Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Genes Dev ; 33(7-8): 388-402, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30808659

ABSTRACT

Tgif1 (thymine-guanine-interacting factor 1) and Tgif2 repress gene expression by binding directly to DNA or interacting with transforming growth factor (TGF) ß-responsive SMADs. Tgifs are essential for embryogenesis and may function in tumor progression. By analyzing both gain and loss of Tgif function in a well-established mouse model of intestinal cancer, we show that Tgifs promote adenoma growth in the context of mutant Apc (adenomatous polyposis coli). Despite the tumor-suppressive role of TGFß signaling, transcriptome profiling of colon tumors suggests minimal effect of Tgifs on the TGFß pathway. Instead, it appears that Tgifs, which are up-regulated in Apc mutant colon tumors, contribute to reprogramming metabolic gene expression. Integrating gene expression data from colon tumors with other gene expression and chromatin-binding data identifies a set of direct Tgif target genes encoding proteins involved in acetyl CoA and pyruvate metabolism. Analysis of both tumor and nontumor tissues indicates that these genes are targets of Tgif repression in multiple settings, suggesting that this is a core Tgif function. We propose that Tgifs play an important role in regulating basic energy metabolism in normal cells, and that this function of Tgifs is amplified in some cancers.


Subject(s)
Acetyl Coenzyme A/genetics , Adenoma , Gene Expression Regulation, Neoplastic/genetics , Homeodomain Proteins/metabolism , Intestinal Neoplasms , Repressor Proteins/metabolism , Adenoma/genetics , Adenoma/physiopathology , Adenomatous Polyposis Coli/genetics , Animals , Cells, Cultured , Disease Models, Animal , Energy Metabolism/genetics , HCT116 Cells , Humans , Intestinal Mucosa/physiopathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/physiopathology , Mice , Mice, Inbred C57BL
2.
PLoS Genet ; 14(5): e1007409, 2018 05.
Article in English | MEDLINE | ID: mdl-29782499

ABSTRACT

Although treatment options for localized prostate cancer (CaP) are initially effective, the five-year survival for metastatic CaP is below 30%. Mutation or deletion of the PTEN tumor suppressor is a frequent event in metastatic CaP, and inactivation of the transforming growth factor (TGF) ß signaling pathway is associated with more advanced disease. We previously demonstrated that mouse models of CaP based on inactivation of Pten and the TGFß type II receptor (Tgfbr2) rapidly become invasive and metastatic. Here we show that mouse prostate tumors lacking Pten and Tgfbr2 have higher expression of stem cell markers and genes indicative of basal epithelial cells, and that basal cell proliferation is increased compared to Pten mutants. To better model the primarily luminal phenotype of human CaP we mutated Pten and Tgfbr2 specifically in luminal cells, and found that these tumors also progress to invasive and metastatic cancer. Accompanying the transition to invasive cancer we observed de-differentiation of luminal tumor cells to an intermediate cell type with both basal and luminal markers, as well as differentiation to basal cells. Proliferation rates in these de-differentiated cells were lower than in either basal or luminal cells. However, de-differentiated cells account for the majority of cells in micro-metastases consistent with a preferential contribution to metastasis. We suggest that active TGFß signaling limits lineage plasticity in prostate luminal cells, and that de-differentiation of luminal tumor cells can drive progression to metastatic disease.


Subject(s)
Cell Lineage/genetics , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , Animals , Cell Proliferation/genetics , Disease Progression , Epithelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Survival Analysis , Transforming Growth Factor beta/metabolism
3.
Mol Cell ; 46(5): 556-7, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22681884

ABSTRACT

In this issue of Molecular Cell, Thillainadesan et al. (2012) provide evidence that Smad proteins promote locus-specific active DNA demethylation as part of the transforming growth factor ß (TGF-ß) transcriptional response.

4.
J Cell Biochem ; 119(6): 4644-4655, 2018 06.
Article in English | MEDLINE | ID: mdl-29291346

ABSTRACT

Myt1 and Myt1l (Myelin transcription factor 1, and Myt1-like) are members of a small family of closely related zinc finger transcription factors, characterized by two clusters of C2HC zinc fingers. Both are widely expressed during early embryogenesis, but are largely restricted to expression within the brain in the adult. Myt1l, as part of a three transcription factor mix, can reprogram fibroblasts to neurons and plays a role in maintaining neuronal identity. Previous analyses have indicated roles in both transcriptional activation and repression and suggested that Myt1 and Myt1l may have opposing functions in gene expression. We show that when targeted to DNA via multiple copies of the consensus Myt1/Myt1l binding site Myt1 represses transcription, whereas Myt1l activates. By targeting via a heterologous DNA binding domain we mapped an activation function in Myt1l to an amino-terminal region that is poorly conserved in Myt1. However, genome wide analyses of the effects of Myt1 and Myt1l expression in a glioblastoma cell line suggest that the two proteins have largely similar effects on endogenous gene expression. Transcriptional repression is likely mediated by binding to DNA via the known consensus site, whereas this site is not associated with the transcriptional start sites of genes with higher expression in the presence of Myt1 or Myt1l. This work suggests that these two proteins function similarly, despite differences observed in analyses based on synthetic reporter constructs.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Response Elements , Transcription Factors/metabolism , Transcription, Genetic , A549 Cells , DNA-Binding Proteins/genetics , Glioblastoma/genetics , Glioblastoma/pathology , HEK293 Cells , HeLa Cells , Humans , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Transcription Factors/genetics
5.
Am J Med Genet C Semin Med Genet ; 178(2): 128-139, 2018 06.
Article in English | MEDLINE | ID: mdl-29749689

ABSTRACT

Holoprosencephaly (HPE) is a frequent human forebrain developmental disorder with both genetic and environmental causes. Multiple loci have been associated with HPE in humans, and potential causative genes at 14 of these loci have been identified. Although TGIF1 (originally TGIF, for Thymine Guanine-Interacting Factor) is among the most frequently screened genes in HPE patients, an understanding of how mutations in this gene contribute to the pathogenesis of HPE has remained elusive. However, mouse models based on loss of function of Tgif1, and the related Tgif2 gene, have shed some light on how human TGIF1 variants might cause HPE. Functional analyses of TGIF proteins and of TGIF1 single nucleotide variants from HPE patients, combined with analysis of forebrain development in mouse embryos lacking both Tgif1 and Tgif2, suggest that TGIFs regulate the transforming growth factor ß/Nodal signaling pathway and sonic hedgehog (SHH) signaling independently. Although, some developmental processes that are regulated by TGIFs may be Nodal-dependent, it appears that the forebrain patterning defects and HPE in Tgif mutant mouse embryos is primarily due to altered signaling via the Shh pathway.


Subject(s)
Brain/embryology , Brain/metabolism , Disease Susceptibility , Holoprosencephaly/etiology , Holoprosencephaly/metabolism , Homeodomain Proteins/genetics , Organogenesis/genetics , Repressor Proteins/genetics , Animals , Body Patterning , Brain/abnormalities , Brain/ultrastructure , Craniofacial Abnormalities/etiology , Craniofacial Abnormalities/metabolism , Developmental Disabilities/etiology , Developmental Disabilities/metabolism , Disease Models, Animal , Gene Deletion , Gene Expression Regulation , Genetic Variation , Homeodomain Proteins/metabolism , Humans , Mice , Nodal Protein/genetics , Nodal Protein/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Repressor Proteins/metabolism , Signal Transduction , Transcription, Genetic
6.
Gynecol Oncol ; 149(2): 350-360, 2018 05.
Article in English | MEDLINE | ID: mdl-29551565

ABSTRACT

OBJECTIVE: Ovarian cancer survival and treatment have improved minimally in the past 20years. Novel treatment strategies are needed to combat this disease. This study investigates the effects of chemical inhibition of the CBFß/RUNX protein-protein interaction on ovarian cancer cell lines. METHODS: Ovarian cancer cell lines were treated with CBFß/RUNX inhibitors, and the effects on proliferation, DNA replication, wound healing, and anchorage-independent growth were measured. RNA-Seq was performed on compound-treated cells to identify differentially expressed genes. Genes altered by compound treatment were targeted with siRNAs, and effects on DNA replication and wound healing were measured. RESULTS: Chemical inhibition of the CBFß/RUNX interaction decreases ovarian cancer cell proliferation. Inhibitor treatment leads to an S-phase cell cycle delay, as indicated by an increased percentage of cells in S-phase, and a decreased DNA replication rate. Inhibitor treatment also reduces wound healing and anchorage-independent growth. RNA-Seq on compound-treated cells revealed changes in a small number of genes related to proliferation and epithelial-to-mesenchymal transition. siRNA-mediated knockdown of INHBA and MMP1 - two genes whose expression decreases with compound treatment - slowed DNA replication and impaired wound healing. CONCLUSIONS: Chemical inhibition of the CBFß/RUNX interaction is a viable strategy for the treatment of ovarian cancer.


Subject(s)
Core Binding Factor alpha Subunits/antagonists & inhibitors , Epithelial-Mesenchymal Transition/genetics , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Small Molecule Libraries/pharmacology , Animals , Carcinoma, Ovarian Epithelial , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Core Binding Factor alpha Subunits/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/metabolism , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Random Allocation , Xenograft Model Antitumor Assays
7.
Prostate ; 77(15): 1452-1467, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28875501

ABSTRACT

BACKGROUND: Phosphoinositide-3 (PI-3) kinase signaling has a pervasive role in cancer. One of the key effectors of PI-3 kinase signaling is AKT, a kinase that promotes growth and survival in a variety of cancers. Genetically engineered mouse models of prostate cancer have shown that AKT signaling is sufficient to induce prostatic epithelial neoplasia (PIN), but insufficient for progression to adenocarcinoma. This contrasts with the phenotype of mice with prostate-specific deletion of Pten, where excessive PI-3 kinase signaling induces both PIN and locally invasive carcinoma. We reasoned that additional PI-3 kinase effector kinases promote prostate cancer progression via activities that provide biological complementarity to AKT. We focused on the PKN kinase family members, which undergo activation in response to PI-3 kinase signaling, show expression changes in prostate cancer, and contribute to cell motility pathways in cancer cells. METHODS: PKN kinase activity was measured by incorporation of 32 P into protein substrates. Phosphorylation of the turn-motif (TM) in PKN proteins by mTOR was analyzed using the TORC2-specific inhibitor torin and a PKN1 phospho-TM-specific antibody. Amino acid substitutions in the TM of PKN were engineered and assayed for effects on kinase activity. Cell motility-related functions and PKN localization was analyzed by depletion approaches and immunofluorescence microscopy, respectively. The contribution of PKN proteins to prostate tumorigenesis was characterized in several mouse models that express PKN transgenes. The requirement for PKN activity in prostate cancer initiated by loss of phosphatase and tensin homolog deleted on chromosome 10 (Pten), and the potential redundancy between PKN isoforms, was analyzed by prostate-specific deletion of Pkn1, Pkn2, and Pten. RESULTS AND CONCLUSIONS: PKN1 and PKN2 contribute to motility pathways in human prostate cancer cells. PKN1 and PKN2 kinase activity is regulated by TORC2-dependent phosphorylation of the TM, which together with published data indicates that PKN proteins receive multiple PI-3 kinase-dependent inputs. Transgenic expression of active AKT and PKN1 is not sufficient for progression beyond PIN. Moreover, Pkn1 is not required for tumorigenesis initiated by loss of Pten. Triple knockout of Pten, Pkn1, and Pkn2 in mouse prostate results in squamous cell carcinoma, an uncommon but therapy-resistant form of prostate cancer.


Subject(s)
Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase C/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/physiology , Disease Progression , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/genetics , Protein Kinase C/genetics , TOR Serine-Threonine Kinases/genetics
8.
Adv Exp Med Biol ; 963: 35-50, 2017.
Article in English | MEDLINE | ID: mdl-28197905

ABSTRACT

Many of the known SUMO substrates are nuclear proteins, which regulate gene expression and chromatin dynamics. Sumoylation, in general, appears to correlate with decreased transcriptional activity, and in many cases modulation of the chromatin template is implicated. Sumoylation of the core histones is associated with transcriptional silencing, and transcription factor sumoylation can decrease gene expression by promoting recruitment of chromatin modifying enzymes. Additionally, sumoylation of transcriptional corepressors and chromatin remodeling enzymes can influence interactions with other transcriptional regulators, and alter their enzymatic activity. In some cases, proteins that are components of transcriptional corepressor complexes have been shown to be SUMO E3 ligases, further emphasizing the integration of sumoylation with the regulation of chromatin remodeling. Despite the evidence suggesting that sumoylation is primarily repressive for access to chromatin, recent analyses suggest that protein sumoylation on the chromatin template may play important roles at highly expressed genes. Elucidating the dynamic interplay of sumoylation with other post-translational modifications of histones and chromatin associated proteins will be key to fully understanding the regulation of access to the chromatin template.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA/metabolism , Histones/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Sumoylation , Animals , Chromatin/chemistry , DNA/chemistry , Histones/chemistry , Humans , Nucleic Acid Conformation , Protein Conformation , Structure-Activity Relationship , Transcription, Genetic , Ubiquitin-Protein Ligases/metabolism
9.
PLoS Genet ; 8(2): e1002524, 2012.
Article in English | MEDLINE | ID: mdl-22383895

ABSTRACT

Holoprosencephaly (HPE) is a severe human genetic disease affecting craniofacial development, with an incidence of up to 1/250 human conceptions and 1.3 per 10,000 live births. Mutations in the Sonic Hedgehog (SHH) gene result in HPE in humans and mice, and the Shh pathway is targeted by other mutations that cause HPE. However, at least 12 loci are associated with HPE in humans, suggesting that defects in other pathways contribute to this disease. Although the TGIF1 (TG-interacting factor) gene maps to the HPE4 locus, and heterozygous loss of function TGIF1 mutations are associated with HPE, mouse models have not yet explained how loss of Tgif1 causes HPE. Using a conditional Tgif1 allele, we show that mouse embryos lacking both Tgif1 and the related Tgif2 have HPE-like phenotypes reminiscent of Shh null embryos. Eye and nasal field separation is defective, and forebrain patterning is disrupted in embryos lacking both Tgifs. Early anterior patterning is relatively normal, but expression of Shh is reduced in the forebrain, and Gli3 expression is up-regulated throughout the neural tube. Gli3 acts primarily as an antagonist of Shh function, and the introduction of a heterozygous Gli3 mutation into embryos lacking both Tgif genes partially rescues Shh signaling, nasal field separation, and HPE. Tgif1 and Tgif2 are transcriptional repressors that limit Transforming Growth Factor ß/Nodal signaling, and we show that reducing Nodal signaling in embryos lacking both Tgifs reduces the severity of HPE and partially restores the output of Shh signaling. Together, these results support a model in which Tgif function limits Nodal signaling to maintain the appropriate output of the Shh pathway in the forebrain. These data show for the first time that Tgif1 mutation in mouse contributes to HPE pathogenesis and provide evidence that this is due to disruption of the Shh pathway.


Subject(s)
Hedgehog Proteins/genetics , Holoprosencephaly/genetics , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Animals , Disease Models, Animal , Genes, Homeobox , Hedgehog Proteins/metabolism , Holoprosencephaly/etiology , Holoprosencephaly/metabolism , Humans , Mice , Mutation , Signal Transduction/genetics , TGF-beta Superfamily Proteins/metabolism
10.
J Lipid Res ; 55(4): 709-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478032

ABSTRACT

Acat2 [gene name: sterol O-acyltransferase 2 (SOAT2)] esterifies cholesterol in enterocytes and hepatocytes. This study aims to identify repressor elements in the human SOAT2 promoter and evaluate their in vivo relevance. We identified TG-interacting factor 1 (Tgif1) to function as an important repressor of SOAT2. Tgif1 could also block the induction of the SOAT2 promoter activity by hepatocyte nuclear factor 1α and 4α. Women have ∼ 30% higher hepatic TGIF1 mRNA compared with men. Depletion of Tgif1 in mice increased the hepatic Soat2 expression and resulted in higher hepatic lipid accumulation and plasma cholesterol levels. Tgif1 is a new player in human cholesterol metabolism.


Subject(s)
Gene Silencing , Homeodomain Proteins/physiology , Repressor Proteins/physiology , Sterol O-Acyltransferase/genetics , Animals , Binding Sites , Cell Line, Tumor , Enzyme Repression , Female , Gallstones/enzymology , Hepatocyte Nuclear Factor 1-alpha/physiology , Hepatocyte Nuclear Factor 4/physiology , Homeodomain Proteins/metabolism , Humans , Lipids/blood , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , Sex Characteristics , Sterol O-Acyltransferase/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Sterol O-Acyltransferase 2
11.
Development ; 137(2): 249-59, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20040491

ABSTRACT

Tgif1 and Tgif2 are transcriptional co-repressors that limit the response to TGFbeta signaling and play a role in regulating retinoic-acid-mediated gene expression. Mutations in human TGIF1 are associated with holoprosencephaly, but it is unclear whether this is a result of deregulation of TGFbeta/Nodal signaling, or of effects on other pathways. Surprisingly, mutation of Tgif1 in mice results in only relatively mild developmental phenotypes in most strain backgrounds. Here, we show that loss-of-function mutations in both Tgif1 and Tgif2 result in a failure of gastrulation. By conditionally deleting Tgif1 in the epiblast, we demonstrate that a single wild-type allele of Tgif1 in the extra-embryonic tissue allows the double null embryos to gastrulate and begin organogenesis, suggesting that extra-embryonic Tgif function is required for patterning the epiblast. Genetically reducing the dose of Nodal in embryos lacking all Tgif function results in partial rescue of the gastrulation defects. Conditional double null embryos have defects in left-right asymmetry, which are also alleviated by reducing the dose of Nodal. Together, these data show that Tgif function is required for gastrulation, and provide the first clear evidence that Tgifs limit the transcriptional response to Nodal signaling during early embryogenesis.


Subject(s)
Gastrulation/physiology , Homeodomain Proteins/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Embryo, Mammalian/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , In Situ Hybridization , Mice , Mice, Mutant Strains
12.
Cancer Res Commun ; 3(4): 592-606, 2023 04.
Article in English | MEDLINE | ID: mdl-37077937

ABSTRACT

The ADP-ribosyltransferase PARP7 modulates protein function by conjugating ADP-ribose to the side chains of acceptor amino acids. PARP7 has been shown to affect gene expression in prostate cancer cells and certain other cell types by mechanisms that include transcription factor ADP-ribosylation. Here, we use a recently developed catalytic inhibitor to PARP7, RBN2397, to study the effects of PARP7 inhibition in androgen receptor (AR)-positive and AR-negative prostate cancer cells. We find that RBN2397 has nanomolar potency for inhibiting androgen-induced ADP-ribosylation of the AR. RBN2397 inhibits the growth of prostate cancer cells in culture when cells are treated with ligands that activate the AR, or the aryl hydrocarbon receptor, and induce PARP7 expression. We show that the growth-inhibitory effects of RBN2397 are distinct from its enhancement of IFN signaling recently shown to promote tumor immunogenicity. RBN2397 treatment also induces trapping of PARP7 in a detergent-resistant fraction within the nucleus, which is reminiscent of how inhibitors such as talazoparib affect PARP1 compartmentalization. Because PARP7 is expressed in AR-negative metastatic tumors and RBN2397 can affect cancer cells through multiple mechanisms, PARP7 may be an actionable target in advanced prostate cancer. Significance: RBN2397 is a potent and selective inhibitor of PARP7 that reduces the growth of prostate cancer cells, including a model for treatment-emergent neuroendocrine prostate cancer. RBN2397 induces PARP7 trapping on chromatin, suggesting its mechanism of action might be similar to clinically used PARP1 inhibitors.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Male , Humans , Receptors, Androgen/genetics , Prostatic Neoplasms/drug therapy , Prostate/metabolism , ADP Ribose Transferases/genetics , Androgens
13.
Cells ; 10(2)2021 02 09.
Article in English | MEDLINE | ID: mdl-33572475

ABSTRACT

Poly-ADP-ribose polymerases (PARPs) are enzymes that catalyze ADP-ribosylation and play critical roles in normal and disease settings. The PARP family member, PARP7, is a mono-ADP-ribosyltransferase that has been suggested to play a tumor suppressive role in breast, ovarian, and colorectal cancer. Here, we have investigated how androgen signaling regulates PARP7 homeostasis in prostate cancer cells, where PARP7 is a direct target gene of AR. We found that the PARP7 protein is extremely short-lived, with a half-life of 4.5 min. We show that in addition to its transcriptional regulation by AR, PARP7 is subject to androgen-dependent post-transcriptional regulation that increases its half-life to 25.6 min. This contrasts with PARP1, PARP2, PARP9, and PARP14, which do not display rapid turnover and are not regulated by androgen signaling. Androgen- and AR-dependent stabilization of PARP7 leads to accumulation in the nucleus, which we suggest is a major site of action. Mutations in the catalytic domain, the Cys3His1 zinc finger, and WWE (tryptophan-tryptophan-glutamate) domains in PARP7 each reduce the degradation rate of PARP7, suggesting the overall structure of the protein is tuned for its rapid turnover. Our finding that PARP7 is regulated by AR signaling both transcriptionally and post-transcriptionally in prostate cancer cells suggests the dosage of PARP7 protein is subject to tight regulation.


Subject(s)
ADP Ribose Transferases/metabolism , Androgens/metabolism , Gene Expression Regulation , Nucleoside Transport Proteins/metabolism , Prostatic Neoplasms/enzymology , ADP Ribose Transferases/chemistry , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Male , Mice , Nucleoside Transport Proteins/genetics , Prostatic Neoplasms/pathology , Protein Domains , Protein Stability , Receptors, Androgen/metabolism , Signal Transduction , Transcription, Genetic
14.
Nat Commun ; 12(1): 2705, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976187

ABSTRACT

Androgen signaling through the androgen receptor (AR) directs gene expression in both normal and prostate cancer cells. Androgen regulates multiple aspects of the AR life cycle, including its localization and post-translational modification, but understanding how modifications are read and integrated with AR activity has been difficult. Here, we show that ADP-ribosylation regulates AR through a nuclear pathway mediated by Parp7. We show that Parp7 mono-ADP-ribosylates agonist-bound AR, and that ADP-ribosyl-cysteines within the N-terminal domain mediate recruitment of the E3 ligase Dtx3L/Parp9. Molecular recognition of ADP-ribosyl-cysteine is provided by tandem macrodomains in Parp9, and Dtx3L/Parp9 modulates expression of a subset of AR-regulated genes. Parp7, ADP-ribosylation of AR, and AR-Dtx3L/Parp9 complex assembly are inhibited by Olaparib, a compound used clinically to inhibit poly-ADP-ribosyltransferases Parp1/2. Our study reveals the components of an androgen signaling axis that uses a writer and reader of ADP-ribosylation to regulate protein-protein interactions and AR activity.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Prostatic Neoplasms/genetics , Protein Processing, Post-Translational , Receptors, Androgen/genetics , ADP-Ribosylation/drug effects , Adenocarcinoma , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Male , Metribolone/pharmacology , Neoplasm Proteins/metabolism , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Survival Analysis
15.
Genesis ; 48(9): 559-62, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20645310

ABSTRACT

We have previously generated a transgenic mouse strain (LSL-TßRI(CA)) containing a Cre-inducible constitutively active TGFß type I receptor (Bartholin et al., 2008, Genesis 46: 724-731). Transgene expression depends on the excision of a floxed-transcriptional STOP (LSL, Lox-STOP-Lox) located upstream the TßRI(CA) coding sequence. To evaluate the correct excision of the STOP signal in the presence of Cre-recombinase, we developed a rapid screening based on an original PCR genotyping strategy. More precisely, we designed a set of primers flanking the LSL containing region. The size of the amplified products will differ according to recombination status of the LSL-TßRI(CA) allele. Indeed, the size of the STOP containing PCR product is 1.93 kb, but is reduced to 0.35 kb when the STOP signal is removed after Cre-mediated recombination. We validated excision in several compartments, including pancreas, liver, T lymphocytes, and embryos using different Cre expressing transgenic mouse strains. This represents a simple and efficient way of monitoring the tissue specific recombination of the LSL-TßRI(CA) allele.


Subject(s)
Polymerase Chain Reaction/methods , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Recombination, Genetic/genetics , Transgenes/genetics , Animals , Codon, Terminator/genetics , Crosses, Genetic , DNA Primers/genetics , Female , Genotype , Integrases/genetics , Liver/metabolism , Mice , Mice, Transgenic , Pancreas/metabolism , Receptor, Transforming Growth Factor-beta Type I , SOXB1 Transcription Factors/genetics , T-Lymphocytes/metabolism
16.
J Cell Biochem ; 111(2): 380-90, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20506222

ABSTRACT

TG-interacting factor (Tgif1) represses gene expression by interaction with general corepressors, and can be recruited to target genes by transforming growth factor beta (TGFß) activated Smads, or by the retinoid X receptor (RXR). Here we show that Tgif1 interacts with the LXRα nuclear receptor and can repress transcription from a synthetic reporter activated by LXRα. In cultured cells reducing endogenous Tgif1 levels resulted in increased expression of LXRα target genes. To test the in vivo role of Tgif1, we analyzed LXRα-dependent gene expression in mice lacking Tgif1. In the livers of Tgif1 null mice, we observed significant derepression of the apolipoprotein genes, Apoa4 and Apoc2, suggesting that Tgif1 is an important in vivo regulator of apolipoprotein gene expression. In contrast, we observed relatively minimal effects on expression of other LXR target genes. This work suggests that Tgif1 can regulate nuclear receptor complexes, in addition to those containing retinoic acid receptors, but also indicates that there is some specificity to which NR target genes are repressed by Tgif1.


Subject(s)
Apolipoproteins/genetics , Gene Expression Regulation , Homeodomain Proteins/physiology , Liver/metabolism , Repressor Proteins/physiology , Animals , Apolipoprotein C-II/analysis , Apolipoprotein C-II/genetics , Apolipoproteins/analysis , Apolipoproteins A/analysis , Apolipoproteins A/genetics , Liver X Receptors , Mice , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/metabolism
17.
Cancer Res ; 80(5): 950-963, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31900260

ABSTRACT

DRAIC is a 1.7 kb spliced long noncoding RNA downregulated in castration-resistant advanced prostate cancer. Decreased DRAIC expression predicts poor patient outcome in prostate and seven other cancers, while increased DRAIC represses growth of xenografted tumors. Here, we show that cancers with decreased DRAIC expression have increased NF-κB target gene expression. DRAIC downregulation increased cell invasion and soft agar colony formation; this was dependent on NF-κB activation. DRAIC interacted with subunits of the IκB kinase (IKK) complex to inhibit their interaction with each other, the phosphorylation of IκBα, and the activation of NF-κB. These functions of DRAIC mapped to the same fragment containing bases 701-905. Thus, DRAIC lncRNA inhibits prostate cancer progression through suppression of NF-κB activation by interfering with IKK activity. SIGNIFICANCE: A cytoplasmic tumor-suppressive lncRNA interacts with and inhibits a major kinase that activates an oncogenic transcription factor in prostate cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/5/950/F1.large.jpg.


Subject(s)
Gene Expression Regulation, Neoplastic , I-kappa B Kinase/genetics , NF-kappa B/metabolism , Prostatic Neoplasms/genetics , RNA, Long Noncoding/metabolism , Animals , Cell Line, Tumor , Disease Progression , Down-Regulation , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Male , Mice , Phosphorylation/genetics , Prostate/pathology , Prostatic Neoplasms/pathology , RNA, Long Noncoding/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays
18.
Dev Biol ; 319(2): 285-97, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18508043

ABSTRACT

The mammalian placenta is the site of exchange of nutrients and waste between mother and embryo. In humans, placental insufficiency can result in intrauterine growth retardation, perinatal death and spontaneous abortion. We show that in C57BL/6J mice a null mutation in the gene encoding the transcriptional corepressor, Tgif, causes placental defects. The major defects are decreased vascularization of the placenta, due to a decrease in the fetal blood vessels, and decreased expression of the gap junction protein Gjb2 (Cx26). These defects result in severe growth retardation in a proportion of Tgif null embryos in Tgif heterozygous mothers, and an overall growth delay in Tgif null animals. Placental defects are much more severe if the mother also completely lacks Tgif function, and placentas from heterozygous Tgif embryos are defective in a Tgif null mother. Embryo transfer experiments show that even the placenta from a wild type embryo is compromised in the absence of maternal Tgif. These results demonstrate that Tgif functions in the normal development of the placenta, and suggest a role for maternal factors in regulating the morphogenesis of embryonically-derived placental tissues.


Subject(s)
Embryo, Mammalian/physiology , Homeodomain Proteins/physiology , Placenta/blood supply , Repressor Proteins/physiology , Animals , Body Weight , Cell Survival , Connexin 26 , Connexins , Embryo Transfer , Embryo, Mammalian/cytology , Embryonic Development/genetics , Embryonic Development/physiology , Female , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Organ Size , Placenta/anatomy & histology , Pregnancy , Repressor Proteins/genetics
19.
Mol Cell Biol ; 26(3): 990-1001, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16428452

ABSTRACT

TGIF (TG-interacting factor) represses transforming growth factor beta (TGF-beta)-activated gene expression and can repress transcription via a specific retinoid response element. Mutations in human TGIF are associated with holoprosencephaly, a severe defect of craniofacial development with both genetic and environmental causes. Both TGF-beta and retinoic acid signaling are implicated in craniofacial development. Here, we analyze the role of TGIF in regulating retinoid responsive gene expression. We demonstrate that TGIF interacts with the ligand binding domain of the RXRalpha retinoid receptor and represses transcription from retinoid response elements. TGIF recruits the general corepressor, CtBP, to RXRalpha, and this recruitment is required for full repression by TGIF. Interaction between TGIF and RXRalpha is reduced by the addition of retinoic acid, consistent with a role for TGIF as an RXRalpha transcriptional corepressor. We created a Tgif null mutation in mice and tested the sensitivity of mutant mice to increased levels of retinoic acid. Tgif mutant embryos are more sensitive to retinoic acid-induced teratogenesis, and retinoid target genes are expressed at a higher level in tissues from Tgif null mice. These results demonstrate an important role for TGIF as a transcriptional corepressor, which regulates developmental signaling by retinoic acid, and raises the possibility that TGIF may repress other RXR-dependent transcriptional responses.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Retinoid X Receptor alpha/antagonists & inhibitors , Retinoid X Receptor alpha/genetics , Tretinoin/metabolism , Animals , Antineoplastic Agents/pharmacology , Dimerization , Drug Resistance, Neoplasm/genetics , Embryo, Mammalian/drug effects , Embryonic Development/genetics , Female , Homeodomain Proteins/genetics , Male , Mice , Mice, Mutant Strains , Repressor Proteins/genetics , Response Elements/genetics , Retinoid X Receptor alpha/metabolism , Sequence Deletion , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Tretinoin/toxicity
20.
Atherosclerosis ; 275: 246-255, 2018 08.
Article in English | MEDLINE | ID: mdl-29980051

ABSTRACT

BACKGROUND AND AIMS: Transforming growth factor ß induced factor homeobox 1 (TGIF1) is a transcriptional repressor that limits the response to transforming growth factor ß signaling and also represses transcription independent of this pathway. Recently, we found higher serum cholesterol levels and more hepatic lipid accumulation in mice lacking Tgif1, and showed that TGIF1 can repress the expression of Soat2, the gene encoding the cholesterol esterifying enzyme acyl-Coenzyme A:cholesterol acyltransferase 2. Although there is evidence that TGIF1 plays a role in lipid metabolism, its role in this metabolic pathway is not fully characterized. Here we investigate whether overexpression of TGIF1 affects intestinal cholesterol absorption. METHODS AND RESULTS: TGIF1 was found to repress human and mouse Niemann-Pick C1 like 1 (Npc1l1) promoter activity in intestinal Caco2 cells. We also found TGIF1 to be able to oppose the induction of the promoter activity by sterol regulatory element binding protein 2 and hepatocyte nuclear factor 1α and 4α. To validate these effects of TGIF1 in vivo, we generated transgenic mice specifically overexpressing TGIF1 in the intestine (Villin-Tgif1). We observed lower intestinal expression levels of Npc1l1 that was associated with lower expression of ATP-binding cassette transporter (Abc) a1, Abcg5, and Abcg8. Villin-Tgif1 mice fed regular chow or a high-fat diet had lower levels of markers of intestinal cholesterol absorption than wild types. CONCLUSIONS: We suggest TGIF1 as a new player in intestinal cholesterol metabolism.


Subject(s)
Cholesterol, Dietary/metabolism , Homeodomain Proteins/metabolism , Intestinal Absorption , Intestinal Mucosa/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Repressor Proteins/metabolism , ATP-Binding Cassette Transporters/metabolism , Animals , Caco-2 Cells , Cholesterol 7-alpha-Hydroxylase/metabolism , Down-Regulation , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Homeodomain Proteins/genetics , Humans , Male , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Mice, Transgenic , Repressor Proteins/genetics , Sterol Regulatory Element Binding Protein 2/genetics , Sterol Regulatory Element Binding Protein 2/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL