Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Cancer Res Commun ; 3(4): 592-606, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37077937

RESUMO

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.


Assuntos
Neoplasias da Próstata , Receptores Androgênicos , Masculino , Humanos , Receptores Androgênicos/genética , Neoplasias da Próstata/tratamento farmacológico , Próstata/metabolismo , ADP Ribose Transferases/genética , Androgênios
2.
Nat Commun ; 12(1): 2705, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33976187

RESUMO

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.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/genética , Poli(ADP-Ribose) Polimerases/genética , Neoplasias da Próstata/genética , Processamento de Proteína Pós-Traducional , Receptores Androgênicos/genética , ADP-Ribosilação/efeitos dos fármacos , Adenocarcinoma , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Humanos , Masculino , Metribolona/farmacologia , Proteínas de Neoplasias/metabolismo , Ftalazinas/farmacologia , Piperazinas/farmacologia , Poli(ADP-Ribose) Polimerase-1/genética , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores Androgênicos/metabolismo , Transdução de Sinais , Análise de Sobrevida
3.
Cells ; 10(2)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33572475

RESUMO

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.


Assuntos
ADP Ribose Transferases/metabolismo , Androgênios/metabolismo , Regulação da Expressão Gênica , Proteínas de Transporte de Nucleosídeos/metabolismo , Neoplasias da Próstata/enzimologia , ADP Ribose Transferases/química , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Masculino , Camundongos , Proteínas de Transporte de Nucleosídeos/genética , Neoplasias da Próstata/patologia , Domínios Proteicos , Estabilidade Proteica , Receptores Androgênicos/metabolismo , Transdução de Sinais , Transcrição Gênica
4.
Cancer Res ; 80(5): 950-963, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31900260

RESUMO

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.


Assuntos
Regulação Neoplásica da Expressão Gênica , Quinase I-kappa B/genética , NF-kappa B/metabolismo , Neoplasias da Próstata/genética , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/metabolismo , Masculino , Camundongos , Fosforilação/genética , Próstata/patologia , Neoplasias da Próstata/patologia , RNA Longo não Codificante/genética , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Genes Dev ; 33(7-8): 388-402, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30808659

RESUMO

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.


Assuntos
Acetilcoenzima A/genética , Adenoma , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Homeodomínio/metabolismo , Neoplasias Intestinais , Proteínas Repressoras/metabolismo , Adenoma/genética , Adenoma/fisiopatologia , Polipose Adenomatosa do Colo/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Metabolismo Energético/genética , Células HCT116 , Humanos , Mucosa Intestinal/fisiopatologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL
6.
Biochim Biophys Acta Gene Regul Mech ; 1861(11): 983-995, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30312684

RESUMO

Myelin transcription factor 1 (Myt1) and Myt1l (Myt1-like) are zinc finger transcription factors that regulate neuronal differentiation. Reduced Myt1l expression has been implicated in glioblastoma (GBM), and the related St18 was originally identified as a potential tumor suppressor for breast cancer. We previously analyzed changes in gene expression in a human GBM cell line with re-expression of either Myt1 or Myt1l. This revealed largely overlapping gene expression changes, suggesting similar function in these cells. Here we show that re-expression of Myt1 or Myt1l reduces proliferation in two different GBM cell lines, activates gene expression programs associated with neuronal differentiation, and limits expression of proliferative and epithelial to mesenchymal transition gene-sets. Consistent with this, expression of both MYT1 and MYT1L is lower in more aggressive glioma sub-types. Examination of the gene expression changes in cells expressing Myt1 or Myt1l suggests that both repress expression of the YAP1 transcriptional coactivator, which functions primarily in the Hippo signaling pathway. Expression of YAP1 and its target genes is reduced in Myt-expressing cells, and there is an inverse correlation between YAP1 and MYT1/MYT1L expression in human brain cancer datasets. Proliferation of GBM cell lines is reduced by lowering YAP1 expression and increased with YAP1 over-expression, which overcomes the anti-proliferative effect of Myt1/Myt1l expression. Finally we show that reducing YAP1 expression in a GBM cell line slows the growth of orthotopic tumor xenografts. Together, our data suggest that Myt1 and Myt1l directly repress expression of YAP1, a protein which promotes proliferation and GBM growth.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Neoplasias Encefálicas , Proliferação de Células , Proteínas de Ligação a DNA/fisiologia , Glioblastoma , Proteínas do Tecido Nervoso/fisiologia , Fosfoproteínas/fisiologia , Fatores de Transcrição/fisiologia , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos Nus , Proteínas de Sinalização YAP
7.
Atherosclerosis ; 275: 246-255, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29980051

RESUMO

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.


Assuntos
Colesterol na Dieta/metabolismo , Proteínas de Homeodomínio/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Repressoras/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Células CACO-2 , Colesterol 7-alfa-Hidroxilase/metabolismo , Regulação para Baixo , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Fator 4 Nuclear de Hepatócito/genética , Fator 4 Nuclear de Hepatócito/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Camundongos Transgênicos , Proteínas Repressoras/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/genética , Proteína de Ligação a Elemento Regulador de Esterol 2/metabolismo , Regulação para Cima
8.
PLoS Genet ; 14(5): e1007409, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29782499

RESUMO

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.


Assuntos
Linhagem da Célula/genética , Neoplasias da Próstata/genética , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Animais , Proliferação de Células/genética , Progressão da Doença , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Análise de Sobrevida , Fator de Crescimento Transformador beta/metabolismo
9.
Am J Med Genet C Semin Med Genet ; 178(2): 128-139, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29749689

RESUMO

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.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Suscetibilidade a Doenças , Holoprosencefalia/etiologia , Holoprosencefalia/metabolismo , Proteínas de Homeodomínio/genética , Organogênese/genética , Proteínas Repressoras/genética , Animais , Padronização Corporal , Encéfalo/anormalidades , Encéfalo/ultraestrutura , Anormalidades Craniofaciais/etiologia , Anormalidades Craniofaciais/metabolismo , Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Regulação da Expressão Gênica , Variação Genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Proteína Nodal/genética , Proteína Nodal/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transcrição Gênica
10.
Gynecol Oncol ; 149(2): 350-360, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29551565

RESUMO

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.


Assuntos
Subunidades alfa de Fatores de Ligação ao Core/antagonistas & inibidores , Transição Epitelial-Mesenquimal/genética , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Carcinoma Epitelial do Ovário , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Subunidades alfa de Fatores de Ligação ao Core/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Cell Biochem ; 119(6): 4644-4655, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29291346

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Elementos de Resposta , Fatores de Transcrição/metabolismo , Transcrição Gênica , Células A549 , Proteínas de Ligação a DNA/genética , Glioblastoma/genética , Glioblastoma/patologia , Células HEK293 , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição/genética
12.
Prostate ; 77(15): 1452-1467, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28875501

RESUMO

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.


Assuntos
Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteína Quinase C/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Diferenciação Celular/fisiologia , Progressão da Doença , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , PTEN Fosfo-Hidrolase/metabolismo , Neoplasias da Próstata/genética , Proteína Quinase C/genética , Serina-Treonina Quinases TOR/genética
13.
Adv Exp Med Biol ; 963: 35-50, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28197905

RESUMO

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.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , DNA/metabolismo , Histonas/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Sumoilação , Animais , Cromatina/química , DNA/química , Histonas/química , Humanos , Conformação de Ácido Nucleico , Conformação Proteica , Relação Estrutura-Atividade , Transcrição Gênica , Ubiquitina-Proteína Ligases/metabolismo
14.
Mol Cell Biol ; 37(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27956704

RESUMO

Mouse embryos conditionally lacking Tgif1 and Tgif2 have holoprosencephaly and defects in left-right asymmetry. To identify pathways affected by loss of Tgif function during embryogenesis, we performed transcriptome profiling on whole mouse embryos. Among the genes with altered expression in embryos lacking Tgifs were a number with links to cilium function. One of these, Evi5l, encodes a RabGAP that is known to block the formation of cilia when overexpressed. Evi5l expression is increased in Tgif1; Tgif2-null embryos and in double-null mouse embryo fibroblasts (MEFs). Knockdown of Tgifs in a human retinal pigment epithelial cell line also increased EVI5L expression. We show that TGIF1 binds to a conserved consensus TGIF site 5' of the human and mouse Evi5l genes and represses Evi5l expression. In primary MEFs lacking both Tgifs, the number of cells with primary cilia was significantly decreased, and we observed a reduction in the transcriptional response to Shh pathway activation. Reducing Evi5l expression in MEFs lacking Tgifs resulted in a partial restoration of cilium numbers and in the transcriptional response to activation of the Shh pathway. In summary, this work shows that Tgifs regulate ciliogenesis and suggests that Evi5l mediates at least part of this effect.


Assuntos
Proteínas Ativadoras de GTPase/genética , Proteínas de Homeodomínio/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Animais , Linhagem Celular , Cílios/metabolismo , Embrião de Mamíferos/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Knockout , Proteínas Repressoras/genética , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo
15.
Eur J Hum Genet ; 25(2): 208-215, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27924807

RESUMO

Holoprosencephaly (HPE) is a prevalent craniofacial developmental disorder that has both genetic and environmental causes. The gene encoding TG-interacting factor 1 (TGIF1) is among those that are routinely screened in HPE patients. However, the mechanisms by which TGIF1 variants cause HPE are not fully understood. TGIF1 is a transcriptional repressor that limits the output of the Transforming Growth Factor ß (TGFß)/Nodal signaling pathway, and HPE in patients with TGIF1 variants has been suggested to be due to increased Nodal signaling. Mice lacking both Tgif1 and its paralog, Tgif2, have HPE, and embryos lacking Tgif function do not survive past mid-gestation. Here, we show that in the presence of a Nodal heterozygous mutation, proliferation defects are rescued and a proportion of embryos lacking all Tgif function survive to late gestation. However, these embryos have a classic HPE phenotype, suggesting that this is a Nodal-independent effect of Tgif loss of function. Further, we show that the Gli3 gene is a direct target for repression by Tgifs, independent of TGFß/Nodal signaling, consistent with Tgif mutations causing HPE via Nodal-independent effects on the Sonic Hedgehog (Shh) pathway. Based on this work, we propose a model for distinct functions of Tgifs in the Nodal and Shh/Gli3 pathways during forebrain development.


Assuntos
Holoprosencefalia/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição Kruppel-Like/genética , Proteínas do Tecido Nervoso/genética , Tubo Neural/embriologia , Proteína Nodal/genética , Proteínas Repressoras/genética , Animais , Linhagem Celular Tumoral , Células Cultivadas , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/metabolismo , Proteína Nodal/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Proteína Gli3 com Dedos de Zinco
16.
PLoS One ; 11(5): e0155837, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27187787

RESUMO

Tgif1 and Tgif2 are transcriptional repressors that inhibit the transcriptional response to transforming growth factor ß signaling, and can repress gene expression by direct binding to DNA. Loss of function mutations in TGIF1 are associated with holoprosencephaly (HPE) in humans. In mice, embryos lacking both Tgif1 and Tgif2 fail to complete gastrulation, and conditional double null embryos that survive past gastrulation have HPE and do not survive past mid-gestation. Here we show that in mice of a relatively pure C57BL/6 strain background, loss of Tgif1 alone results in defective axial patterning and altered expression of Hoxc6. The primary defects in Tgif1 null embryos are the presence of extra ribs on the C7 vertebra, consistent with a posterior transformation phenotype. In addition we observed defective cervical vertebrae, primarily C1-C5, in both adult mice and embryos that lacked Tgif1. The combination of Tgif1 and Tgif2 mutations increases the severity and penetrance of the posterior transformation phenotype, without altering the type of defects seen. Similarly, exposure of Tgif1 mutant embryos to retinoic acid at E8.5 increased the severity and penetrance of the Tgif1 phenotype. This suggests that Tgif1 and Tgif2 regulate axial patterning and that reduced TGIF function sensitizes embryos to the effects of retinoic acid.


Assuntos
Padronização Corporal/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas Repressoras/fisiologia , Animais , Padronização Corporal/efeitos dos fármacos , Padronização Corporal/genética , Vértebras Cervicais/anormalidades , Vértebras Cervicais/embriologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Proteínas Repressoras/genética , Costelas/anormalidades , Costelas/embriologia , Esqueleto/anormalidades , Tretinoína/farmacologia
17.
Cancer Discov ; 4(10): 1123-5, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25274684

RESUMO

A subset of glioblastomas (GBM) has high levels of TGFß signaling, and anti-TGFß therapies are being pursued as treatments for GBM. The work presented here identifies CREB1 as a potential biomarker for TGFß-dependent GBM. CREB1 integrates signaling from TGFß and the PI3K pathway and nucleates a self-sustaining signaling loop that maintains TGFß2 expression in GBM with high CREB1 levels.


Assuntos
Comunicação Autócrina , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Glioblastoma/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Animais , Humanos
18.
PLoS One ; 9(3): e92800, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24651496

RESUMO

Recent work with mouse models of prostate cancer (CaP) has shown that inactivation of TGFß signaling in prostate epithelium can cooperate with deletion of the Pten tumor suppressor to drive locally aggressive cancer and metastatic disease. Here, we show that inactivating the TGFß pathway by deleting the gene encoding the TGFß type II receptor (Tgfbr2) in combination with a deletion of the Apc tumor suppressor gene specifically in mouse prostate epithelium, results in the rapid onset of invasive CaP. Micro-metastases were observed in the lymph nodes and lungs of a proportion of the double mutant mice, whereas no metastases were observed in Apc single mutant mice. Prostate-specific Apc;Tgfbr2 mutants had a lower frequency of metastasis and survived significantly longer than Pten;Tgfbr2 double mutants. However, all Apc;Tgfbr2 mutants developed invasive cancer by 30 weeks of age, whereas invasive cancer was rarely observed in Apc single mutant animals, even by one year of age. Further comparison of the Pten and Apc models of CaP revealed additional differences, including adenosquamous carcinoma in the Apc;Tgfbr2 mutants that was not seen in the Pten model, and a lack of robust induction of the TGFß pathway in Apc null prostate. In addition to causing high-grade prostate intra-epithelial neoplasia (HGPIN), deletion of either Pten or Apc induced senescence in affected prostate ducts, and this restraint was overcome by loss of Tgfbr2. In summary, this work demonstrates that TGFß signaling restrains the progression of CaP induced by different tumor suppressor mutations, suggesting that TGFß signaling exerts a general tumor suppressive effect in prostate.


Assuntos
Proteína da Polipose Adenomatosa do Colo/deficiência , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular , Senescência Celular/genética , Modelos Animais de Doenças , Progressão da Doença , Deleção de Genes , Homozigoto , Queratina-10 , Masculino , Camundongos , Camundongos Knockout , Mutação , Invasividade Neoplásica , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Neoplasias da Próstata/mortalidade , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Células Estromais/metabolismo
19.
J Lipid Res ; 55(4): 709-17, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24478032

RESUMO

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.


Assuntos
Inativação Gênica , Proteínas de Homeodomínio/fisiologia , Proteínas Repressoras/fisiologia , Esterol O-Aciltransferase/genética , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Repressão Enzimática , Feminino , Cálculos Biliares/enzimologia , Fator 1-alfa Nuclear de Hepatócito/fisiologia , Fator 4 Nuclear de Hepatócito/fisiologia , Proteínas de Homeodomínio/metabolismo , Humanos , Lipídeos/sangue , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Ligação Proteica , Caracteres Sexuais , Esterol O-Aciltransferase/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Esterol O-Aciltransferase 2
20.
Mol Cell Biol ; 33(24): 4824-33, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24100014

RESUMO

TG-interacting factor 1 (TGIF1) is a transcriptional repressor that can modulate retinoic acid and transforming growth factor ß signaling pathways. It is required for myeloid progenitor cell differentiation and survival, and mutations in the TGIF1 gene cause holoprosencephaly. Furthermore, we have previously observed that acute myelogenous leukemia (AML) patients with low TGIF1 levels had worse prognoses. Here, we explored the role of Tgif1 in murine hematopoietic stem cell (HSC) function. CFU assays showed that Tgif1(-/-) bone marrow cells produced more total colonies and had higher serial CFU potential. These effects were also observed in vivo, where Tgif1(-/-) bone marrow cells had higher repopulation potential in short- and long-term competitive repopulation assays than wild-type cells. Serial transplantation and replating studies showed that Tgif1(-/-) HSCs exhibited greater self-renewal and were less proliferative and more quiescent than wild-type cells, suggesting that Tgif1 is required for stem cells to enter the cell cycle. Furthermore, HSCs from Tgif1(+/-) mice had a phenotype similar to that of HSCs from Tgif1(-/-) mice, while bone marrow cells with overexpressing Tgif1 showed increased proliferation and lower survival in long-term transplant studies. Taken together, our data suggest that Tgif1 suppresses stem cell self-renewal and provide clues as to how reduced expression of TGIF1 may contribute to poor long-term survival in patients with AML.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas Repressoras/fisiologia , Animais , Células da Medula Óssea/metabolismo , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA