51
|
Strudwick XL, Cowin AJ. Cytoskeletal regulation of dermal regeneration. Cells 2012; 1:1313-27. [PMID: 24710556 PMCID: PMC3901152 DOI: 10.3390/cells1041313] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/15/2012] [Accepted: 12/04/2012] [Indexed: 12/21/2022] Open
Abstract
Wound healing results in the repair of injured tissues however fibrosis and scar formation are, more often than not the unfortunate consequence of this process. The ability of lower order vertebrates and invertebrates to regenerate limbs and tissues has been all but lost in mammals; however, there are some instances where glimpses of mammalian regenerative capacity do exist. Here we describe the unlocked potential that exists in mammals that may help us understand the process of regeneration post-injury and highlight the potential role of the actin cytoskeleton in this process. The precise function and regulation of the cytoskeleton is critical to the success of the healing process and its manipulation may therefore facilitate regenerative healing. The gelsolin family of actin remodelling proteins in particular has been shown to have important functions in wound healing and family member Flightless I (Flii) is involved in both regeneration and repair. Understanding the interactions between different cytoskeletal proteins and their dynamic control of processes including cellular adhesion, contraction and motility may assist the development of therapeutics that will stimulate regeneration rather than repair.
Collapse
Affiliation(s)
- Xanthe L Strudwick
- Wound Healing Laboratory, Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, South Australia 5006, Australia.
| | - Allison J Cowin
- Wound Healing Laboratory, Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, South Australia 5006, Australia.
| |
Collapse
|
52
|
G9a functions as a molecular scaffold for assembly of transcriptional coactivators on a subset of glucocorticoid receptor target genes. Proc Natl Acad Sci U S A 2012; 109:19673-8. [PMID: 23151507 DOI: 10.1073/pnas.1211803109] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Histone H3 lysine-9 methyltransferase G9a/EHMT2/KMT1C is a key corepressor of gene expression. However, activation of a limited number of genes by G9a (independent of its catalytic activity) has also been observed, although the precise molecular mechanisms are unknown. By using RNAi in combination with gene expression microarray analysis, we found that G9a functions as a positive and a negative transcriptional coregulator for discrete subsets of genes that are regulated by the hormone-activated Glucocorticoid Receptor (GR). G9a was recruited to GR-binding sites (but not to the gene body) of its target genes and interacted with GR, suggesting recruitment of G9a by GR. In contrast to its corepressor function, positive regulation of gene expression by G9a involved G9a-mediated enhanced recruitment of coactivators CARM1 and p300 to GR target genes. Further supporting a role for G9a as a molecular scaffold for its coactivator function, the G9a-specific methyltransferase inhibitor UNC0646 did not affect G9a coactivator function but selectively decreased G9a corepressor function for endogenous target genes. Overall, G9a functioned as a coactivator for hormone-activated genes and as a corepressor in support of hormone-induced gene repression, suggesting that the positive or negative actions of G9a are determined by the gene-specific regulatory environment and chromatin architecture. These findings indicate distinct mechanisms of G9a coactivator vs. corepressor functions in transcriptional regulation and provide insight into the molecular mechanisms of G9a coactivator function. Our results also suggest a physiological role of G9a in fine tuning the set of genes that respond to glucocorticoids.
Collapse
|
53
|
Tien JCY, Xu J. Steroid receptor coactivator-3 as a potential molecular target for cancer therapy. Expert Opin Ther Targets 2012; 16:1085-96. [PMID: 22924430 DOI: 10.1517/14728222.2012.718330] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Steroid receptor coactivator-3 (SRC-3), also called amplified-in-breast cancer-1 (AIB1), is an oncogenic coactivator in endocrine and non-endocrine cancers. Functional studies demonstrate SRC-3 promotes numerous aspects of cancer, through its capacity as a coactivator for nuclear hormone receptors and other transcription factors, and via its ability to control multiple growth pathways simultaneously. Targeting SRC-3 with specific inhibitors therefore holds future promise for clinical cancer therapy. AREAS COVERED We discuss critical advances in understanding SRC-3 as a cancer mediator and prospective drug target. We review SRC-3 structure and function and its role in distinct aspects of cancer. In addition, we discuss SRC-3 regulation and degradation. Finally, we comment on a recently discovered SRC-3 small molecular inhibitor. EXPERT OPINION Most targeted chemotherapeutic drugs block only a single cellular pathway. In response, cancers frequently acquire resistance by upregulating alternative pathways. SRC-3 coordinates multiple signaling networks, suggesting SRC-3 inhibition offers a promising therapeutic strategy. Development of an effective SRC-3 inhibitor faces critical challenges. Better understanding of SRC-3 function and interacting partners, in both the nucleus and cytosol, is required for optimized inhibitor development. Ultimately, blockade of SRC-3 oncogenic function may inhibit multiple cancer-related signaling pathways.
Collapse
Affiliation(s)
- Jean Ching-Yi Tien
- Baylor College of Medicine, Department of Molecular and Cellular Biology, One Baylor Plaza, Houston, TX 77030, USA
| | | |
Collapse
|
54
|
A functional study of nucleocytoplasmic transport signals of the EhNCABP166 protein from Entamoeba histolytica. Parasitology 2012; 139:1697-710. [PMID: 22906852 DOI: 10.1017/s0031182012001199] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
EhNCABP166 is an Entamoeba histolytica actin-binding protein that localizes to the nucleus and cytoplasm. Bioinformatic analysis of the EhNCABP166 amino acid sequence shows the presence of 3 bipartite nuclear localization signals (NLS) and a nuclear export signal (NES). The present study aimed to investigate the functionality of these signals in 3 ways. First, we fused each potential NLS to a cytoplasmic domain of ehFLN to determine whether the localization of this domain could be altered by the presence of the NLSs. Furthermore, the localization of each domain of EhNCABP166 was determined. Similarly, we generated mutations in the first block of bipartite signals from the domains that contained these signals. Additionally, we added an NES to 2 constructs that were then evaluated. We confirmed the intranuclear localization of EhNCABP166 using transmission electron microscopy. Fusion of each NLS resulted in shuttling of the cytoplasmic domain to the nucleus. With the exception of 2 domains, all of the evaluated domains localized within the nucleus. A mutation in the first block of bipartite signals affected the localization of the domains containing an NLS. The addition of an NES shifted the localization of these domains to the cytoplasm. The results presented here establish EhNCABP166 as a protein containing functional nuclear localization signals and a nuclear export signal.
Collapse
|
55
|
Khurana S, Chakraborty S, Zhao X, Liu Y, Guan D, Lam M, Huang W, Yang S, Kao HY. Identification of a novel LXXLL motif in α-actinin 4-spliced isoform that is critical for its interaction with estrogen receptor α and co-activators. J Biol Chem 2012; 287:35418-35429. [PMID: 22908231 DOI: 10.1074/jbc.m112.401364] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
α-Actinins (ACTNs) are a family of proteins cross-linking actin filaments that maintain cytoskeletal organization and cell motility. Recently, it has also become clear that ACTN4 can function in the nucleus. In this report, we found that ACTN4 (full length) and its spliced isoform ACTN4 (Iso) possess an unusual LXXLL nuclear receptor interacting motif. Both ACTN4 (full length) and ACTN4 (Iso) potentiate basal transcription activity and directly interact with estrogen receptor α, although ACTN4 (Iso) binds ERα more strongly. We have also found that both ACTN4 (full length) and ACTN4 (Iso) interact with the ligand-independent and the ligand-dependent activation domains of estrogen receptor α. Although ACTN4 (Iso) interacts efficiently with transcriptional co-activators such as p300/CBP-associated factor (PCAF) and steroid receptor co-activator 1 (SRC-1), the full length ACTN4 protein either does not or does so weakly. More importantly, the flanking sequences of the LXXLL motif are important not only for interacting with nuclear receptors but also for the association with co-activators. Taken together, we have identified a novel extended LXXLL motif that is critical for interactions with both receptors and co-activators. This motif functions more efficiently in a spliced isoform of ACTN4 than it does in the full-length protein.
Collapse
Affiliation(s)
- Simran Khurana
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Sharmistha Chakraborty
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Xuan Zhao
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Yu Liu
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Dongyin Guan
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Minh Lam
- Comprehensive Cancer Center, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Wei Huang
- Center for Proteomics and Department of Pharmacology, School of Medicine, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Sichun Yang
- Center for Proteomics and Department of Pharmacology, School of Medicine, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106
| | - Hung-Ying Kao
- Department of Biochemistry, Case Western Reserve University and the Research Institute of University Hospitals of Cleveland, Cleveland, Ohio 44106.
| |
Collapse
|
56
|
Jackson JE, Kopecki Z, Adams DH, Cowin AJ. Flii neutralizing antibodies improve wound healing in porcine preclinical studies. Wound Repair Regen 2012; 20:523-36. [PMID: 22672080 DOI: 10.1111/j.1524-475x.2012.00802.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 02/27/2012] [Indexed: 12/27/2022]
Abstract
Wound healing is an important area of widely unmet medical need, with millions of procedures carried out worldwide which could potentially benefit from a product to improve the wound repair process. Our studies investigating the actin-remodeling protein Flightless I (Flii) show it to be an important regulator of wound healing. Flii-deficient mice have enhanced wound healing in comparison to Flii overexpressing mice which have impaired wound healing. For the first time, we show that a Flightless I neutralizing monoclonal antibody (FnAb) therapy is effective in a large animal model of wound repair. Porcine 5 cm incisional and 6.25 cm(2) excisional wounds were treated with FnAb at the time of wounding and for two subsequent days. The wounds were dressed in Tegaderm dressings and left to heal by secondary intention for 7 and 35 days, respectively. At the relevant end points, the wounds were excised and processed for histological analysis. Parameters of wound area, collagen deposition, and scar appearance were analyzed. The results show that treatment with FnAb accelerates reepithelialization and improves the macroscopic appearance of early scars. FnAbs have the potential to enhance wound repair and reduce scar formation.
Collapse
Affiliation(s)
- Jessica E Jackson
- Women's and Children's Health Research Institute, North Adelaide, South Australia, Australia.
| | | | | | | |
Collapse
|
57
|
Won Jeong K, Chodankar R, Purcell DJ, Bittencourt D, Stallcup MR. Gene-specific patterns of coregulator requirements by estrogen receptor-α in breast cancer cells. Mol Endocrinol 2012; 26:955-66. [PMID: 22543272 DOI: 10.1210/me.2012-1066] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Progesterone receptor (PgR) controls the menstrual cycle, pregnancy, embryonic development, and homeostasis, and it plays important roles in breast cancer development and progression. However, the requirement of coregulators for estrogen-induced expression of the PgR gene has not been fully explored. Here we used RNA interference to demonstrate dramatic differences in requirements of 10 different coregulators for estrogen-regulated expression of six different genes, including PgR and the well-studied TFF1 (or pS2) gene in MCF-7 breast cancer cells. Full estrogen-induced expression of TFF1 required all ten coregulators, but PgR induction required only four of the 10 coregulators. Chromatin immunoprecipitation studies demonstrated several mechanisms responsible for the differential coregulator requirements. Actin-binding coregulator Flightless-I, required for TFF1 expression and recruited to that gene by estrogen receptor-α (ERα), is not required for PgR expression and not recruited to that gene. Protein acetyltransferase tat-interactive protein 60 and ATP-dependent chromatin remodeler Brahma Related Gene 1 are recruited to both genes but are required only for TFF1 expression. Histone methyltransferase G9a is recruited to both genes and required for estrogen-induced expression of TFF1 but negatively regulates estrogen-induced expression of PgR. In contrast, histone methyltransferase myeloid/lymphoid or mixed-lineage leukemia 1 (MLL1), pioneer factor Forkhead box A1, and p160 coregulator steroid receptor coactivator-3 are required for expression of and are recruited to both genes. Depletion of MLL1 decreased ERα binding to the PgR and TFF1 genes. In contrast, depletion of G9a enhanced ERα binding to the PgR gene but had no effect on ERα binding to the TFF1 gene. These studies suggest that differential promoter architecture is responsible for promoter-specific mechanisms of gene regulation.
Collapse
Affiliation(s)
- Kwang Won Jeong
- Department of Biochemistry and Molecular Biology, University of Southern California/Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90089, USA
| | | | | | | | | |
Collapse
|
58
|
King HA, Trotter KW, Archer TK. Chromatin remodeling during glucocorticoid receptor regulated transactivation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:716-26. [PMID: 22425674 DOI: 10.1016/j.bbagrm.2012.02.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/24/2012] [Accepted: 02/28/2012] [Indexed: 12/21/2022]
Abstract
Steroid hormone receptor (SR) signaling leads to widespread changes in gene expression, and aberrant SR signaling can lead to malignancies including breast, prostate, and lung cancers. Chromatin remodeling is an essential component of SR signaling, and defining the process of chromatin and nucleosome remodeling during signaling is critical to the continued development of related therapies. The glucocorticoid receptor (GR) is a key SR that activates numerous promoters including the well defined MMTV promoter. The activation of MMTV by GR provides an excellent model for teasing apart the sequence of events between hormone treatment and changes in gene expression. Comparing hormone-induced transcription from stably integrated promoters with defined nucleosomal structure to that from transiently expressed, unstructured promoters permits key distinctions between interactions that require remodeling and those that do not. The importance of co-activators and histone modifications prior to remodeling and the formation of the preinitiation complex that follows can also be clarified by defining key transition points in the propagation of hormonal signals. Combined with detailed mapping of proteins along the promoter, a temporal and spatial understanding of the signaling and remodeling processes begins to emerge. In this review, we examine SR signaling with a focus on GR activation of the MMTV promoter. We also discuss the ATP-dependent remodeling complex SWI/SNF, which provides the necessary remodeling activity during GR signaling and interacts with several SRs. BRG1, the central ATPase of SWI/SNF, also interacts with a set of BAF proteins that help determine the specialized function and fine-tuned regulation of BRG1 remodeling activity. BRG1 regulation comes from its own subdomains as well as its interactive partners. In particular, the HSA domain region of BRG1 and unique features of its ATPase homology appear to play key roles in regulating remodeling function. Details of the inter-workings of this chromatin remodeling protein continue to be revealed and promise to improve our understanding of the mechanism of chromatin remodeling during steroid hormone signaling. This article is part of a Special Issue entitled: Chromatin in time and space.
Collapse
Affiliation(s)
- Heather A King
- Laboratory of Molecular Carcinogenesis, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | | |
Collapse
|
59
|
Johnson AB, O'Malley BW. Steroid receptor coactivators 1, 2, and 3: critical regulators of nuclear receptor activity and steroid receptor modulator (SRM)-based cancer therapy. Mol Cell Endocrinol 2012; 348:430-9. [PMID: 21664237 PMCID: PMC3202666 DOI: 10.1016/j.mce.2011.04.021] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 04/04/2011] [Accepted: 04/22/2011] [Indexed: 01/17/2023]
Abstract
Coactivators are a diverse group of non-DNA binding proteins that induce structural changes in agonist-bound nuclear receptors (NRs) that are essential for NR-mediated transcriptional activation. Once bound, coactivators function to bridge enhancer binding proteins to the general transcription machinery, as well as to recruit secondary coactivators that modify promoter and enhancer chromatin in a manner permissive for transcriptional activation. In the following review article, we focus on one of the most in-depth studied families of coactivators, the steroid receptor coactivators (SRC) 1, 2, and 3. SRCs are widely implicated in NR-mediated diseases, especially in cancers, with the majority of studies focused on their roles in breast cancer. We highlight the relevant literature supporting the oncogenic activity of SRCs and their future as diagnostic and prognostic indicators. With much interest in the development of selective receptor modulators (SRMs), we focus on how these coactivators regulate the interactions between SRMs and their respective NRs; and, importantly, the influence that coactivators have on the functional output of SRMs. Furthermore, we speculate that coactivator-specific inhibitors could provide powerful, all-encompassing treatments that target multiple modes of oncogenic regulation in cancers resistant to typical anti-endocrine treatments.
Collapse
Affiliation(s)
- Amber B Johnson
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, United States
| | | |
Collapse
|
60
|
Lei N, Franken L, Ruzehaji N, Offenhäuser C, Cowin AJ, Murray RZ. Flightless, secreted through a late endosome/lysosome pathway, binds LPS and dampens cytokine secretion. J Cell Sci 2012; 125:4288-96. [DOI: 10.1242/jcs.099507] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Flightless (Flii) is upregulated in response to wounding and has been shown to function in wound closure and scarring. In macrophages intracellular Flii negatively modulates TLR signalling and dampens cytokine production. We now show that Flii is constitutively secreted from macrophages and fibroblasts and is present in human plasma. Secretion from fibroblasts is upregulated in response to scratch wounding and LPS-activated macrophages also temporally upregulate their secretion of Flii. Using siRNA, wild-type and mutant proteins we show that Flii is secreted via a late endosomal/lysosomal pathway that is regulated by Rab7 and Stx11. Flii contains 11 leucine rich repeat (LRR) domains in its N-terminus that have nearly 50% similarity to those in the extracellular pathogen binding portion of Toll-like receptor 4 (TLR4). We show secreted Flii can also bind LPS and has the ability to alter macrophage activation. LPS activation of macrophages in Flii depleted conditioned media leads to enhanced macrophage activation and increased TNF secretion compared to cells activated in the presence of Flii. These results show secreted Flii binds to LPS and in doing so alters macrophage activation and cytokine secretion, suggesting that like the intracellular pool of Flii, secreted Flii also has the ability to alter inflammation.
Collapse
|
61
|
Glucocorticoid-dependent phosphorylation of the transcriptional coregulator GRIP1. Mol Cell Biol 2011; 32:730-9. [PMID: 22158970 DOI: 10.1128/mcb.06473-11] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Much of the regulatory diversity in eukaryotic transcription is provided by coregulators, which are recruited by DNA-binding factors to propagate signaling to basal machinery or chromatin. p160 family members, including the glucocorticoid receptor (GR)-interacting protein 1 (GRIP1), function as coactivators for GR, a ligand-dependent transcription factor of the nuclear receptor superfamily. Unlike other p160s, GRIP1 also potentiates GR-mediated repression of AP1 and NF-κB targets and, surprisingly, transcriptional activation by interferon regulatory factors. What enables GRIP1 activating or repressing properties or discrimination between physiologically antagonistic pathways is unknown. We found that endogenous GRIP1 in mammalian cells undergoes glucocorticoid-induced, GR interaction-dependent phosphorylation and identified one constitutive and six inducible phosphorylation sites and two putative GRIP1 kinases, casein kinase 2 and cyclin-dependent kinase 9. We raised phosphospecific antibodies to the four closely spaced sites in a previously uncharacterized part of GRIP1 which, combined with mutagenesis, revealed the conservation of GRIP1 phosphorylation across several cell types and species and its functional relevance to GR-activated transcription and to response element-specific recruitment of phospho-GRIP1 to native GR targets. We propose that cofactor engagement by GR is neither passive nor stochastic; rather, GR actively imparts modifications that dictate GRIP1 function in a subset of complexes, adding a layer of specificity to GR transcriptional control.
Collapse
|
62
|
Kopecki Z, Arkell RM, Strudwick XL, Hirose M, Ludwig RJ, Kern JS, Bruckner-Tuderman L, Zillikens D, Murrell DF, Cowin AJ. Overexpression of the Flii
gene increases dermal-epidermal blistering in an autoimmune ColVII mouse model of epidermolysis bullosa acquisita. J Pathol 2011; 225:401-13. [PMID: 21984127 DOI: 10.1002/path.2973] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 07/18/2011] [Indexed: 11/06/2022]
Affiliation(s)
- Zlatko Kopecki
- Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, SA, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
An JH, Kim JW, Jang SM, Kim CH, Kang EJ, Choi KH. Gelsolin negatively regulates the activity of tumor suppressor p53 through their physical interaction in hepatocarcinoma HepG2 cells. Biochem Biophys Res Commun 2011; 412:44-9. [PMID: 21801713 DOI: 10.1016/j.bbrc.2011.07.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 07/08/2011] [Indexed: 10/18/2022]
Abstract
As a transcription factor, p53 modulates several cellular responses including cell-cycle control, apoptosis, and differentiation. In this study, we have shown that an actin regulatory protein, gelsolin (GSN), can physically interact with p53. The nuclear localization of p53 is inhibited by GSN overexpression in hepatocarcinoma HepG2 cells. Additionally, we demonstrate that GSN negatively regulates p53-dependent transcriptional activity of a reporter construct, driven by the p21-promoter. Furthermore, p53-mediated apoptosis was repressed in GSN-transfected HepG2 cells. Taken together, these results suggest that GSN binds to p53 and this interaction leads to the inhibition of p53-induced apoptosis by anchoring of p53 in the cytoplasm in HepG2 cells.
Collapse
Affiliation(s)
- Joo-Hee An
- Department of Life Science (BK21 Program), College of Natural Sciences, Chung-Ang University, Seoul 156-756, Republic of Korea
| | | | | | | | | | | |
Collapse
|
64
|
Thomsen N, Chappell A, Ali RG, Jones T, Adams DH, Matthaei KI, Campbell HD, Cowin AJ, Arkell RM. Mouse strains for the ubiquitous or conditional overexpression of the Flii gene. Genesis 2011; 49:681-8. [PMID: 21786402 DOI: 10.1002/dvg.20735] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 01/31/2011] [Accepted: 02/07/2011] [Indexed: 11/07/2022]
Abstract
The gelsolin related actin binding protein, Flii, is able to regulate wound healing; mice with decreased Flii expression show improved wound healing whereas mice with elevated Flii expression exhibit impaired wound healing. In both mice and humans Flii expression increases with age and amelioration of FLII activity represents a possible therapeutic strategy for improved wound healing in humans. Despite analysis of Flii function in a variety of organisms we know little of the molecular mechanisms underlying Flii action. Two new murine alleles of Flii have been produced to drive constitutive or tissue-specific expression of Flii. Each strain is able to rescue the embryonic lethality associated with a Flii null allele and to impair wound healing. These strains provide valuable resources for ongoing investigation of Flii function in a variety of biological processes.
Collapse
Affiliation(s)
- Nicole Thomsen
- Centre for Molecular Genetics of Development, Research School of Biology, The Australian National University, Canberra, ACT, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Regeneration of Hair Follicles Is Modulated by Flightless I (Flii) in a Rodent Vibrissa Model. J Invest Dermatol 2011; 131:838-47. [DOI: 10.1038/jid.2010.393] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
66
|
Lin CH, Waters JM, Powell BC, Arkell RM, Cowin AJ. Decreased expression of Flightless I, a gelsolin family member and developmental regulator, in early-gestation fetal wounds improves healing. Mamm Genome 2011; 22:341-52. [PMID: 21400204 DOI: 10.1007/s00335-011-9320-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 02/08/2011] [Indexed: 02/07/2023]
Abstract
Up until late in the third trimester of gestation and through to adulthood, the healing response acts more to regenerate than to repair a wound. The mechanisms underlying this "scar-free" healing remain unknown although the actin cytoskeleton has a major role. Flightless I (Flii), an actin-remodelling protein and essential developmental regulator, negatively affects wound repair but its effect on scar-free fetal healing is unknown. Using fetal skin explants from E17 (regenerate) and E19 (repair) rats, the function of Flii in fetal wound repair was determined. Expression of Flii increased between E17 and E19 days of gestation and wounding transiently increased Flii expression in E17 but not E19 wounds. However, both confocal and immunofluorescent analysis showed E17 keratinocytes immediately adjacent to the wounds downregulated Flii. As a nuclear coactivator and inhibitor of proliferation and migration, the absence of Flii in cells at the edge of the wound could be instrumental in allowing these cells to proliferate and migrate into the wound deficit. In contrast, Flii was strongly expressed within the cytoplasm and nucleus of keratinocytes within epidermal cells at the leading edge of E19 wounded fetal skin explants. This increase in Flii expression in E19 wounds could affect the way these cells migrate into the wound space and contribute to impaired wound healing. Neutralising Flii protein improved healing of early- but not late-gestation wounds. Flii did not colocalise with actin cables formed around E17 wounds suggesting an independent mechanism of action distinct from its actin-binding function in scar-free wound repair.
Collapse
Affiliation(s)
- Cheng-Hung Lin
- Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, SA 5006, Australia
| | | | | | | | | |
Collapse
|
67
|
Lee YH, Bedford MT, Stallcup MR. Regulated recruitment of tumor suppressor BRCA1 to the p21 gene by coactivator methylation. Genes Dev 2011; 25:176-88. [PMID: 21245169 DOI: 10.1101/gad.1975811] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Tumor suppression by p53 and BRCA1 involves regulation of cell cycle, apoptosis, and DNA repair and is influenced by transcriptional coactivators and post-translational modifications. Here we show that coactivator-associated arginine methyltransferase 1 (CARM1) methylates Arg 754 in the KIX region of coactivator p300. Methylated p300 and p300 protein fragments are preferentially recognized by BRCT domains of BRCA1, identifying the BRCT domain as a novel methylarginine-binding module. CARM1 and p300 cooperate with BRCA1 and p53 to induce expression of the critical cell cycle and proliferation regulator p21(WAF1/CIP1) in response to DNA damage. This induction was severely attenuated by elimination of CARM1 or its methyltransferase activity, or by mutation of Arg 754 of p300. Absence of CARM1 methyltransferase activity led to failure of cells to arrest in the G1 phase of the cell cycle in response to DNA damage. CARM1 methyltransferase activity was required for induction of some p53 target genes (p21 and Gadd45) but not others (Bax) by DNA damage. Recruitment of BRCA1 to the p53-binding region of the p21 promoter in response to DNA damage required methylation of Arg 754 of p300 by CARM1. Thus, coactivator methylation may be crucial for fine-tuning the tumor suppressor function of BRCA1 and other BRCT domain proteins.
Collapse
Affiliation(s)
- Young-Ho Lee
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California 90089, USA
| | | | | |
Collapse
|
68
|
Forsthoefel NR, Vernon DM. Effect of sporophytic PIRL9 genotype on post-meiotic expression of the Arabidopsis pirl1;pirl9 mutant pollen phenotype. PLANTA 2011; 233:423-31. [PMID: 21140166 DOI: 10.1007/s00425-010-1324-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2010] [Accepted: 11/12/2010] [Indexed: 05/07/2023]
Abstract
Plant intracellular ras-group-related leucine-rich repeat proteins (PIRLs) are a novel class of plant leucine-rich repeat (LRR) proteins structurally related to animal ras-group LRRs involved in cell signaling and gene regulation. Gene knockout analysis has shown that two members of the Arabidopsis thaliana PIRL gene family, PIRL1 and PIRL9, are redundant and essential for pollen development and viability: pirl1;pirl9 microspores produced by pirl1/PIRL1;pirl9 plants consistently abort just before pollen mitosis I. qrt1 tetrad analysis demonstrated that the genes become essential after meiosis, during anther stage 10. In this study, we characterized the phenotype of pirl1;pirl9 pollen produced by plants heterozygous for pirl9 (pirl1;pirl9/PIRL9). Alexander's staining, scanning electron microscopy, and fluorescence microscopy indicated that pirl1;pirl9 double mutants produced by pirl9 heterozygotes have a less severe phenotype and more variable morphology than pirl1;pirl9 pollen from pirl1/PIRL1;pirl9 plants. Mutant pollen underwent developmental arrest with variable timing, often progressing beyond pollen mitosis I and arresting at the binucleate stage. Thus, although the pirl1 and pirl9 mutations act post-meiosis, the timing and expressivity of the pirl1;pirl9 pollen phenotype depends on the pirl9 genotype of the parent plant. These results suggest a continued requirement for PIRL1 and PIRL9 beyond the initiation of pollen mitosis. Furthermore, they reveal a modest but novel sporophytic effect in which parent plant genotype influences a mutant phenotype expressed in the haploid generation.
Collapse
|
69
|
|
70
|
Oma Y, Harata M. Actin-related proteins localized in the nucleus: from discovery to novel roles in nuclear organization. Nucleus 2011; 2:38-46. [PMID: 21647298 PMCID: PMC3104808 DOI: 10.4161/nucl.2.1.14510] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 12/15/2010] [Accepted: 12/17/2010] [Indexed: 12/18/2022] Open
Abstract
The actin family consists of conventional actin and actin-related proteins (ARPs), and the members show moderate similarity and share the same basal structure. Following the finding of various ARPs in the cytoplasm in the 1990s, multiple subfamilies that are localized predominantly in the nucleus were identified. Consistent with these cytological observations, subsequent biochemical analyses revealed the involvement of the nuclear ARPs in ATP-dependent chromatin-remodeling and histone acetyltransferase complexes. In addition to their contribution to chromatin remodeling, recent studies have shown that nuclear ARPs have roles in the organization of the nucleus that are independent of the activity of the above-mentioned complexes. Therefore, nuclear ARPs are recognized as novel key regulators of genome function, and affect not only the remodeling of chromatin but also the spatial arrangement and dynamics of chromatin within the nucleus.
Collapse
Affiliation(s)
- Yukako Oma
- Laboratory of Molecular Biology, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | | |
Collapse
|
71
|
Li GH, Arora PD, Chen Y, McCulloch CA, Liu P. Multifunctional roles of gelsolin in health and diseases. Med Res Rev 2010; 32:999-1025. [PMID: 22886630 DOI: 10.1002/med.20231] [Citation(s) in RCA: 184] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Gelsolin, a Ca(2+) -regulated actin filament severing, capping, and nucleating protein, is an ubiquitous, multifunctional regulator of cell structure and metabolism. More recent data show that gelsolin can act as a transcriptional cofactor in signal transduction and its own expression and function can be influenced by epigenetic changes. Here, we review the functions of the plasma and cytoplasmic forms of gelsolin, and their manifold impacts on cancer, apoptosis, infection and inflammation, cardiac injury, pulmonary diseases, and aging. An improved understanding of the functions and regulatory mechanisms of gelsolin may lead to new considerations of this protein as a potential biomarker and/or therapeutic target.
Collapse
Affiliation(s)
- Guo Hua Li
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | |
Collapse
|
72
|
Forsthoefel NR, Dao TP, Vernon DM. PIRL1 and PIRL9, encoding members of a novel plant-specific family of leucine-rich repeat proteins, are essential for differentiation of microspores into pollen. PLANTA 2010; 232:1101-1114. [PMID: 20697737 DOI: 10.1007/s00425-010-1242-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 07/26/2010] [Indexed: 05/29/2023]
Abstract
Plant intracellular Ras-group-related leucine-rich repeat proteins (PIRLs) are a plant-specific class of leucine-rich repeat (LRR) proteins related to animal and fungal LRRs that take part in developmental signaling and gene regulation. As part of a systematic functional study of the Arabidopsis thaliana PIRL gene family, T-DNA knockout mutants defective in the closely related PIRL1 and PIRL9 genes were identified and characterized. Pirl1 and pirl9 single mutants displayed normal transmission and did not exhibit an obvious developmental phenotype. To investigate the possibility of functional redundancy, crosses to generate double mutants were carried out; however, pirl1;pirl9 plants were not recovered. Reciprocal crosses between wild type and pirl1/PIRL1;pirl9 plants, which produce 50% pirl1;pirl9 gametophytes, indicated male-specific transmission failure of the double-mutant allele combination. Scanning electron microscopy and viability staining showed that approximately half of the pollen produced by pirl1/PIRL1;pirl9 plants was inviable and severely malformed. Tetrad analyses with qrt1 indicated that pollen defects segregated with the double-mutant allele combination, thus demonstrating that PIRL1 and PIRL9 function after meiosis. Pollen development was characterized with bright field, fluorescence, and transmission electron microscopy. Pirl1;pirl9 mutants stopped growing as microspores, failed to initiate vacuolar fission, aborted, and underwent cytoplasmic degeneration. Development consistently arrested at the late microspore stage, just prior to pollen mitosis I. Thus, PIRL1 and PIRL9 have redundant roles essential at a key transition point early in pollen development. Together, these results define a functional context for these two members of this distinct class of plant LRR genes.
Collapse
|
73
|
Ambrosino C, Tarallo R, Bamundo A, Cuomo D, Franci G, Nassa G, Paris O, Ravo M, Giovane A, Zambrano N, Lepikhova T, Jänne OA, Baumann M, Nyman TA, Cicatiello L, Weisz A. Identification of a hormone-regulated dynamic nuclear actin network associated with estrogen receptor alpha in human breast cancer cell nuclei. Mol Cell Proteomics 2010; 9:1352-67. [PMID: 20308691 PMCID: PMC2877992 DOI: 10.1074/mcp.m900519-mcp200] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 02/22/2010] [Indexed: 11/06/2022] Open
Abstract
Estrogen receptor alpha (ERalpha) is a modular protein of the steroid/nuclear receptor family of transcriptional regulators that upon binding to the hormone undergoes structural changes, resulting in its nuclear translocation and docking to specific chromatin sites. In the nucleus, ERalpha assembles in multiprotein complexes that act as final effectors of estrogen signaling to the genome through chromatin remodeling and epigenetic modifications, leading to dynamic and coordinated regulation of hormone-responsive genes. Identification of the molecular partners of ERalpha and understanding their combinatory interactions within functional complexes is a prerequisite to define the molecular basis of estrogen control of cell functions. To this end, affinity purification was applied to map and characterize the ERalpha interactome in hormone-responsive human breast cancer cell nuclei. MCF-7 cell clones expressing human ERalpha fused to a tandem affinity purification tag were generated and used to purify native nuclear ER-containing complexes by IgG-Sepharose affinity chromatography and glycerol gradient centrifugation. Purified complexes were analyzed by two-dimensional DIGE and mass spectrometry, leading to the identification of a ligand-dependent multiprotein complex comprising beta-actin, myosins, and several proteins involved in actin filament organization and dynamics and/or known to participate in actin-mediated regulation of gene transcription, chromatin dynamics, and ribosome biogenesis. Time course analyses indicated that complexes containing ERalpha and actin are assembled in the nucleus early after receptor activation by ligands, and gene knockdown experiments showed that gelsolin and the nuclear isoform of myosin 1c are key determinants for assembly and/or stability of these complexes. Based on these results, we propose that the actin network plays a role in nuclear ERalpha actions in breast cancer cells, including coordinated regulation of target gene activity, spatial and functional reorganization of chromatin, and ribosome biogenesis.
Collapse
Affiliation(s)
- Concetta Ambrosino
- From the Departments of General Pathology and
- Department of Biological and Environmental Sciences, University of Sannio, 82100 Benevento, Italy
| | | | | | | | | | | | - Ornella Paris
- From the Departments of General Pathology and
- Associazione Italiana per la Ricerca sul Cancro (AIRC) Naples Oncogenomics Center, 80145 Naples, Italy
| | - Maria Ravo
- From the Departments of General Pathology and
| | - Alfonso Giovane
- Biochemistry and Biophysics “F. Cedrangolo,” Second University of Naples, 80138 Naples, Italy
| | - Nicola Zambrano
- CEINGE Biotecnologie Avanzate and Department of Biochemistry and Medical Biotechnologies, University of Naples Federico II, 80145 Naples, Italy
| | | | | | - Marc Baumann
- Protein Chemistry Unit, Biomedicum Helsinki, University of Helsinki, 00290 Helsinki, Finland
| | - Tuula A. Nyman
- Protein Chemistry Research Group, Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland, and
| | - Luigi Cicatiello
- From the Departments of General Pathology and
- Associazione Italiana per la Ricerca sul Cancro (AIRC) Naples Oncogenomics Center, 80145 Naples, Italy
| | - Alessandro Weisz
- From the Departments of General Pathology and
- Associazione Italiana per la Ricerca sul Cancro (AIRC) Naples Oncogenomics Center, 80145 Naples, Italy
- Molecular Medicine Laboratory, Faculty of Medicine and Surgery, University of Salerno, 84081 Baronissi, Italy
| |
Collapse
|
74
|
Castano E, Philimonenko VV, Kahle M, Fukalová J, Kalendová A, Yildirim S, Dzijak R, Dingová-Krásna H, Hozák P. Actin complexes in the cell nucleus: new stones in an old field. Histochem Cell Biol 2010; 133:607-26. [PMID: 20443021 DOI: 10.1007/s00418-010-0701-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2010] [Indexed: 01/13/2023]
Abstract
Actin is a well-known protein that has shown a myriad of activities in the cytoplasm. However, recent findings of actin involvement in nuclear processes are overwhelming. Actin complexes in the nucleus range from very dynamic chromatin-remodeling complexes to structural elements of the matrix with single partners known as actin-binding proteins (ABPs). This review summarizes the recent findings of actin-containing complexes in the nucleus. Particular attention is given to key processes like chromatin remodeling, transcription, DNA replication, nucleocytoplasmic transport and to actin roles in nuclear architecture. Understanding the mechanisms involving ABPs will definitely lead us to the principles of the regulation of gene expression performed via concerting nuclear and cytoplasmic processes.
Collapse
Affiliation(s)
- E Castano
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Hayashi T, Funato Y, Terabayashi T, Morinaka A, Sakamoto R, Ichise H, Fukuda H, Yoshida N, Miki H. Nucleoredoxin negatively regulates Toll-like receptor 4 signaling via recruitment of flightless-I to myeloid differentiation primary response gene (88). J Biol Chem 2010; 285:18586-93. [PMID: 20400501 DOI: 10.1074/jbc.m110.106468] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We previously characterized nucleoredoxin (NRX) as a negative regulator of the Wnt signaling pathway through Dishevelled (Dvl). We perform a comprehensive search for other NRX-interacting proteins and identify Flightless-I (Fli-I) as a novel NRX-binding partner. Fli-I binds to NRX and other related proteins, such as Rod-derived cone viability factor (RdCVF), whereas Dvl binds only to NRX. Endogenous NRX and Fli-I in vivo interactions are confirmed. Both NRX and RdCVF link Fli-I with myeloid differentiation primary response gene (88) (MyD88), an important adaptor protein for innate immune response. NRX and RdCVF also potentiate the negative effect of Fli-I upon lipopolysaccharide-induced activation of NF-kappaB through the Toll-like receptor 4/MyD88 pathway. Embryonic fibroblasts derived from NRX gene-targeted mice show aberrant NF-kappaB activation upon lipopolysaccharide stimulation. These results suggest that the NRX subfamily of proteins forms a link between MyD88 and Fli-I to mediate negative regulation of the Toll-like receptor 4/MyD88 pathway.
Collapse
Affiliation(s)
- Tatsuya Hayashi
- Laboratory of Intracellular Signaling, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Carascossa S, Dudek P, Cenni B, Briand PA, Picard D. CARM1 mediates the ligand-independent and tamoxifen-resistant activation of the estrogen receptor alpha by cAMP. Genes Dev 2010; 24:708-19. [PMID: 20360387 DOI: 10.1101/gad.568410] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The estrogen receptor alpha (ERalpha) is activated as a transcription factor by both estrogen and a large variety of other extracellular signals. The mechanisms of this ligand-independent activation, notably by cAMP signaling, are still largely unknown. We now close the gap in the signaling pathway between cAMP and ERalpha. Whereas the direct phosphorylation of ERalpha by the cAMP-activated protein kinase A (PKA) is dispensable, the phosphorylation of the coactivator-associated arginine methyltransferase 1 (CARM1) by PKA at a single serine is necessary and sufficient for direct binding to the unliganded hormone-binding domain (HBD) of ERalpha, and the interaction is necessary for cAMP activation of ERalpha. Sustained PKA activity promoting a constitutive interaction may contribute to tamoxifen resistance of breast tumors. Binding and activation involve a novel regulatory groove of the ERalpha HBD. As a result, depending on the activating signal, ERalpha recruits different coactivator complexes to regulate alternate sets of target genes.
Collapse
Affiliation(s)
- Sophie Carascossa
- Département de Biologie Cellulaire, Université de Genève, Sciences III, CH-1211 Genève 4, Switzerland
| | | | | | | | | |
Collapse
|
77
|
Harrison MJ, Tang YH, Dowhan DH. Protein arginine methyltransferase 6 regulates multiple aspects of gene expression. Nucleic Acids Res 2010; 38:2201-16. [PMID: 20047962 PMCID: PMC2853117 DOI: 10.1093/nar/gkp1203] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
It is well established that transcription and alternative splicing events are functionally coupled during gene expression. Here, we report that protein arginine N-methyltransferase 6 (PRMT6) may play a key role in this coupling process by functioning as a transcriptional coactivator that can also regulate alternative splicing. PRMT6 coactivates the progesterone, glucocorticoid and oestrogen receptors in luciferase reporter assays in a hormone-dependent manner. In addition, small interfering RNA (siRNA) oligonucleotide duplex knockdown of PRMT6 disrupts oestrogen-stimulated transcription of endogenous GREB1 and progesterone receptor in MCF-7 breast cancer cells, demonstrating the importance of PRMT6 in hormone-dependent transcription. In contrast, the regulation of alternative splicing by PRMT6 is hormone independent. siRNA knockdown of PRMT6 increases the exon inclusion:skipping ratio of alternatively spliced exons in endogenous vascular endothelial growth factor and spleen tyrosine kinase RNA transcripts in both the presence and absence of oestrogen. These results demonstrate that PRMT6 has a dual role in regulating gene expression and that these two functions can occur independently of each other.
Collapse
Affiliation(s)
- Matthew J Harrison
- The University of Queensland, Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, Woolloongabba, QLD 4102, Australia
| | | | | |
Collapse
|
78
|
Kim D, Lee J, Cheng D, Li J, Carter C, Richie E, Bedford MT. Enzymatic activity is required for the in vivo functions of CARM1. J Biol Chem 2009; 285:1147-52. [PMID: 19897492 DOI: 10.1074/jbc.m109.035865] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CARM1 is one of nine protein arginine methyltransferases that methylate arginine residues in proteins. CARM1 is recruited by many different transcription factors as a positive regulator. Gene targeting of CARM1 in mice has been performed, and knock-out mice, which are smaller than their wild-type littermates, die just after birth. It has been proposed that CARM1 has functions that are independent of its enzymatic activity. Indeed, CARM1 is found to interact with a number of proteins and may have a scaffolding function in this context. However, CARM1 methylates histone H3, PABP1, AIB1, and a number of splicing factors, which strongly suggests that its impact on transcription and splicing is primarily through its ability to modify these substrates. To unequivocally establish the importance of CARM1 enzymatic activity in vivo, we generated an enzyme-dead knock-in of this protein arginine methyltransferase. We determined that knock-in cells and mice have defects similar to those seen in their knock-out counterparts with respect to the time of embryo lethality, T cell development, adipocyte differentiation, and transcriptional coactivator activity. CARM1 requires its enzymatic activity for all of its known cellular functions. Thus, small molecule inhibitors of CARM1 will incapacitate all of the enzyme's cellular functions.
Collapse
Affiliation(s)
- Daehoon Kim
- Science Park-Research Division, The University of Texas M. D. Anderson Cancer Center, Smithville, Texas 78957, USA
| | | | | | | | | | | | | |
Collapse
|
79
|
Abstract
Coregulators (coactivators and corepressors) occupy the driving seat for actions of all nuclear receptors, and consequently, selective receptor modulator drugs. The potency and selectivity for subreactions of transcription reside in the coactivators, and thus, they are critically important for tissue-selective gene function. Each tissue has a "quantitative finger print" of coactivators based on its relative inherited concentrations of these molecules. When the cellular concentration of a coactivator is altered, genetic dysfunction usually leads to a pathologic outcome. For example, many cancers overexpress "growth coactivators." In this way, the cancer cell can hijack these coactivator molecules to drive proliferation and metastasis. The present review contains summaries of selective coactivators and corepressors that have been demonstrated to play important roles in the malignant process and emphasizes their importance for future therapeutic interventions.
Collapse
Affiliation(s)
- Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
80
|
Kuhn P, Xu W. Protein arginine methyltransferases: nuclear receptor coregulators and beyond. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2009; 87:299-342. [PMID: 20374708 DOI: 10.1016/s1877-1173(09)87009-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Protein arginine methyltransferases (PRMTs) are a family of enzymes that play a crucial role in diverse cellular functions. Several PRMTs have been associated with gene expression regulation, in which PRMTs act as histone methyltransferases, secondary coregulators of transcription, or facilitate mRNA splicing and stability. Additional functions include modulation of protein localization, ribosomal assembly, and signal transduction. At the organismal level, several PRMTs appear to be important for development and may play an important role in cancer. The relationships between their cellular and organismal functions are poorly understood; at least in part due to the large body of enzymatic substrates for PRMTs and their transcriptional targets that remain to be determined. Specific PRMT inhibitors have been developed in recent years, which should help to shed light on their diverse biological roles. Connecting PRMT cellular functions with their global effects on an organism will facilitate development of novel treatments for human diseases.
Collapse
Affiliation(s)
- Peter Kuhn
- McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
81
|
Xu J, Wu RC, O’Malley BW. Normal and cancer-related functions of the p160 steroid receptor co-activator (SRC) family. Nat Rev Cancer 2009; 9:615-30. [PMID: 19701241 PMCID: PMC2908510 DOI: 10.1038/nrc2695] [Citation(s) in RCA: 388] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The three homologous members of the p160 SRC family (SRC1, SRC2 and SRC3) mediate the transcriptional functions of nuclear receptors and other transcription factors, and are the most studied of all the transcriptional co-activators. Recent work has indicated that the SRCgenes are subject to amplification and overexpression in various human cancers. Some of the molecular mechanisms responsible for SRC overexpression, along with the mechanisms by which SRCs promote breast and prostate cancer cell proliferation and survival, have been identified, as have the specific contributions of individual SRC family members to spontaneous breast and prostate carcinogenesis in genetically manipulated mouse models. These studies have identified new challenges for cancer research and therapy.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
- Luzhou Medical College, Luzhou, Sichuan 646000, China
| | - Ray-Chang Wu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| | - Bert W. O’Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030 USA
| |
Collapse
|
82
|
Jeong KW, Lee YH, Stallcup MR. Recruitment of the SWI/SNF chromatin remodeling complex to steroid hormone-regulated promoters by nuclear receptor coactivator flightless-I. J Biol Chem 2009; 284:29298-309. [PMID: 19720835 DOI: 10.1074/jbc.m109.037010] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
ATP-dependent chromatin remodeling complexes, such as SWI/SNF, are required for transcriptional activation of specific genes and are believed to be recruited to gene promoters by direct interaction with DNA binding transcription factors. However, we report here that recruitment of SWI/SNF to target genes of estrogen receptor alpha (ERalpha) requires the previously described nuclear receptor coactivator protein Flightless-I (Fli-I). Fli-I can bind directly to both ER and BAF53, an actin-related component of the SWI/SNF complex, suggesting that Fli-I may recruit SWI/SNF to ER target genes via interaction with BAF53. Point mutations in Fli-I that disrupt binding to ER or BAF53 compromised the ability of Fli-I to enhance ER-mediated activation of a transiently transfected reporter gene. Depletion of endogenous Fli-I or BAF53 inhibited estrogen-responsive expression of endogenous target genes of ER, indicating a critical role for Fli-I and BAF53. Moreover, depletion of endogenous Fli-I or BAF53 specifically eliminated part of the complex cyclical pattern of recruitment of SWI/SNF to estrogen-responsive promoters in a way that indicates multiple roles and multiple mechanisms of recruitment for SWI/SNF in estrogen-dependent target gene expression. These results begin to establish the functional relationships and interdependencies that coordinate the actions of the many coactivators participating in the transcriptional activation process.
Collapse
Affiliation(s)
- Kwang Won Jeong
- Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, USA
| | | | | |
Collapse
|
83
|
Adams D, Ruzehaji N, Strudwick X, Greenwood J, Campbell H, Arkell R, Cowin A. Attenuation of Flightless I, an actin-remodelling protein, improves burn injury repair via modulation of transforming growth factor (TGF)-β1 and TGF-β3. Br J Dermatol 2009; 161:326-36. [DOI: 10.1111/j.1365-2133.2009.09296.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
84
|
Zheng B, Han M, Bernier M, Wen JK. Nuclear actin and actin-binding proteins in the regulation of transcription and gene expression. FEBS J 2009; 276:2669-85. [PMID: 19459931 PMCID: PMC2978034 DOI: 10.1111/j.1742-4658.2009.06986.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Nuclear actin is involved in the transcription of all three RNA polymerases, in chromatin remodeling and in the formation of heterogeneous nuclear ribonucleoprotein complexes, as well as in recruitment of the histone modifier to the active gene. In addition, actin-binding proteins (ABPs) control actin nucleation, bundling, filament capping, fragmentation and monomer availability in the cytoplasm. In recent years, more and more attention has focused on the role of actin and ABPs in the modulation of the subcellular localization of transcriptional regulators. This review focuses on recent developments in the study of transcription and transcriptional regulation by nuclear actin, and the regulation of muscle-specific gene expression, nuclear receptor and transcription complexes by ABPs. Among the ABPs, striated muscle activator of Rho signaling and actin-binding LIM protein regulate actin dynamics and serum response factor-dependent muscle-specific gene expression. Functionally and structurally unrelated cytoplasmic ABPs interact cooperatively with nuclear receptor and regulate its transactivation. Furthermore, ABPs also participate in the formation of transcription complexes.
Collapse
Affiliation(s)
- Bin Zheng
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Michel Bernier
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Jin-kun Wen
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
85
|
Kopecki Z, Arkell R, Powell BC, Cowin AJ. Flightless I regulates hemidesmosome formation and integrin-mediated cellular adhesion and migration during wound repair. J Invest Dermatol 2009; 129:2031-45. [PMID: 19212345 DOI: 10.1038/jid.2008.461] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Flightless I (Flii), a highly conserved member of the gelsolin family of actin-remodelling proteins associates with actin structures and is involved in cellular motility and adhesion. Our previous studies have shown that Flii is an important negative regulator of wound repair. Here, we show that Flii affects hemidesmosome formation and integrin-mediated keratinocyte adhesion and migration. Impaired hemidesmosome formation and sparse arrangements of keratin cytoskeleton tonofilaments and actin cytoskeleton anchoring fibrils were observed in Flii(Tg/+) and Flii(Tg/Tg) mice with their skin being significantly more fragile than Flii(+/-) and WT mice. Flii(+/-) primary keratinocytes showed increased adhesion on laminin and collagen I than WT and Flii(Tg/Tg) primary keratinocytes. Decreased expression of CD151 and laminin-binding integrins alpha3, beta1, alpha6 and beta4 were observed in Flii overexpressing wounds, which could contribute to the impaired wound re-epithelialization observed in these mice. Flii interacts with proteins directly linked to the cytoplasmic domain of integrin receptors suggesting that it may be a mechanical link between ligand-bound integrin receptors and the actin cytoskeleton driving adhesion-signaling pathways. Therefore Flii may regulate wound repair through its effect on hemidesmosome formation and integrin-mediated cellular adhesion and migration.
Collapse
Affiliation(s)
- Zlatko Kopecki
- Women's and Children's Health Research Institute, North Adelaide, South Australia, Australia
| | | | | | | |
Collapse
|
86
|
Hofmann WA. Cell and molecular biology of nuclear actin. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2009; 273:219-63. [PMID: 19215906 DOI: 10.1016/s1937-6448(08)01806-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Actin is a highly conserved protein and one of the major components of the cytoplasm and the nucleus in eukaryotic cells. In the nucleus, actin is involved in a variety of nuclear processes that include transcription and transcription regulation, RNA processing and export, intranuclear movement, and structure maintenance. Recent advances in the field of nuclear actin have established that functions of actin in the nucleus are versatile, complex, and interconnected. It also has become increasingly evident that the cytoplasmic and nuclear pools of actin are functionally linked. However, while the biological significance of nuclear actin has become clear, we are only beginning to understand the mechanisms that lie behind the regulation of nuclear actin. This review provides an overview of our current understanding of the functions of actin in the nucleus.
Collapse
Affiliation(s)
- Wilma A Hofmann
- Department of Physiology and Biophysics, State University of New York, Buffalo, NY, USA
| |
Collapse
|
87
|
Liang J, Zhang H, Zhang Y, Zhang Y, Shang Y. GAS, a new glutamate-rich protein, interacts differentially with SRCs and is involved in oestrogen receptor function. EMBO Rep 2008; 10:51-7. [PMID: 19039327 DOI: 10.1038/embor.2008.223] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 11/03/2008] [Accepted: 11/04/2008] [Indexed: 01/07/2023] Open
Abstract
Steroid receptor coactivators (SRCs) exert profound effects on animal development and physiology. Genetic ablation experiments indicate that various SRC proteins might have differential physiological roles; however, clear evidence of functional specificity has not yet been shown at the molecular level. Here we report the identification of a new SRC1 interacting protein, glutamate-rich coactivator interacting with SRC1 (GAS), which contains a central glutamate-rich region and has transactivation activity. Interestingly, GAS interacts only with SRC1, and not with glucocorticoid receptor interacting protein 1 (GRIP1) or amplified in breast cancer 1 (AIB1), the other two members of the SRC family. It interacts with oestrogen receptor-alpha (ERalpha) and participates in both oestrogen receptor-regulated gene transcription and oestrogen-stimulated G1/S cell-cycle transition. Our data thus indicate that GAS is a new transcription cofactor and that different SRCs are associated with distinct secondary cofactors.
Collapse
Affiliation(s)
- Jing Liang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education,Peking University Health Science Center, Beijing, China
| | | | | | | | | |
Collapse
|
88
|
Seward ME, Easley CA, McLeod JJ, Myers AL, Tombes RM. Flightless-I, a gelsolin family member and transcriptional regulator, preferentially binds directly to activated cytosolic CaMK-II. FEBS Lett 2008; 582:2489-95. [DOI: 10.1016/j.febslet.2008.06.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 05/29/2008] [Accepted: 06/05/2008] [Indexed: 11/28/2022]
|
89
|
FLI-1 Flightless-1 and LET-60 Ras control germ line morphogenesis in C. elegans. BMC DEVELOPMENTAL BIOLOGY 2008; 8:54. [PMID: 18485202 PMCID: PMC2396608 DOI: 10.1186/1471-213x-8-54] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Accepted: 05/16/2008] [Indexed: 11/12/2022]
Abstract
Background In the C. elegans germ line, syncytial germ line nuclei are arranged at the cortex of the germ line as they exit mitosis and enter meiosis, forming a nucleus-free core of germ line cytoplasm called the rachis. Molecular mechanisms of rachis formation and germ line organization are not well understood. Results Mutations in the fli-1 gene disrupt rachis organization without affecting meiotic differentiation, a phenotype in C. elegans referred to here as the germ line morphogenesis (Glm) phenotype. In fli-1 mutants, chains of meiotic germ nuclei spanned the rachis and were partially enveloped by invaginations of germ line plasma membrane, similar to nuclei at the cortex. Extensions of the somatic sheath cells that surround the germ line protruded deep inside the rachis and were associated with displaced nuclei in fli-1 mutants. fli-1 encodes a molecule with leucine-rich repeats and gelsolin repeats similar to Drosophila flightless 1 and human Fliih, which have been shown to act as cytoplasmic actin regulators as well as nuclear transcriptional regulators. Mutations in let-60 Ras, previously implicated in germ line development, were found to cause the Glm phenotype. Constitutively-active LET-60 partially rescued the fli-1 Glm phenotype, suggesting that LET-60 Ras and FLI-1 might act together to control germ line morphogenesis. Conclusion FLI-1 controls germ line morphogenesis and rachis organization, a process about which little is known at the molecular level. The LET-60 Ras GTPase might act with FLI-1 to control germ line morphogenesis.
Collapse
|
90
|
Collins RE, Northrop JP, Horton JR, Lee DY, Zhang X, Stallcup MR, Cheng X. The ankyrin repeats of G9a and GLP histone methyltransferases are mono- and dimethyllysine binding modules. Nat Struct Mol Biol 2008; 15:245-50. [PMID: 18264113 PMCID: PMC2586904 DOI: 10.1038/nsmb.1384] [Citation(s) in RCA: 215] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Accepted: 01/08/2008] [Indexed: 01/17/2023]
Abstract
Histone modifications have important roles in transcriptional control, mitosis and heterochromatin formation. G9a and G9a-like protein (GLP) are euchromatin-associated methyltransferases that repress transcription by mono- and dimethylating histone H3 at Lys9 (H3K9). Here we demonstrate that the ankyrin repeat domains of G9a and GLP bind with strong preference to N-terminal H3 peptides containing mono- or dimethyl K9. X-ray crystallography revealed the basis for recognition of the methylated lysine by a partial hydrophobic cage with three tryptophans and one acidic residue. Substitution of key residues in the cage eliminated the H3 tail interaction. Hence, G9a and GLP contain a new type of methyllysine binding module (the ankyrin repeat domains) and are the first examples of protein (histone) methyltransferases harboring in a single polypeptide the activities that generate and read the same epigenetic mark.
Collapse
Affiliation(s)
- Robert E Collins
- Department of Biochemistry, Emory University School of Medicine, 1510 Clifton Road, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
91
|
Chinenov Y, Rogatsky I. Glucocorticoids and the innate immune system: crosstalk with the toll-like receptor signaling network. Mol Cell Endocrinol 2007; 275:30-42. [PMID: 17576036 DOI: 10.1016/j.mce.2007.04.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2007] [Accepted: 04/28/2007] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) are responsible for the recognition of a variety of microbial pathogens and the initial induction of immune and inflammatory responses. These responses are normally restricted by the adrenally produced glucocorticoid hormones which provide a feedback mechanism to curb unabated inflammation. Glucocorticoids act through a ligand-dependent transcription factor-the glucocorticoid receptor (GR), which engages in a complex network of protein:protein and protein:DNA interactions ultimately activating or repressing target gene transcription. Not surprisingly, multiple mechanisms account for the glucocorticoid interference with TLR signaling including enhanced expression of the natural inhibitors of TLR pathways, direct repression of TLR-activated transcriptional regulators and cross-utilization of cofactors essential for both GR and TLR signaling. Here we discuss recent and unexpected examples of crosstalk between the two transcriptional networks and the emerging role of GR in the regulation of innate immunity.
Collapse
Affiliation(s)
- Yurii Chinenov
- Hospital for Special Surgery, Department of Microbiology & Immunology, Weill Medical College of Cornell University, 535 E70th Street, Research Building Room 425, New York, NY 10021,USA
| | | |
Collapse
|
92
|
Cowin AJ, Adams DH, Strudwick XL, Chan H, Hooper JA, Sander GR, Rayner TE, Matthaei KI, Powell BC, Campbell HD. Flightless I deficiency enhances wound repair by increasing cell migration and proliferation. J Pathol 2007; 211:572-581. [PMID: 17326236 DOI: 10.1002/path.2143] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Wound healing disorders are a therapeutic problem of increasing clinical importance involving substantial morbidity, mortality, and rising health costs. Our studies investigating flightless I (FliI), a highly conserved actin-remodelling protein, now reveal that FliI is an important regulator of wound repair whose manipulation may lead to enhanced wound outcomes. We demonstrate that FliI-deficient + /- mice are characterized by improved wound healing with increased epithelial migration and enhanced wound contraction. In contrast, FliI-overexpressing mice have significantly impaired wound healing with larger less contracted wounds and reduced cellular proliferation. We show that FliI is secreted in response to wounding and that topical application of antibodies raised against the leucine-rich repeat domain of the FliI protein (FliL) significantly improves wound repair. These studies reveal that FliI affects wound repair via mechanisms involving cell migration and proliferation and that FliI might represent an effective novel therapeutic factor to improve conditions in which wound healing is impaired.
Collapse
Affiliation(s)
- A J Cowin
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
- Department of Paediatrics, University of Adelaide, South Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Australia
| | - D H Adams
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
| | - X L Strudwick
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
| | - H Chan
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
| | - J A Hooper
- Molecular Genetics and Evolution Group and Centre for the Molecular Genetics of Development, Research School of Biological Sciences, Australian National University, Canberra, ACT, Australia
| | - G R Sander
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
- Department of Paediatrics, University of Adelaide, South Australia
| | - T E Rayner
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
| | - K I Matthaei
- Division of Molecular Bioscience, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - B C Powell
- Child Health Research Institute, 72 King William Road, North Adelaide, South Australia
- Department of Paediatrics, University of Adelaide, South Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Australia
| | - H D Campbell
- Molecular Genetics and Evolution Group and Centre for the Molecular Genetics of Development, Research School of Biological Sciences, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
93
|
Li S, Shang Y. Regulation of SRC family coactivators by post-translational modifications. Cell Signal 2007; 19:1101-12. [PMID: 17368849 DOI: 10.1016/j.cellsig.2007.02.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2007] [Revised: 02/01/2007] [Accepted: 02/01/2007] [Indexed: 02/05/2023]
Abstract
Initially identified as a group of auxiliary protein factors involved in transcriptional regulation by steroid hormone receptors as well as by other members of the nuclear receptor superfamily, the steroid receptor coactivators (SRCs) have since then been implicated in the transcriptional regulation of other transcription factors which are important components of very different signaling pathways. Members of the SRC family have been shown to interact with myogenin, MEF-2, transcriptional enhancer factor (TEF), NF-kappaB, AP-1, STAT, p53, and E2F1, suggesting that SRC coactivators participate in diverse cellular processes. Recent evidence indicates that various post-translational modifications play critical roles in determining the final transcriptional output and specificity of SRC coactivators. In this review, we summarized the current knowledge concerning post-translational modifications, dynamic interplay between different modifications, and patho-physiological relevance of the modifications of SRC proteins.
Collapse
Affiliation(s)
- Shaosi Li
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center, 38 Xue Yuan Road, Beijing 100083, PR China
| | | |
Collapse
|
94
|
Ono S. Mechanism of depolymerization and severing of actin filaments and its significance in cytoskeletal dynamics. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 258:1-82. [PMID: 17338919 DOI: 10.1016/s0074-7696(07)58001-0] [Citation(s) in RCA: 212] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The actin cytoskeleton is one of the major structural components of the cell. It often undergoes rapid reorganization and plays crucial roles in a number of dynamic cellular processes, including cell migration, cytokinesis, membrane trafficking, and morphogenesis. Actin monomers are polymerized into filaments under physiological conditions, but spontaneous depolymerization is too slow to maintain the fast actin filament dynamics observed in vivo. Gelsolin, actin-depolymerizing factor (ADF)/cofilin, and several other actin-severing/depolymerizing proteins can enhance disassembly of actin filaments and promote reorganization of the actin cytoskeleton. This review presents advances as well as a historical overview of studies on the biochemical activities and cellular functions of actin-severing/depolymerizing proteins.
Collapse
Affiliation(s)
- Shoichiro Ono
- Department of Pathology, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
95
|
Germain P, Staels B, Dacquet C, Spedding M, Laudet V. Overview of nomenclature of nuclear receptors. Pharmacol Rev 2006; 58:685-704. [PMID: 17132848 DOI: 10.1124/pr.58.4.2] [Citation(s) in RCA: 469] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Nuclear receptor pharmacology has, to a certain extent, led the way, compared with other receptor systems, in the appreciation that ligands may exert very diverse pharmacology, based on their individual chemical structure and the allosteric changes induced in the receptor/accessory protein complex. This can lead to very selective pharmacological effects, which may not necessarily be predicted from the experience with other agonists/partial agonists/antagonists. If this is the case, then drug discovery may be back to drug-specific pharmacology (where each drug may have an original profile), rather than specific-drug pharmacology (where agents specific for a receptor have a distinct profile). As functional selectivity is indeed a crucial mechanism to be considered when going through the drug discovery development process, then initial screens using reconstituted systems may not show the appropriate pharmacology, simply because the required stoichiometry of corepressors and coactivators may not be present to select the best compounds; therefore, multiple effector systems are necessary to screen for differential activation, and, even then, screening with in vivo pathophysiological models may ultimately be required for the selection process-a massive but necessary task for pharmacologists. Thus, the characterization of nuclear receptors and their associated proteins and the ligands that interact with them will remain a challenge to pharmacologists.
Collapse
Affiliation(s)
- Pierre Germain
- Department of Cell Biology and Signal Transduction, Institut de Genetique et de Biologie Moleculaire et Cellulaire (IGBMC), 1 rue Laurent Fries, BP 10142, 67404 Illkirch Cedex, France.
| | | | | | | | | |
Collapse
|
96
|
Lee YH, Stallcup MR. Interplay of Fli-I and FLAP1 for regulation of beta-catenin dependent transcription. Nucleic Acids Res 2006; 34:5052-9. [PMID: 16990252 PMCID: PMC1636430 DOI: 10.1093/nar/gkl652] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
β-catenin mediates Wnt/wingless signaling and transcriptional activation by lymphocyte enhancer binding factor 1/T cell factor (LEF1/TCF) proteins with the assistance of multiple coregulators, including positive cofactors like p300/CBP and negative cofactors like HDACs. We previously demonstrated that a developmentally essential protein, Flightless-I (Fli-I), serves as a coactivator for nuclear receptor-mediated transcription. To further understand the action mechanism of Fli-I, we investigated the functional roles of Fli-I and Fli-I leucine rich repeat associated protein 1 (FLAP1) in transcriptional activation by β-catenin and LEF1/TCF. β-catenin-dependent transcription was activated by exogenous FLAP1 but inhibited by Fli-I. Reduction of endogenous FLAP1 levels compromised transcriptional activation by LEF1/TCF, β-catenin and the p160 coactivator GRIP1. FLAP1 interacted directly with β-catenin, GRIP1 and p300 and enhanced their activity. Furthermore, FLAP1 was strongly synergistic with p300 in supporting transcriptional activation by β-catenin and LEF1/TCF, but Fli-I disrupted the synergy of FLAP1 with p300 and β-catenin. Thus the opposing effects of Fli-I and FLAP1 may be a key regulatory mechanism for β-catenin and LEF1/TCF-mediated transcription and thus for Wnt signaling, and some mutations of Fli-I may result in developmental defects, such as the flightless phenotype of Drosophila, by causing dysregulation of the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
| | - Michael R. Stallcup
- To whom correspondence should be addressed. Tel: +1 323 442 1145; Fax: +1 323 442 1224;
| |
Collapse
|
97
|
Kim JH, Yang CK, Stallcup MR. Downstream signaling mechanism of the C-terminal activation domain of transcriptional coactivator CoCoA. Nucleic Acids Res 2006; 34:2736-50. [PMID: 16717280 PMCID: PMC1464418 DOI: 10.1093/nar/gkl361] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/08/2006] [Accepted: 04/24/2006] [Indexed: 12/03/2022] Open
Abstract
The coiled-coil coactivator (CoCoA) is a transcriptional coactivator for nuclear receptors and enhances nuclear receptor function by the interaction with the bHLH-PAS domain (AD3) of p160 coactivators. The C-terminal activation domain (AD) of CoCoA possesses strong transactivation activity and is required for the coactivator function of CoCoA with nuclear receptors. To understand how CoCoA AD transmits its activating signal to the transcription machinery, we defined specific subregions, amino acid motifs and protein binding partners involved in the function of CoCoA AD. The minimal transcriptional AD was mapped to approximately 91 C-terminal amino acids and consists of acidic, serine/proline-rich and phenylalanine-rich subdomains. Transcriptional activation by the CoCoA AD was p300-dependent, and p300 interacted physically and functionally with CoCoA AD and was recruited to a promoter by the interaction with CoCoA AD. The FYDVASAF motif in the CoCoA AD was critical for the transcriptional activity of CoCoA AD, the interaction of CoCoA with p300, the coactivator function of CoCoA for estrogen receptor alpha and GRIP1 and the transcriptional synergy among coactivators GRIP1, CARM1, p300 and CoCoA. Taken together these data extend our understanding of the mechanism of downstream signaling by the essential C-terminal AD of the nuclear receptor coactivator CoCoA; they indicate that p300 is a functionally important interaction partner of CoCoA AD and that their interaction potentiates transcriptional activation by the p160 coactivator complex.
Collapse
Affiliation(s)
- Jeong Hoon Kim
- Department of Biochemistry and Molecular Biology, University of Southern California Los Angeles, CA 90089, USA.
| | | | | |
Collapse
|
98
|
van de Wijngaart DJ, van Royen ME, Hersmus R, Pike ACW, Houtsmuller AB, Jenster G, Trapman J, Dubbink HJ. Novel FXXFF and FXXMF motifs in androgen receptor cofactors mediate high affinity and specific interactions with the ligand-binding domain. J Biol Chem 2006; 281:19407-16. [PMID: 16690616 DOI: 10.1074/jbc.m602567200] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Upon hormone binding, a hydrophobic coactivator binding groove is induced in the androgen receptor (AR) ligand-binding domain (LBD). This groove serves as high affinity docking site for alpha-helical FXXLF motifs present in the AR N-terminal domain and in AR cofactors. Study of the amino acid requirements at position +4 of the AR FXXLF motif revealed that most amino acid substitutions strongly reduced or completely abrogated AR LBD interaction. Strong interactions were still observed following substitution of Leu+4 by Phe or Met residues. Leu+4 to Met or Phe substitutions in the FXXLF motifs of AR cofactors ARA54 and ARA70 were also compatible with strong AR LBD binding. Like the corresponding FXXLF motifs, interactions of FXXFF and FXXMF variants of AR and ARA54 motifs were AR specific, whereas variants of the less AR-selective ARA70 motif displayed increased AR specificity. A survey of currently known AR-binding proteins revealed the presence of an FXXFF motif in gelsolin and an FXXMF motif in PAK6. In vivo fluorescence resonance energy transfer and functional protein-protein interaction assays showed direct, efficient, and specific interactions of both motifs with AR LBD. Mutation of these motifs abrogated interaction of gelsolin and PAK6 proteins with AR. In conclusion, we have demonstrated strong interaction of FXXFF and FXXMF motifs to the AR coactivator binding groove, thereby mediating specific binding of a subgroup of cofactors to the AR LBD.
Collapse
Affiliation(s)
- Dennis J van de Wijngaart
- Department of Urology, Josephine Nefkens Institute, Erasmus MC, P. O. Box 1738, 3000 DR Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Lee DY, Northrop JP, Kuo MH, Stallcup MR. Histone H3 lysine 9 methyltransferase G9a is a transcriptional coactivator for nuclear receptors. J Biol Chem 2006; 281:8476-85. [PMID: 16461774 PMCID: PMC1770944 DOI: 10.1074/jbc.m511093200] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Methylation of Lys-9 of histone H3 has been associated with repression of transcription. G9a is a histone H3 Lys-9 methyltransferase localized in euchromatin and acts as a corepressor for specific transcription factors. Here we demonstrate that G9a also functions as a coactivator for nuclear receptors, cooperating synergistically with nuclear receptor coactivators glucocorticoid receptor interacting protein 1, coactivator-associated arginine methyltransferase 1 (CARM1), and p300 in transient transfection assays. This synergy depends strongly on the arginine-specific protein methyltransferase activity of CARM1 but does not absolutely require the enzymatic activity of G9a and is specific to CARM1 and G9a among various protein methyltransferases. Reduction of endogenous G9a diminished hormonal activation of an endogenous target gene by the androgen receptor, and G9a associated with regulatory regions of this same gene. G9a fused to Gal4 DNA binding domain can repress transcription in a lysine methyltransferase-dependent manner; however, the histone modifications associated with transcriptional activation can inhibit the methyltransferase activity of G9a. These findings suggest a link between histone arginine and lysine methylation and a mechanism for controlling whether G9a functions as a corepressor or coactivator.
Collapse
Affiliation(s)
- David Y. Lee
- From the Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA 90089, and the
| | - Jeffrey P. Northrop
- From the Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA 90089, and the
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824
| | - Michael R. Stallcup
- From the Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA 90089, and the
- To whom correspondence should be addressed. . Mailing address: Department of Biochemistry and Molecular Biology, University of Southern California, 1333 San Pablo Street, MCA-51A, Los Angeles, CA 90089-9151. Phone: (323) 442-1289; Fax: (323) 442-1224
| |
Collapse
|
100
|
von Schalburg KR, McCarthy SP, Rise ML, Hutson JC, Davidson WS, Koop BF. Expression of morphogenic genes in mature ovarian and testicular tissues: Potential stem-cell niche markers and patterning factors. Mol Reprod Dev 2006; 73:142-52. [PMID: 16224774 DOI: 10.1002/mrd.20359] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Morphogens are developmental regulators that modulate different tissue patterning, proliferation, differentiation, or remodeling processes in embryonic and adult tissues. Morphogens may also evoke specific regulatory programs in stem cells. Some of the morphogens involved in these processes have been characterized, while others remain unidentified. A microarray containing 3,557 salmonid cDNAs was used to compare the transcriptomes of rainbow trout precocious ovary at three different stages during second year (June, August, and October) with a reference (June normal ovary) transcriptome. During this study, we detected morphogen transcript hybridizations to salmonid elements and the study was enlarged to investigate these activities in various developmental stages of both ovary and testis. Genes from diverse development regulator families such as Anterior gradient-2, BMP, Epimorphin, Flightless, Frizzled, Notch, Tiarin, Twisted gastrulation, and Wnt were demonstrated to be expressed in the adult trout gonads. In mice or rats, expression of mammalian bmp-4, epimorphin, flightless, twisted gastrulation, and GW112 transcripts were localized to cell types isolated from the developed ovary and testis. Comparisons of salmonid and mammalian morphogens at the amino acid residue level show high similarities, suggesting functional conservation. This report provides evidence for local regulation by various morphogens and their potential to control distinct programs of gene expression in the gametes and their accessory cells during gametogenesis.
Collapse
|