: 10.1016/j.yexcr.2005.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 11/21/2005] [Accepted: 11/30/2005] [Indexed: 01/06/2023]
Abstract
We investigated post-translational modification and subcellular localisation of endogenous platelet-derived growth factor-C (PDGF-C) in human thyroid papillary carcinomas (PTC), non-neoplastic thyroid tissues, and a selection of cultured cell lines. PDGF-C expressed nuclear localisation in 95% of all tested cell types in culture and in 10% of the thyrocytes from both PTC and non-neoplastic tissue. The cell lines expressed two forms of full-length PDGF-C, approximately 39 and approximately 55 kDa, in cell membrane and cytosol, while the approximately 55 kDa form dominated in the nucleus where it was partly chromatin-associated. The approximately 55 kDa form was post-translationally modified by SUMO-1. The putative PDGF-C SUMOylation site is the surface exposed (314)lysine part of a positively charged loop ((312)RPKTGVRGLHK(322)) with characteristics of a nuclear localisation signal. The tissue thyrocytes expressed a non-SUMOylated approximately 43 kDa and the 55 kDa PDGF-C. The SUMO-1 modified approximately 55 kDa PDGF-C expression was low in PTC where the approximately 43 kDa PDGF-C dominated. This is in contrast to non-neoplastic tissue and cultured cells where the SUMOylated approximately 55 kDa PDGF-C was strongly expressed. Our data provide novel evidence for nuclear localisation of PDGF-C, post-translational modification by SUMOylation and the expression of a novel form of PDGF-C in human papillary thyroid carcinomas.
Collapse
Affiliation(s)
- Laila J Reigstad
- Department of Molecular Biology, University of Bergen, Section of Surgery, Haukeland University Hospital, Bergen 5020, Norway
| | | | | | | |
Collapse
|
76
|
Van Maele B, Busschots K, Vandekerckhove L, Christ F, Debyser Z. Cellular co-factors of HIV-1 integration. Trends Biochem Sci 2006; 31:98-105. [PMID: 16403635 DOI: 10.1016/j.tibs.2005.12.002] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2005] [Revised: 11/23/2005] [Accepted: 12/19/2005] [Indexed: 11/15/2022]
Abstract
To achieve productive infection, the reverse transcribed cDNA of human immunodeficiency virus type 1 (HIV-1) is inserted in the host cell genome. The main protein responsible for this reaction is the viral integrase. However, studies indicate that the virus is assisted by cellular proteins, or co-factors, to achieve integration into the infected cell. The barrier-to-autointegration factor (BAF) might prevent autointegration. Its ability to bridge DNA and the finding that the nuclear lamina-associated polypeptide-2alpha interacts with BAF suggest a role in nuclear structure organization. Integrase interactor 1 was found to directly interact with HIV-1 integrase and to activate its DNA-joining activity, and the high mobility group chromosomal protein A1 might approximate both long terminal repeat (LTR) ends and facilitate integrase binding by unwinding the LTR termini. Furthermore, the lens-epithelium-derived growth factor (LEDGF; also known as p75) seems to tether HIV-1 integrase to the chromosomes. Although a direct role in integration has only been demonstrated for LEDGF/p75, to date, each validated cellular co-factor for HIV-1 integration could constitute a promising new target for antiviral therapy.
Collapse
Affiliation(s)
- Bénédicte Van Maele
- Molecular Virology and Gene Therapy, Molecular Medicine, KULAK and K.U. Leuven, Kapucijnenvoer 33 3000 Leuven, Flanders, Belgium
| | | | | | | | | |
Collapse
|
77
|
Singh DP, Kubo E, Takamura Y, Shinohara T, Kumar A, Chylack LT, Fatma N. DNA Binding Domains and Nuclear Localization Signal of LEDGF: Contribution of two Helix-Turn-Helix (HTH)-like Domains and a Stretch of 58 Amino Acids of the N-terminal to the Trans-activation Potential of LEDGF. J Mol Biol 2006; 355:379-94. [PMID: 16318853 DOI: 10.1016/j.jmb.2005.10.054] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2005] [Revised: 10/15/2005] [Accepted: 10/19/2005] [Indexed: 11/26/2022]
Abstract
Lens epithelium derived growth factor (LEDGF), a nuclear protein, plays a role in regulating the transcription of stress-associated genes such as heat shock proteins by binding to consensus core DNA sequences nAGGn or nGAAn or their repeats, and in doing so helps to provide cyto-protection. However, additional information is required to identify the specific structural features of LEDGF involved in gene transcription. Here we have investigated the functional domains activating and repressing DNA-binding modules, by using a DNA binding assay and trans-activation experiments performed by analyzing proteins prepared from deletion constructs. The results disclosed the DNA-binding domain of N-terminal LEDGF mapped between amino acid residues 5 and 62, a 58 amino acid residue stretch PWWP domain which binds to stress response elements (STRE; A/TGGGGA/T). C-terminal LEDGF contains activation domains, an extensive loop-region (aa 418-530) with two helix-turn-helix (HTH)-like domains, and binds to a heat shock element (HSE; nGAAn). A trans-activation assay using Hsp27 promoter revealed that both HTH domains contribute in a cooperative manner to the trans-activation potential of LEDGF. Interestingly, removal of N-terminal LEDGF (aa 1-187) significantly enhances the gene activation potential of C-terminal LEDGF (aa 199-530); thus the N-terminal domain (aa 5-62), exhibits auto-transcriptional repression activity. It appears that this domain is involved in stabilizing the LEDGF-DNA binding complex. Collectively, our results demonstrate that LEDGF contains three DNA-binding domains, which regulate gene expression depending on cellular microenvironment and thus modify the physiology of cells to maintain cellular homeostasis.
Collapse
Affiliation(s)
- Dhirendra P Singh
- Department of Ophthalmology, and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | | | | | | | | | | | | |
Collapse
|
78
|
Vandegraaff N, Devroe E, Turlure F, Silver PA, Engelman A. Biochemical and genetic analyses of integrase-interacting proteins lens epithelium-derived growth factor (LEDGF)/p75 and hepatoma-derived growth factor related protein 2 (HRP2) in preintegration complex function and HIV-1 replication. Virology 2005; 346:415-26. [PMID: 16337983 DOI: 10.1016/j.virol.2005.11.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2005] [Revised: 09/28/2005] [Accepted: 11/16/2005] [Indexed: 11/29/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) integrase (IN) functions in cells within the context of high molecular weight preintegration complexes (PICs). Lens epithelium-derived growth factor (LEDGF) transcriptional coactivator/p75 and hepatoma-derived growth factor related protein 2 (HRP2) tightly bind to HIV-1 IN and stimulate its integration activity in vitro. Here, we show that each recombinant host cell factor efficiently reconstitutes the in vitro activity of HIV-1 PICs disrupted for functional integration by pre-treatment with high concentrations of salt. Mutational analysis reveals that both the IN-binding and DNA-binding activities of LEDGF/p75 contribute to functional PIC reconstitution. We also investigate a role(s) for these proteins in HIV-1 infection by using short-interfering RNA. HIV-1 infection was essentially unaffected in HeLa-P4 cells depleted for LEDGF/p75, HRP2, or both proteins. We conclude that cells knocked-out for LEDGF/p75 and/or HRP2 will be useful genetic tools to address the roles of these host cell factors in HIV-1 replication.
Collapse
Affiliation(s)
- Nick Vandegraaff
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
79
|
Fatma N, Kubo E, Sharma P, Beier DR, Singh DP. Impaired homeostasis and phenotypic abnormalities in Prdx6-/-mice lens epithelial cells by reactive oxygen species: increased expression and activation of TGFbeta. Cell Death Differ 2005; 12:734-50. [PMID: 15818411 DOI: 10.1038/sj.cdd.4401597] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
PRDX6, a member of the peroxiredoxins (PRDXs) family, is a key player in the removal of reactive oxygen species (ROS). Using targeted inactivation of the Prdx6 gene, we present evidence that the corresponding protein offsets the deleterious effects of ROS on lens epithelial cells (LECs) and regulates gene expression by limiting its levels. PRDX6-depleted LECs displayed phenotypic alterations and elevated alpha-smooth muscle actin and betaig-h3 expression (markers for cataractogenesis), indistinguishable from transforming growth factor beta (TGFbeta)-induced changes. Biochemical assays disclosed enhanced levels of ROS, as well as high expression and activation of TGFbeta1 in Prdx6-/- LECs. A CAT assay revealed transcriptional repression of lens epithelium-derived growth factor (LEDGF), HSP27, and alphaB-crystallin promoter activities in these cells. A gel mobility shift assay demonstrated the attenuation of LEDGF binding to heat shock or stress response elements present in these genes. A supply of PRDX6 toPrdx6-/- LECs reversed these changes. Based on the above data, we propose a rheostat role for PRDX6 in regulating gene expression by controlling the ROS level to maintain cellular homeostasis.
Collapse
Affiliation(s)
- N Fatma
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | |
Collapse
|
80
|
Okamoto M, Ogawa Y, Watanabe A, Sugiura K, Shimomura Y, Aoki N, Nagasaka T, Tomita Y, Muro Y. Autoantibodies to DFS70/LEDGF are increased in alopecia areata patients. J Autoimmun 2005; 23:257-66. [PMID: 15501396 DOI: 10.1016/j.jaut.2004.07.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2004] [Revised: 06/29/2004] [Accepted: 07/12/2004] [Indexed: 11/19/2022]
Abstract
Alopecia areata (AA) has been suspected to be an autoimmune disease, although there is no distinct evidence, we investigated the relationship between AA and autoantibodies against dense fine speckles 70 kDa (DFS70) in 111 patients with alopecia and 105 healthy controls. The sera from 59 out of 111 (53%) Japanese alopecia patients were positive for anti-nuclear antibody (ANA), as compared to the sera of 16 out of 105 (15%) healthy controls (p < 0.001). Twenty percent (22/111) of the alopecia patients were shown to be positive for the prevalence of anti-DFS70 antibodies, as compared to 8% (8/105) of the healthy controls (p < 0.01). IgG subclass analysis by ELISA showed that IgG1 and IgG2-anti-DFS70 antibodies were dominant in alopecia patients. The DFS70 gene expression in the hair structures was clearly detected in both those with and those without the anti-DFS70 antibody by RT-PCR. Immunohistochemical techniques showed that the DFS70 was localized predominantly in the outer root sheath (ORS) cells. The elevated anti-DFS70 antibodies in alopecia patients and the localization of the DFS70 in the ORS suggest that autoantibodies against the DFS70 are related to the etiology in a certain population of AA.
Collapse
Affiliation(s)
- Miyako Okamoto
- Division of Connective Tissue Disease & Autoimmunity, Department of Dermatology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Kubo E, Singh DP, Akagi Y. Gene expression profiling of diabetic and galactosaemic cataractous rat lens by microarray analysis. Diabetologia 2005; 48:790-8. [PMID: 15761720 DOI: 10.1007/s00125-005-1687-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2004] [Accepted: 11/07/2004] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS Osmotic and oxidative stress is associated with the progression and advancement of diabetic cataract. In the present study, we used a cDNA microarray method to analyse gene expression patterns in streptozotocin-induced diabetic rats and galactose-fed cataractous lenses. In addition, we investigated the regulation and interaction(s) of anti-oxidant protein 2 and lens epithelium-derived growth factor in these models. METHODS To identify differential gene expression patterns, one group of Sprague-Dawley rats was made diabetic with streptozotocin and a second group was made galactosaemic. Total RNA was extracted from the lenses of both groups and their controls. Labelled cDNA was hybridised to Atlas Rat Arrays. Changes in gene expression level were analysed. Real-time PCR and western analysis were used to validate the microarray results. RESULTS The expression of 31 genes was significantly modulated in hyperglycaemic lenses compared with galactosaemic lenses. Notably, transcript and protein levels of B-cell leukaemia/lymphoma protein 2 and nuclear factor-kappaB were significantly elevated in rat lenses at 4 weeks after injection of streptozotocin. At a later stage, mRNA and protein levels of TGF-beta were elevated. However, levels of mRNA for IGF-1, lens epithelium-derived growth factor and anti-oxidant protein 2 were diminished in streptozotocin-induced diabetic cataract. CONCLUSIONS/INTERPRETATIONS These results provide evidence that progression of sugar cataract involves oxidative- and TGF-beta-mediated signalling. These pathways may promote abnormal gene expression in the hyperglycaemic and galactosaemic states and thus may contribute to the symptoms associated with these conditions. Since oxidative stress seems to be a major event in cataract formation, supply of anti-oxidant may postpone the progression of such disorders.
Collapse
Affiliation(s)
- E Kubo
- Department of Ophthalmology, Faculty of Medical Science, University of Fukui, 23-3 Shimoaiduki, Matsuoka, Fukui, 910-1193, Japan
| | | | | |
Collapse
|
82
|
Busschots K, Vercammen J, Emiliani S, Benarous R, Engelborghs Y, Christ F, Debyser Z. The interaction of LEDGF/p75 with integrase is lentivirus-specific and promotes DNA binding. J Biol Chem 2005; 280:17841-7. [PMID: 15749713 DOI: 10.1074/jbc.m411681200] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
We have previously shown that the p75 isoform of the transcriptional co-activator lens epithelium-derived growth factor (LEDGF) interacts tightly with human immunodeficiency virus (HIV)-1 integrase (IN) and is essential for nuclear targeting of this protein in human cells (Cherepanov, P., Maertens, G., Proost, P., Devreese, B., Van Beeumen, J., Engelborghs, Y., De Clercq, E., and Debyser, Z. (2003) J. Biol. Chem. 278, 372-381; Maertens, G., Cherepanov, P., Pluymers, W., Busschots, K., De Clercq, E., Debyser, Z., and Engelborghs, Y. (2003) J. Biol. Chem. 278, 33528-33539). Here the interaction between recombinant LEDGF/p75 and HIV-1 IN was examined in a pull-down binding test. LEDGF/p75 was shown to increase the solubility of HIV-1 IN. Next, fluorescent correlation spectroscopy was used to measure the interaction of LEDGF/p75 or the complex of HIV-1 IN and LEDGF/p75 with a specific double-stranded DNA oligonucleotide. Whereas LEDGF/p75 displayed only a moderate affinity for DNA, it strongly promoted the binding of HIV-1 IN to DNA. This effect was specific for the p75 isoform of LEDGF and was not seen with p52. In the pull-down assay LEDGF/p75 interacted with HIV-1, HIV-2, and feline immunodeficiency virus IN, but not with the IN of human T-cell lymphotropic virus type 2, Moloney murine leukemia virus, or Rous sarcoma virus. These results strongly suggest that the interaction of LEDGF/p75 with IN is specific to lentiviridae. LEDGF/p75 stimulated the binding of HIV-1 and HIV-2 IN, but not Moloney murine leukemia virus or Rous sarcoma virus IN, to an aspecific DNA. These results provide supporting evidence for our hypothesis that LEDGF/p75 plays a role in the tethering of lentiviral IN to the chromosomal DNA.
Collapse
Affiliation(s)
- Katrien Busschots
- Laboratory for Molecular Virology and Gene Therapy, Katholieke Universiteit Leuven and Interdisciplinary Research Center Katholieke Universiteit Leuven Campus Kortrijk, Kapucijnenvoer 33, B-3000 Leuven, Flanders, Belgium
| | | | | | | | | | | | | |
Collapse
|
83
|
Abstract
The intraocular lens has recently been recognized as a potential source for neuroprotective and neurite-promoting activities. The lens is ontogenetically and functionally a peculiar intraocular tissue with the unique feature of performing incomplete cellular apoptosis throughout the lifetime. The ectodermally derived epithelial cells permanently divide to produce the nuclei- and organelle-free lens fibre cells that allow for the optical transparency. The underlying extremely specific physical, biochemical, metabolic and structural mechanism lead to efficient protection from photo-oxidative stress caused by exposure to short-wavelength light. The fact that fibre cells undergo incomplete apoptosis is also of crucial importance to other cellular systems. In particular, injured nerve cells such as axotomized retinal ganglion cells may profit from the apoptosis-blocking mechanisms operating within the lens fibres. In this review we first discuss some factors involved in the lens differentiation and partial apoptosis as a basic principle of long-term survival. We then present recent experimental evidence that lenticular factors also operate outside the lens, and in particular within the retina to contribute to axonal regeneration, e.g. after a trauma. In turn, factors such as GAP-43 that were thought to be exclusively expressed within nervous tissue have now also been discovered within the lenticular tissue. Experiments of the direct confrontation of lenticular epithelial and fibre cells with regenerating ganglion cell axons in vitro are presented. It is concluded that survival factors supplied by the lens might be used to facilitate survival within neuronal tissue.
Collapse
Affiliation(s)
- T Stupp
- Department of Experimental Ophthalmology, School of Medicine, University Eye Hospital of Münster and Interdisciplinary Clinical Research Center (IZKF), Domagkstrasse 15, D-48149 Münster, Germany
| | | |
Collapse
|
84
|
Chylack LT, Fu L, Mancini R, Martin-Rehrmann MD, Saunders AJ, Konopka G, Tian D, Hedley-Whyte ET, Folkerth RD, Goldstein LE. Lens epithelium-derived growth factor (LEDGF/p75) expression in fetal and adult human brain. Exp Eye Res 2004; 79:941-8. [PMID: 15642333 DOI: 10.1016/j.exer.2004.08.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2004] [Accepted: 08/19/2004] [Indexed: 11/17/2022]
Abstract
Lens epithelium-derived growth factor (LEDGF/p75) is a novel transcription co-activator that is critically involved in lens epithelial cell gene regulation and stress responses. Recent evidence indicates that LEDGF/p75 may play an important role in lens epithelial to fibre cell terminal differentiation. Since the lens and the brain are both ectodermally derived organs generated from epithelioid progenitor cells, we hypothesize that LEDGF/p75 is expressed and subserving similar functions in both organs. To investigate this hypothesis, we studied LEDGF/p75 expression and localization in the human brain. We detected LEDGF/p75-specific RT-PCR reaction products in both fetal and adult human brain. LEDGF/p75 mRNA expression in the brain exhibited differential developmental and regional specificity. LEDGF/p75 transcript was markedly elevated in fetal as compared to adult brain. In the adult brain, LEDGF/p75 mRNA expression was substantial in the subventricular zone (SVZ), scant in hippocampus, and undetectable elsewhere. To study LEDGF/p75 protein expression and localization, we developed and purified a new anti-LEDGF/p75 polyclonal antibody directed against a unique C-terminal region of LEDGF/p75. Western blot analysis of fetal and adult human brain revealed a approximately 75 kDa protein that demonstrated developmental and regional specificity similar to that detected by RT-PCR analysis. LEDGF/p75 protein expression was high in fetal brain and in the adult SVZ. Immunohistochemical studies of human fetal brain showed prominent LEDGF/p75-immunoreactive cells in the germinal neuroepithelium and cortical plate regions. Analysis of adult and aged human brain revealed LEDGF/p75-immunoreactive cell enrichment in the SVZ adjacent to the ventral region of the lateral ventricle at the level of the anterior commissure, a region implicated in adult neurogenesis. We utilised a primary mixed cortical cell culture system to identify LEDGF/p75 in neurons, but not astrocytes. Neuronal LEDGF/p75 exhibited a predominantly perinuclear distribution pattern. These data demonstrate that LEDGF/p75 is expressed in discrete regions and cell types within the fetal and adult human brain. Moreover, the developmental and regional expression patterns of LEDGF/p75 suggest that this transcriptional co-activator may be involved in neuroepithelial stem cell differentiation and neurogenesis.
Collapse
Affiliation(s)
- Leo T Chylack
- Molecular Aging and Development Laboratory, Center for Ophthalmic Research, Department of Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Mittermann I, Aichberger KJ, Bünder R, Mothes N, Renz H, Valenta R. Autoimmunity and atopic dermatitis. Curr Opin Allergy Clin Immunol 2004; 4:367-71. [PMID: 15349035 DOI: 10.1097/00130832-200410000-00007] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW It has been demonstrated that a considerable percentage of patients suffering from atopic dermatitis mount IgE autoantibodies against a broad variety of human proteins. This review summarizes evidence for autoimmune mechanisms in atopic dermatitis and suggests novel pathomechanisms that may be involved in this disease. RECENT FINDINGS It has been shown that patients suffering from atopic dermatitis exhibit IgE autoreactivity to human proteins. These autoantigens are expressed in a variety of cell and tissue types. Complementary DNAs coding for IgE autoantigens have been identified, cloned and characterized at the molecular level. Using purified recombinant IgE autoantigens, it has been shown in paradigmatic models that IgE autoimmunity may be a pathogenetic mechanism in atopic dermatitis. Moreover, it has been shown that the levels of IgE autoantibodies are associated with severity of disease. SUMMARY Patients suffering from severe manifestations of atopy mount IgE autoantibodies against a variety of human proteins. The levels of IgE autoantibodies correspond with disease severity. Several mechanisms of IgE autoimmunity may contribute to the pathogenesis of atopic dermatitis.
Collapse
Affiliation(s)
- Irene Mittermann
- Department of Pathophysiology, Division of Immunopathology, Vienna General Hospital, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
86
|
Llano M, Vanegas M, Fregoso O, Saenz D, Chung S, Peretz M, Poeschla EM. LEDGF/p75 determines cellular trafficking of diverse lentiviral but not murine oncoretroviral integrase proteins and is a component of functional lentiviral preintegration complexes. J Virol 2004; 78:9524-37. [PMID: 15308744 PMCID: PMC506940 DOI: 10.1128/jvi.78.17.9524-9537.2004] [Citation(s) in RCA: 242] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1), feline immunodeficiency virus (FIV), and Moloney murine leukemia virus (MoMLV) integrases were stably expressed to determine their intracellular trafficking. Each lentiviral integrase localized to cell nuclei in close association with chromatin while the murine oncoretroviral integrase was cytoplasmic. Fusions of pyruvate kinase to the lentiviral integrases did not reveal transferable nuclear localization signals. The intracellular trafficking of each was determined instead by the transcriptional coactivator LEDGF/p75, which was required for nuclear localization. Stable small interfering RNA expression eliminated detectable LEDGF/p75 expression and caused dramatic, stable redistribution of each lentiviral integrase from nucleus to cytoplasm while the distribution of MoMLV integrase was unaffected. In addition, endogenous LEDGF/p75 coimmunoprecipitated specifically with each lentiviral integrase. In vitro integration assays with preintegration complexes (PICs) showed that endogenous LEDGF/p75 is a component of functional HIV-1 and FIV PICs. However, HIV-1 and FIV infection and replication in LEDGF/p75-deficient cells was equivalent to that in control cells, whether cells were dividing or growth arrested. Two-long terminal repeat circle accumulation in nondividing cell nuclei was also equivalent to that of LEDGF/p75 wild-type cells. Virions produced in LEDGF/p75-deficient cells had normal infectivity. We conclude that LEDGF/p75 fully accounts for cellular trafficking of diverse lentiviral, but not oncoretroviral, integrases and is the main lentiviral integrase-to-chromatin tethering factor. While lentiviral PIC nuclear import is unaffected by LEDGF/p75 knockdown, this protein is a component of functional lentiviral PICs. A role in HIV-1 integration site distribution merits investigation.
Collapse
Affiliation(s)
- Manuel Llano
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
87
|
Cherepanov P, Devroe E, Silver PA, Engelman A. Identification of an evolutionarily conserved domain in human lens epithelium-derived growth factor/transcriptional co-activator p75 (LEDGF/p75) that binds HIV-1 integrase. J Biol Chem 2004; 279:48883-92. [PMID: 15371438 DOI: 10.1074/jbc.m406307200] [Citation(s) in RCA: 229] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Human lens epithelium-derived growth factor/transcriptional co-activator p75 (LEDGF/p75) protein was recently identified as a binding partner for HIV-1 integrase (IN) in human cells. In this work, we used biochemical and bioinformatic approaches to define the domain organization of LEDGF/p75. Using limited proteolysis and deletion mutagenesis we show that the protein contains a pair of evolutionarily conserved domains, assuming about 35% of its sequence. Whereas the N-terminal PWWP domain had been recognized previously, the second domain is novel. It is comprised of approximately 80 amino acid residues and is both necessary and sufficient for binding to HIV-1 IN. Strikingly, the integrase binding domain (IBD) is not unique to LEDGF/p75, as a second human protein, hepatoma-derived growth factor-related protein 2 (HRP2), contains a homologous sequence. LEDGF/p75 and HRP2 IBDs avidly bound HIV-1 IN in an in vitro GST pull-down assay and each full-length protein potently stimulated HIV-1 IN activity in vitro. LEDGF/p75 and HRP2 are predicted to share a similar domain organization and have an evident evolutionary and likely functional relationship.
Collapse
Affiliation(s)
- Peter Cherepanov
- Departments of Cancer Immunology and AIDS and Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
88
|
Fatma N, Kubo E, Chylack LT, Shinohara T, Akagi Y, Singh DP. LEDGF regulation of alcohol and aldehyde dehydrogenases in lens epithelial cells: stimulation of retinoic acid production and protection from ethanol toxicity. Am J Physiol Cell Physiol 2004; 287:C508-16. [PMID: 15238362 DOI: 10.1152/ajpcell.00076.2004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Retinoic acid (RA) is required for the normal growth and maintenance of many cell types, including lens epithelial cells (LECs). Alcohol (ADH) and aldehyde (ALDH) dehydrogenases are implicated in cellular detoxification and conversion of vitamin A to RA. Lens epithelium-derived growth factor (LEDGF) provides cellular protection against stress by transactivating stress-associated genes. Here we show evidence that LEDGF binds and transactivates heat shock (nGAAn) and stress response (A/TGGGGA/T) elements in the promoters of ADH1, ADH4, and retinaldehyde 2 (RALDH2) genes. Electrophoretic mobility and supershift assays disclosed specific binding of LEDGF to nGAAn and A/TGGGGA/T elements in these gene promoters. Transfection experiments in LECs with promoters linked to a chloramphenicol acetyltransferase (CAT) reporter gene along with LEDGF cDNA revealed higher CAT activity. RT-PCR results confirmed that LECs overexpressing LEDGF contained increased levels of ADH1, ADH4, and RALDH2 mRNA. Notably, LECs displayed higher LEDGF mRNA and protein expression during ethanol stress. Cells overexpressing LEDGF typically exhibited elevated RA levels and survived well during ethanol stress. The present findings indicate that LEDGF is one of the transcriptional activators of these genes that facilitates cellular protection against ethanol stress and plays a role in RA production.
Collapse
Affiliation(s)
- Nigar Fatma
- Department of Ophthalmology, 985840 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198-5840, USA
| | | | | | | | | | | |
Collapse
|
89
|
Feng H, Xiang H, Mao YW, Wang J, Liu JP, Huang XQ, Liu Y, Liu SJ, Luo C, Zhang XJ, Liu Y, Li DWC. Human Bcl-2 activates ERK signaling pathway to regulate activating protein-1, lens epithelium-derived growth factor and downstream genes. Oncogene 2004; 23:7310-21. [PMID: 15326476 DOI: 10.1038/sj.onc.1208041] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The proto-oncogene, bcl-2, has various functions besides its role in protecting cells from apoptosis. One of the functions is to regulate expression of other genes. Previous studies have demonstrated that Bcl-2 regulates activities of several important transcription factors including NF-kappaB and p53, and also their downstream genes. In our recent studies, we reported that Bcl-2 substantially downregulates expression of the endogenous alphaB-crystallin gene through modulating the transcriptional activity of lens epithelium-derived growth factor (LEDGF). In the present communication, we report that human Bcl-2 can positively regulate expression of the proto-oncogenes c-jun and c-fos. Moreover, it enhances the DNA binding activity and transactivity of the activating protein-1 (AP-1). Furthermore, we present evidence to show that Bcl-2 can also activate both ERK1 and ERK2 MAP kinases. Inhibition of the activities of these kinases or the upstream activating kinases by pharmacological inhibitors or dominant-negative mutants abolishes the Bcl-2-mediated regulation of AP-1, LEDGF and their downstream genes. Together, our results demonstrate that through activation of the ERK kinase signaling pathway, Bcl-2 regulates the transcriptional activities of multiple transcription factors, and hence modulates the expression of their downstream genes. Thus, our results provide a mechanism to explain how Bcl-2 may regulate expression of other genes.
Collapse
Affiliation(s)
- Hao Feng
- College of Life Sciences, Hunan Normal University, Changsha, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Maertens G, Cherepanov P, Debyser Z, Engelborghs Y, Engelman A. Identification and characterization of a functional nuclear localization signal in the HIV-1 integrase interactor LEDGF/p75. J Biol Chem 2004; 279:33421-9. [PMID: 15163664 DOI: 10.1074/jbc.m404700200] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human lens epithelium-derived growth factor (LEDGF)/p75 protein forms a specific nuclear complex with human immunodeficiency virus type 1 (HIV-1) integrase and is essential for nuclear localization and chromosomal association of the viral protein. We now studied nuclear import of LEDGF/p75 in live and semipermeabilized cells. We show that nuclear import of LEDGF/p75 is GTP-, Ran-, importin-alpha/beta-, and energy-dependent and that the protein competes with the canonical SV40 large T antigen nuclear localization signal (NLS) for nuclear import receptors. We identified the NLS of LEDGF/p75 through deletion analysis and site-directed mutagenesis. The LEDGF/p75 NLS, 148GRKRKAEKQ156, belongs to the canonical SV40-like family. Fusion of this short peptide to the amino terminus of Escherichia coli beta-galactosidase rendered the fusion protein nuclear, confirming that the LEDGF/p75 NLS is transferable. Moreover, a single amino acid change in the NLS was sufficient to exclude the mutant LEDGF/p75 protein from the nucleus and abolish nuclear import of HIV-1 integrase.
Collapse
Affiliation(s)
- Goedele Maertens
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
91
|
Abouzied MM, Baader SL, Dietz F, Kappler J, Gieselmann V, Franken S. Expression patterns and different subcellular localization of the growth factors HDGF (hepatoma-derived growth factor) and HRP-3 (HDGF-related protein-3) suggest functions in addition to their mitogenic activity. Biochem J 2004; 378:169-76. [PMID: 14572309 PMCID: PMC1223924 DOI: 10.1042/bj20030916] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2003] [Revised: 10/01/2003] [Accepted: 10/22/2003] [Indexed: 11/17/2022]
Abstract
HDGF (hepatoma-derived growth factor) and the HRPs (HDGF-related proteins) comprise a family of six proteins which display high identity in their N-terminus, but differ at the C-terminus. Here we investigate the patterns of expression of HDGF and HRP-3, by generating antisera specifically recognizing each growth factor. Whereas HRP-3 protein is expressed only in brain, HDGF can be found in a broad range of tissues, with highest levels in brain, testis, lung and spleen. The expression of HDGF and HRP-3 was found to be regulated during brain development, with highest levels around birth, followed by a decline until postnatal day 9. Interestingly, expression of HRP-3 increases again in adult brain. In situ hybridization and immunohistochemistry of cerebellar, cerebral and hippocampal brain slices showed that expression of both growth factors is not limited to areas of high proliferative activity. Both mRNAs and proteins are expressed in neuronal as well as glial cells. Immunocytochemistry of cultured neocortical neurons revealed that HDGF and HRP-3 can be found in the nucleus as well as the cytoplasm. HDGF is restricted to the neuronal soma, whereas HRP-3 can also be found in neurites. Thus the expression of HDGF and HRP-3 in differentiated cells, post-mitotic neurons and primary cultures of rat neocortex points to functions in brain that might not be limited to proliferation. In addition, their simultaneous expression in the same cell and their different subcellular localization in cultured neurons suggest different functions of HDGF and HRP-3 within single cells.
Collapse
Affiliation(s)
- Mekky M Abouzied
- Institut für Physiologische Chemie, Rheinische Friedrich-Wilhelms Universität, Nussallee 11, 53115 Bonn, Germany
| | | | | | | | | | | |
Collapse
|
92
|
Inomata Y, Hirata A, Koga T, Kimura A, Singh DP, Shinohara T, Tanihara H. Lens epithelium-derived growth factor: neuroprotection on rat retinal damage induced by N-methyl-D-aspartate. Brain Res 2004; 991:163-70. [PMID: 14575888 DOI: 10.1016/j.brainres.2003.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The purpose of this study is to investigate possible neuroprotective effects of lens epithelium-derived growth factor (LEDGF) against cell death induced by N-methyl-D-aspartate (NMDA) in the rat retina. LEDGF and/or NMDA were intravitreally injected into rat eyes. NMDA-induced retinal death and protective effects of LEDGF were evaluated by morphometric analysis, cell numbers in the ganglion cell layer (GCL) and the thickness of the inner plexiform layer (IPL). Retrograde labeling with a fluorescent tracer (Fluoro-Gold) was applied for counting retinal ganglion cells (RGCs) that survived after NMDA injection. Terminal deoxyribonucleotidyl transferase (TdT)-mediated fluroscein-16-dUTP nick end-labeling (TUNEL) staining was used to evaluate of retinal cell death. Morphometric analysis and retrograde labeling analysis showed that retinal damage induced by NMDA was protected significantly by LEDGF. TUNEL assay revealed that pretreatment with LEDGF prevents NMDA-induced apoptosis. Retinal damage (ganglion and amacrine cells) induced by NMDA was protected by an intravitreal injection of LEDGF.
Collapse
Affiliation(s)
- Yasuya Inomata
- Department of Ophthalmology and Visual Science, Kumamoto University Graduate School of Medical Sciences, 1-1-1, Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | |
Collapse
|
93
|
Ganapathy V, Daniels T, Casiano CA. LEDGF/p75: a novel nuclear autoantigen at the crossroads of cell survival and apoptosis. Autoimmun Rev 2003; 2:290-7. [PMID: 12965181 DOI: 10.1016/s1568-9972(03)00063-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Intracellular autoantigens recognized by autoantibodies in systemic autoimmune and other chronic inflammatory disorders often undergo proteolytic cleavage during apoptosis. Cleaved autoantigens may display altered functions and higher immunogenicity, and could potentially elicit autoantibody responses under a pro-inflammatory environment. LEDGF/p75 (lens epithelium derived growth factor p75) is a ubiquitous nuclear autoantigen targeted by autoantibodies in subsets of patients with atopic disorders, mainly atopic dermatitis, and other inflammatory conditions involving dysregulated apoptosis. Anti-LEDGF/p75 autoantibodies have been shown to have cytotoxic activity, suggesting their involvement in pathogenesis. LEDGF/p75 confers cellular protection against stress-induced apoptosis via transcriptional activation of stress-related genes. Recent studies in our laboratory established that LEDGF/p75 belongs to a selected group of autoantigens that are targeted for cleavage during cell death. In apoptotic cells, caspases cleave this protein at three sites located within functionally important domains, generating two fragments of 65 and 58 kD. Caspase cleavage not only abolishes the survival function of LEDGF/p75 but may generate variants of the protein that enhance apoptosis. A model is proposed in which caspase-induced LEDGF/p75 cleavage and the generation of autoantibodies to the protein might contribute to the pathogenesis of various human atopic and inflammatory disorders associated with dysregulated apoptosis.
Collapse
Affiliation(s)
- Vidya Ganapathy
- Department of Biochemistry and Microbiology, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | | | | |
Collapse
|
94
|
Maertens G, Cherepanov P, Pluymers W, Busschots K, De Clercq E, Debyser Z, Engelborghs Y. LEDGF/p75 is essential for nuclear and chromosomal targeting of HIV-1 integrase in human cells. J Biol Chem 2003; 278:33528-39. [PMID: 12796494 DOI: 10.1074/jbc.m303594200] [Citation(s) in RCA: 393] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We have reported that human immunodeficiency virus type 1 (HIV-1) integrase (IN) forms a specific nuclear complex with human lens epithelium-derived growth factor/transcription co-activator p75 (LEDGF/p75) protein. We now studied the IN-LEDGF/p75 interaction and nuclear import of IN in living cells using fusions of IN and LEDGF/p75 with enhanced green fluorescent protein and far-red fluorescent protein HcRed1. We show that both the N-terminal zinc binding domain and the central core domains of IN are involved in the interaction with LEDGF/p75. Both domains are essential for nuclear localization of IN as well as for the association of IN with condensed chromosomes during mitosis. However, upon overexpression of LEDGF/p75, the core domain fragment of IN was recruited to the nuclei and mitotic chromosomes with a distribution pattern characteristic of the full-length protein, indicating that it harbors the main determinant for interaction with LEDGF/p75. Although the C-terminal domain of IN was dispensable for nuclear/chromosomal localization, a fusion of the C-terminal IN fragment with enhanced green fluorescent protein was found exclusively in the nucleus, with a diffuse nuclear/nucleolar distribution, suggesting that the C-terminal domain may also play a role in the nuclear import of IN. In contrast to LEDGF/p75, its alternative splice variant, p52, did not interact with HIV-1 IN in vitro and in living cells. Finally, RNA interference-mediated knock-down of endogenous LEDGF/p75 expression abolished nuclear/chromosomal localization of IN. We conclude, therefore, that the interaction with LEDGF/p75 accounts for the karyophilic properties and chromosomal targeting of HIV-1 IN.
Collapse
Affiliation(s)
- Goedele Maertens
- Laboratory of Biomolecular Dynamics, Katholieke Universiteit Leuven, Celestijnenlaan 200D, B-3001 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
95
|
Bhat SP. Crystallins, genes and cataract. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2003; 60:205-62. [PMID: 12790344 DOI: 10.1007/978-3-0348-8012-1_7] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Far from being a physical entity, assembled of inanimate structural proteins, the ocular lens epitomizes the biological ingenuity that sustains an essential and near-perfect physical system of immaculate optics. Crystallins (alpha, beta, and gamma) provide transparency by dint of their high concentration, but it is debatable whether proteins that provide transparency are any different, biologically or structurally, from those that are present in non-transparent structures or tissues. It is becoming increasingly clear that crystallins may have a plethora of metabolic and regulatory functions, both within the lens as well as outside of it. Alpha-crystallins are members of a small heat shock family of proteins and beta/gamma-crystallins belong to the family of epidermis-specific differentiation proteins. Crystallin gene expression has been studied from the perspective of the lens specificity of their promoters. Mutations in alpha-, beta-, and gamma-crystallins are linked with the phenotype of the loss of transparency. Understanding catalytic, non-structural properties of crystallins may be critical for understanding the malfunction in molecular cascades that lead to cataractogenesis and its eventual therapeutic amelioration.
Collapse
Affiliation(s)
- Suraj P Bhat
- Jules Stein Eye Institute and Brain Research Institute, Geffen School of Medicine at UCLA, Los Angeles, CA 90077-7000, USA.
| |
Collapse
|
96
|
Sharma P, Fatma N, Kubo E, Shinohara T, Chylack LT, Singh DP. Lens epithelium-derived growth factor relieves transforming growth factor-beta1-induced transcription repression of heat shock proteins in human lens epithelial cells. J Biol Chem 2003; 278:20037-46. [PMID: 12649267 DOI: 10.1074/jbc.m212016200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lens epithelium-cell derived growth factor (LEDGF) is a transcriptional activator. It protects the cells by binding to cis-stress response ((A/T)GGGG(T/A)), and heat shock (HSE; nGAAn) elements in the stress genes and activating their transcription. Transforming growth factor-beta (TGF-beta) has been implicated in the control of tissue homeostasis, terminal differentiation, and apoptosis. Here we provide evidence that TGF-beta1 down-regulates LEDGF expression and diminishes its affinity for DNA during TGF-beta1-induced phenotypic changes and apoptosis in human lens epithelial cells. Surprisingly, TGF-beta1 treatment for 48 h markedly decreased the LEDGF, Hsp27, and alphaB-crystallin promoter activities with the decrease of abundance of LEDGF mRNA and protein. Deletion mutants of the LEDGF promoter showed that one TGF-beta1 inhibitory element (TIE) like sequence nnnTTGGnnn (-444 to -433) contributed to this negative regulation. Mutation of TIE (TTGG to TATT) abolished the down-regulation of the LEDGF promoter. Gel mobility and supershift assays showed that LEDGF in the nuclear extracts of TGF-beta1-treated human lens epithelial cells did not bind to stress-response elements and HSE. The TGF-beta1-induced down-regulation of LEDGF, Hsp27, and alphaB-crystallin promoters activity was reversed by cotransfection with a plasmid expressing LEDGF. Because overexpression of LEDGF was able to relieve TGF-beta1 and/or stress-induced changes, it would be a candidate molecule to postpone age-related degenerating disorders.
Collapse
Affiliation(s)
- Preeti Sharma
- Center for Ophthalmic Research, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
97
|
Nguyen TA, Boyle DL, Wagner LM, Shinohara T, Takemoto DJ. LEDGF activation of PKC gamma and gap junction disassembly in lens epithelial cells. Exp Eye Res 2003; 76:565-72. [PMID: 12697420 DOI: 10.1016/s0014-4835(03)00049-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Lens epithelium-derived growth factor (LEDGF) has been shown to enhance survival of lens epithelial cells (LECs) against stress. The objectives of these studies are to determine how LEDGF controls PKC gamma activity in normal LECs: how this control of PKC gamma regulates the phosphorylation of Connexin 43, the inhibition of gap junction activity, and the prevention of assembly of gap junctions in LECs. A rabbit LEC line, N/N1003A, was grown in the absence or presence of LEDGF. PKC gamma protein was translocated from the cytosolic fractions to the membrane fractions upon addition of LEDGF at 10 ng ml(-1). In whole cell extracts of N/N1003A cells, co-immunoprecipitation assays showed a protein-protein interaction between PKC gamma and Connexin 43. In the presence of LEDGF the activation of PKC gamma enhanced the phosphorylation of Connexin 43 by four-fold compared to the absence of LEDGF. The addition of LEDGF for 30 min resulted in a 65% decrease in gap junction Connexin 43 at the cell surface and a 70% decrease in gap junction activity. These results suggest that the activation of PKC gamma by LEDGF plays a major role in gap junction assembly/disassembly, which may enhance survival of LECs against osmolarity-stress induced by high sugar concentration.
Collapse
Affiliation(s)
- Thu Annelise Nguyen
- Department of Biochemistry, Kansas State University, Manhattan, KS 66506, USA
| | | | | | | | | |
Collapse
|
98
|
Kubo E, Singh DP, Fatma N, Shinohara T, Zelenka P, Reddy VN, Chylack LT. Cellular distribution of lens epithelium-derived growth factor (LEDGF) in the rat eye: loss of LEDGF from nuclei of differentiating cells. Histochem Cell Biol 2003; 119:289-99. [PMID: 12692670 DOI: 10.1007/s00418-003-0518-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2003] [Indexed: 01/07/2023]
Abstract
Lens epithelium-derived growth factor (LEDGF) enhances the survival and growth of cells. To understand LEDGF's spatial localization and its putative function(s) during proliferation and differentiation, we localized LEDGF during terminal differentiation in whole rat lenses, lens epithelial cell (LEC) explants stimulated with FGF-2, and insulin, iris, human LECs with lentoids. In addition, intracellular localization of LEDGF was performed in other ocular tissues: ciliary body, retina, and cornea. We found the immunopositivity of nuclear LEDGF decreased in LECs of the equatorial region. In contrast, immunopositivity of LEDGF was detected in the cytoplasm of LECs and superficial fiber cells. After treating LEC explants with FGF-2 and insulin, which are known to be differentiating factors for LECs, the nuclei of these cells showed no LEDGF immunopositivity, but explants did express p57(kip2), a differentiation marker protein. Also, immunopositive LEDGF was not detected in the nuclei of differentiated cells, lentoid body, and corneal epithelial cells. This demonstrated that the loss of LEDGF from the nucleus may be associated with the process of terminal differentiation that might be in some way common with the biochemical mechanisms of apoptosis. The spatial and temporal distribution of LEDGF in the present study also provides a vision for further investigation as to how this protein is involved in cell fate determination.
Collapse
Affiliation(s)
- Eri Kubo
- The Center for Ophthalmic Research, Brigham and Women's Hospital, Havard Medical School, 221 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
99
|
Cherepanov P, Maertens G, Proost P, Devreese B, Van Beeumen J, Engelborghs Y, De Clercq E, Debyser Z. HIV-1 integrase forms stable tetramers and associates with LEDGF/p75 protein in human cells. J Biol Chem 2003; 278:372-81. [PMID: 12407101 DOI: 10.1074/jbc.m209278200] [Citation(s) in RCA: 549] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
We studied human immunodeficiency virus, type 1 (HIV-1) integrase (IN) complexes derived from nuclei of human cells stably expressing the viral protein from a synthetic gene. We show that in the nuclear extracts IN exists as part of a large distinct complex with an apparent Stokes radius of 61 A, which dissociates upon dilution yielding a core molecule of 41 A. We isolated the IN complexes from cells expressing FLAG-tagged IN and demonstrated that the 41 A core is a tetramer of IN, whereas 61 A molecules are composed of IN tetramers associated with a cellular protein with an apparent molecular mass of 76 kDa. This novel integrase interacting protein was found to be identical to lens epithelium-derived growth factor (LEDGF/p75), a protein implicated in regulation of gene expression and cellular stress response. HIV-1 IN and LEDGF co-localized in the nuclei of human cells stably expressing IN. Furthermore, recombinant LEDGF robustly enhanced strand transfer activity of HIV-1 IN in vitro. Our findings indicate that the minimal IN molecule in human cells is a homotetramer, suggesting that at least an octamer of IN is required to accomplish coordinated integration of both retroviral long terminal repeats and that LEDGF is a cellular factor involved in this process.
Collapse
Affiliation(s)
- Peter Cherepanov
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000, Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
100
|
Sakaguchi H, Miyagi M, Darrow RM, Crabb JS, Hollyfield JG, Organisciak DT, Crabb JW. Intense light exposure changes the crystallin content in retina. Exp Eye Res 2003; 76:131-3. [PMID: 12589783 DOI: 10.1016/s0014-4835(02)00249-x] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Toward a better understanding of light-induced photoreceptor damage, the crystallin content of rat retina was examined following intense light exposure. Nine crystallin species were identified by mass spectrometric analysis of rat retina fractionated by 2D gel electrophoresis. The Coomassie blue staining intensity of all crystallin 2D gel components was 2- to 3-fold greater in light exposed than in control retinas. Following light exposure, anti-alphaB-crystallin immunoreactivity was increased in rod outer segments and retinal pigment epithelium. These findings support a possible role for crystallins in protecting photoreceptors from light damage.
Collapse
|