Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 11595, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773164

RESUMEN

Despite growing evidence implicating the calcium-activated chloride channel anoctamin1 (ANO1) in cancer metastasis, its direct impact on the metastatic potential of prostate cancer and the possible significance of epigenetic alteration in this process are not fully understood. Here, we show that ANO1 is minimally expressed in LNCap and DU145 prostate cancer cell lines with low metastatic potential but overexpressed in high metastatic PC3 prostate cancer cell line. The treatment of LNCap and DU145 cells with DNMT inhibitor 5-aza-2'-deoxycytidine (5-Aza-CdR) potentiates ANO1 expression, suggesting that DNA methylation is one of the mechanisms controlling ANO1 expression. Consistent with this notion, hypermethylation was detected at the CpG island of ANO1 promoter region in LNCap and DU145 cells, and 5-Aza-CdR treatment resulted in a drastic demethylation at promoter CpG methylation sites. Upon 5-Aza-CdR treatment, metastatic indexes, such as cell motility, invasion, and metastasis-related gene expression, were significantly altered in LNCap and DU145 cells. These 5-Aza-CdR-induced metastatic hallmarks were, however, almost completely ablated by stable knockdown of ANO1. These in vitro discoveries were further supported by our in vivo observation that ANO1 expression in xenograft mouse models enhances the metastatic dissemination of prostate cancer cells into tibial bone and the development of osteolytic lesions. Collectively, our results help elucidate the critical role of ANO1 expression in prostate cancer bone metastases, which is epigenetically modulated by promoter CpG methylation.


Asunto(s)
Anoctamina-1 , Neoplasias Óseas , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias , Regiones Promotoras Genéticas , Neoplasias de la Próstata , Masculino , Anoctamina-1/metabolismo , Anoctamina-1/genética , Humanos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Óseas/secundario , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Ratones , Islas de CpG , Decitabina/farmacología , Movimiento Celular/genética , Epigénesis Genética , Azacitidina/farmacología
2.
Mol Oncol ; 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38600695

RESUMEN

Matrix metalloproteinase 9 (MMP-9) is a member of the MMP family and has been recently identified as a nuclear protease capable of clipping histone H3 N-terminal tails (H3NT). This MMP-9-dependent H3NT proteolysis is critical for establishing an active state of gene transcription during osteoclast differentiation and melanoma development. However, whether H3NT cleavage by MMP-9 plays a similar role in other cellular events has not been explored. Here, we dissect the functional contribution of MMP-9-dependent H3NT clipping to colonic tumorigenesis by using a combination of genome-wide transcriptome data, ChIP/ChIPac-qPCR, CRISPR/dCas9 gene-targeting system, and in vivo xenograft models. We show that MMP-9 is overexpressed in colon cancer cells and catalyzes H3NT proteolysis to drive transcriptional activation of growth stimulatory genes. Our studies using knockdown and inhibition approaches clearly indicate that MMP-9 mediates transcriptional activation and promotes colonic tumorigenesis in a manner dependent on its protease activity toward H3NT. Remarkably, artificial H3NT proteolysis at target gene promoters with dCAS9-MMP-9 is sufficient for establishing their transcriptional competence in colon cancer cells, underscoring the importance of MMP-9-dependent H3NT proteolysis per se in the transactivation process. Our data establish new functions and mechanisms for MMP-9 in driving the oncogenic transcription program in colon cancer through H3NT proteolysis, and demonstrate how this epigenetic pathway can be exploited as a potential therapeutic target for cancer treatment.

3.
Biomedicines ; 11(9)2023 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-37760992

RESUMEN

Vpr binding protein (VprBP), also known as DDB1- and CUL4-associated factor1 (DCAF1), is a recently identified atypical kinase and plays an important role in downregulating the transcription of tumor suppressor genes as well as increasing the risk for colon and prostate cancers. Melanoma is the most aggressive form of skin cancer arising from pigment-producing melanocytes and is often associated with the dysregulation of epigenetic factors targeting histones. Here, we demonstrate that VprBP is highly expressed and phosphorylates threonine 120 (T120) on histone H2A to drive the transcriptional inactivation of growth-regulatory genes in melanoma cells. As is the case for its epigenetic function in other types of cancers, VprBP acts to induce a gene silencing program dependent on H2AT120 phosphorylation (H2AT120p). The significance of VprBP-mediated H2AT120p is further underscored by the fact that VprBP knockdown- or VprBP inhibitor-induced lockage of H2AT120p mitigates melanoma tumor growth in xenograft models. Collectively, our results establish VprBP-mediated H2AT120p as a key epigenetic signal for melanomagenesis and suggest the therapeutic potential of targeting VprBP kinase activity for effective melanoma treatment.

4.
Res Sq ; 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37293029

RESUMEN

Background: Melanoma is the most aggressive form of skin cancer arising from pigment-producing melanocytes and is often associated with dysregulation of epigenetic factors targeting histones. VprBP, also known as DCAF1, is a recently identified kinase and plays an important role in downregulating the transcription of tumor suppressor genes as well as increasing the risk for colon and prostate cancers. However, it remains unknown whether VprBP is also involved in triggering the pathogenesis of other types of cancer. Results: We demonstrate that VprBP is highly expressed and phosphorylates threonine 120 (T120) on histone H2A to drive transcriptional inactivation of growth regulatory genes in melanoma cells. As is the case for its epigenetic function in colon and prostate cancers, VprBP acts to induce gene silencing program dependently of H2AT120 phosphorylation (H2AT120p). The significance of VprBP-mediated H2AT120p is further underscored by the fact that VprBP knockdown- or VprBP inhibitor-induced lockage of H2AT120p mitigates melanoma tumor growth in xenograft models. Moreover, artificial tethering of VprBP wild type, but not VprBP kinase-dead mutant, to its responsive genes is sufficient for achieving an inactive transcriptional state in VprBP-depleted cells, indicating that VprBP drives gene silencing program in an H2AT120p-dependent manner. Conclusions: Our results establish VprBP-mediated H2AT120p as a key epigenetic signal for melanomagenesis and suggest the therapeutic potential of targeting VprBP kinase activity for effective melanoma treatment.

5.
Nat Commun ; 14(1): 2140, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069142

RESUMEN

Our recent work has shown that DCAF1 (also known as VprBP) is overexpressed in colon cancer and phosphorylates histone H2AT120 to drive epigenetic gene inactivation and oncogenic transformation. We have extended these observations by investigating whether DCAF1 also phosphorylates non-histone proteins as an additional mechanism linking its kinase activity to colon cancer development. We now demonstrate that DCAF1 phosphorylates EZH2 at T367 to augment its nuclear stabilization and enzymatic activity in colon cancer cells. Consistent with this mechanistic role, DCAF1-mediated EZH2 phosphorylation leads to elevated levels of H3K27me3 and altered expression of growth regulatory genes in cancer cells. Furthermore, our preclinical studies using organoid and xenograft models revealed that EZH2 requires phosphorylation for its oncogenic function, which may have therapeutic implications for gene reactivation in colon cancer cells. Together, our data define a mechanism underlying DCAF1-driven colonic tumorigenesis by linking DCAF1-mediated EZH2 phosphorylation to EZH2 stability that is crucial for establishing H3K27me3 and gene silencing program.


Asunto(s)
Neoplasias del Colon , Proteína Potenciadora del Homólogo Zeste 2 , Histonas , Proteínas Serina-Treonina Quinasas , Ubiquitina-Proteína Ligasas , Humanos , Neoplasias del Colon/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Silenciador del Gen , Genes Reguladores , Histonas/genética , Histonas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
6.
Oncogene ; 42(17): 1405-1416, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37041410

RESUMEN

VprBP (also known as DCAF1) is a recently identified kinase that is overexpressed in cancer cells and serves as a major determinant for epigenetic gene silencing and tumorigenesis. The role of VprBP in driving target gene inactivation has been largely attributed to its ability to mediate histone H2A phosphorylation. However, whether VprBP also phosphorylates non-histone proteins and whether these phosphorylation events drive oncogenic signaling pathways have not been explored. Here we report that serine 367 phosphorylation (S367p) of p53 by VprBP is a key player in attenuating p53 transcriptional and growth suppressive activities. VprBP catalyzes p53S367p through a direct interaction with the C-terminal domain of p53. Mechanistically, VprBP-mediated S367p inhibits p53 function in the wake of promoting p53 proteasomal degradation, because blocking p53S367p increases p53 protein levels, thereby enhancing p53 transactivation. Furthermore, abrogation of VprBP-p53 interaction by p53 acetylation is critical for preventing p53S367p and potentiating p53 function in response to DNA damage. Together, our findings establish VprBP-mediated S367p as a negative regulator of p53 function and identify a previously uncharacterized mechanism by which S367p modulates p53 stability.


Asunto(s)
Transducción de Señal , Proteína p53 Supresora de Tumor , Humanos , Fosforilación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Fosfotransferasas
7.
Oncogene ; 41(4): 560-570, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34785776

RESUMEN

Melanoma is a type of skin cancer that develops in pigment-producing melanocytes and often spreads to other parts of the body. Aberrant gene expression has been considered as a crucial step for increasing the risk of melanomagenesis, but how chromatin reorganization contributes to this pathogenic process is still not well understood. Here we report that matrix metalloproteinase 9 (MMP-9) localizes to the nucleus of melanoma cells and potentiates gene expression by proteolytically clipping the histone H3 N-terminal tail (H3NT). From genome-wide studies, we discovered that growth-regulatory genes are selectively targeted and activated by MMP-9-dependent H3NT proteolysis in melanoma cells. MMP-9 cooperates functionally with p300/CBP because MMP-9 cleaves H3NT in a manner that is dependent on p300/CBP-mediated acetylation of H3K18. The functional significance of MMP-9-dependent H3NT proteolysis is further underscored by the fact that RNAi knockdown and small-molecule inhibition of MMP-9 and p300/CBP impede melanomagenic gene expression and melanoma tumor growth. Together, our data establish new functions and mechanisms for nuclear MMP-9 in promoting melanomagenesis and demonstrate how MMP-9-dependent H3NT proteolysis can be exploited to prevent and treat melanoma skin cancer.


Asunto(s)
Histonas/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Melanoma/genética , Animales , Humanos , Melanoma/patología , Ratones , Proteolisis
8.
Mol Oncol ; 15(10): 2801-2817, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34312968

RESUMEN

Histone modification is aberrantly regulated in cancer and generates an unbalanced state of gene transcription. VprBP, a recently identified kinase, phosphorylates histone H2A on threonine 120 (T120) and is involved in oncogenic transcriptional dysregulation; however, its specific role in colon cancer is undefined. Here, we show that VprBP is overexpressed in colon cancer and directly contributes to epigenetic gene silencing and cancer pathogenesis. Mechanistically, the observed function of VprBP is mediated through H2AT120 phosphorylation (H2AT120p)-driven transcriptional repression of growth regulatory genes, resulting in a significantly higher proliferative capacity of colon cancer cells. Our preclinical studies using organoid and xenograft models demonstrate that treatment with the VprBP inhibitor B32B3 impairs colonic tumor growth by blocking H2AT120p and reactivating a transcriptional program resembling that of normal cells. Collectively, our work describes VprBP as a master kinase contributing to the development and progression of colon cancer, making it a new molecular target for novel therapeutic strategies.


Asunto(s)
Neoplasias del Colon , Histonas , Proteínas Serina-Treonina Quinasas , Ubiquitina-Proteína Ligasas , Neoplasias del Colon/genética , Epigénesis Genética , Silenciador del Gen , Histonas/metabolismo , Humanos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Ubiquitina-Proteína Ligasas/fisiología
9.
Am J Physiol Cell Physiol ; 320(3): C448-C461, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33471620

RESUMEN

Gram-negative bacterial lipopolysaccharide (LPS) increases the susceptibility of cells to pathogenic diseases, including inflammatory diseases and septic syndrome. In our experiments, we examined whether LPS induces epithelial barrier disruption in secretory epithelia and further investigated its underlying mechanism. The activities of Ca2+-activated Cl- channels (CACC) and epithelial Na+ channels (ENaC) were monitored with a short-circuit current using an Ussing chamber. Epithelial membrane integrity was estimated via transepithelial electrical resistance and paracellular permeability assays. We found that the apical application of LPS evoked short-circuit current (Isc) through the activation of CACC and ENaC. Although LPS disrupted epithelial barrier integrity, this was restored with the inhibition of CACC and ENaC, indicating the role of CACC and ENaC in the regulation of paracellular pathways. We confirmed that LPS, CACC, or ENaC activation evoked apical membrane depolarization. The exposure to a high-K+ buffer increased paracellular permeability. LPS induced the rapid redistribution of zonula occludens-1 (ZO-1) and reduced the expression levels of ZO-1 in tight junctions through apical membrane depolarization and tyrosine phosphorylation. However, the LPS-induced epithelial barrier disruption and degradation of ZO-1 were largely recovered by blocking CACC and ENaC. Furthermore, although LPS-impaired epithelial barrier became vulnerable to secondary bacterial infections, this vulnerability was prevented by inhibiting CACC and ENaC. We concluded that LPS induces the disruption of epithelial barrier integrity through the activation of CACC and ENaC, resulting in apical membrane depolarization and the subsequent tyrosine phosphorylation of ZO-1.


Asunto(s)
Canales de Cloruro/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Lipopolisacáridos/farmacología , Canales de Sodio/metabolismo , Animales , Células Cultivadas , Masculino , Potenciales de la Membrana/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
10.
Front Psychol ; 12: 678803, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35185663

RESUMEN

Many messengers and social networking services (SNSs) use emojis and stickers as a means of communication. Stickers express individual emotions well, allowing long texts to be replaced with small pictures. As the use of stickers increased, stickers were commercialized on a few platforms and showed remarkable growth as people bought and used stickers with their favorite characters, products, or entertainers online. Depending on their personality, individuals have different motivations for using stickers that determine the usefulness and enjoyment of stickers, affecting their purchase decisions. In the present study, participants (n = 302) who were randomly recruited from a university completed an online questionnaire assessing the Big Five personality characteristics, motivations for using stickers, and the technology acceptance model (TAM). Results using partial least squares structural equation modeling (PLS-SEM) revealed that each personality trait affected different motivations for using stickers. Moreover, motivations for using stickers also influenced different technology acceptance variables. Finally, perceived usefulness, enjoyment, and ease of use had a positive effect on the intention to purchase stickers. This study has implications in that it is an exploratory approach to the intention to purchase stickers, which has been investigated by few prior studies, and it sheds light on the relationship between personality, motivation, and TAM in purchasing stickers. It also suggests that personality and motivation factors can be considered in personalized recommendation services.

11.
J Clin Med ; 9(3)2020 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-32182826

RESUMEN

Cystic fibrosis transmembrane conductance regulator (CFTR), a cyclic AMP (cAMP)-regulated chloride channel, is critical for secretion and absorption across diverse epithelia. Mutations or absence of CFTR result in pathogeneses, including cancer. While CFTR has been proposed as a tumor suppressing gene in tumors of the intestine, lung, and breast cancers, its effects in head and neck cancer (HNC) have yet to be investigated. This study aimed to define expression patterns and epigenetic modifications of CFTR in HNC. CFTR was expressed in normal but not in HNC cells and tissues. Treatment with 5-aza-2'-deoxycytidine (5-Aza-CdR) was associated with rescued expression of CFTR, whose function was confirmed by patch clamp technique. Further experiments demonstrated that CFTR CpG islands were hypermethylated in cancer cells and tissues and hypomethylated in normal cells and tissue. Our results suggest that CFTR epigenetic modifications are critical in both down-regulation and up-regulation of CFTR expression in HNC and normal cells respectively. We then investigated the impact of CFTR on expressions and functions of cancer-related genes. CFTR silencing was closely associated with changes to other cancer-related genes, suppressing apoptosis while enhancing proliferation, cell motility, and invasion in HNC. Our findings demonstrate that hypermethylation of CFTR CpG islands and CFTR deficiency is closely related to HNC.

12.
Int J Mol Sci ; 20(24)2019 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-31847128

RESUMEN

Anoctamin1 (ANO1), a calcium activated chloride channel, is known to play a critical role in salivary secretion. In the salivary gland, ANO1 is expressed exclusively in the acinar cells, with no expression in the ductal cells. However, the mechanisms that determine this distinctive cell type-dependent expression pattern of ANO1 remain unknown. In this study, we discovered that the cell-dependent expression of ANO1 during salivary gland organogenesis is regulated by DNA methylation of ANO1 CpG islands. ANO1 CpG islands in e12 embryonic submandibular glands (eSMG) are highly methylated, but those in e14 eSMG or adult SMG are significantly unmethylated. The differential expression pattern of ANO1 in duct and acini is defined at e14. Artificial demethylation by treatment with the demethylating agent 5-aza-2'-deoxycytidine (5-Aza-CdR), induced the expression of ANO1 in both the ductal cell line Human Submandibular Gland (HSG) and in the duct cells of adult mouse SMG. During the trans-differentiation in Matrigel of duct-origin HSG cells into acinar-like phenotype, significant demethylation of ANO1 CpG islands is observed. This may be due to the reduced expression of DNA methyltransferase (DNMT) 3a and 3b. These results suggest that the differential expression of ANO1 in salivary glands during organogenesis and differentiation is mainly regulated by epigenetic demethylation of the ANO1 gene.


Asunto(s)
Anoctamina-1/biosíntesis , Islas de CpG , Metilación de ADN , Epigénesis Genética , Proteínas de Neoplasias/biosíntesis , Glándulas Salivales/metabolismo , Animales , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN Metiltransferasa 3A , Decitabina/farmacología , Humanos , Ratones , Glándulas Salivales/citología , ADN Metiltransferasa 3B
13.
Mol Oncol ; 13(9): 1976-1992, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31293051

RESUMEN

p53 is a sequence-specific transcription factor, and proper regulation of p53 transcriptional activity is critical for orchestrating different tumor-suppressive mechanisms. p32 is a multifunctional protein which interacts with a large number of viral proteins and transcription factors. Here, we investigate the effect of p32 on p53 transactivation and identify a novel mechanism by which p32 alters the functional characteristics of p53. Specifically, p32 attenuates p53-dependent transcription through impairment of p53 binding to its response elements on target genes. Upon p32 expression, p53 levels bound at target genes are decreased, and p53 target genes are inactivated, strongly indicating that p32 restricts p53 occupancy and function at target genes. The primary mechanism contributing to the observed action of p32 is the ability of p32 to interact with the p53 tetramerization domain and to block p53 tetramerization, which in turn enhances nuclear export and degradation of p53, leading to defective p53 transactivation. Collectively, these data establish p32 as a negative regulator of p53 function and suggest the therapeutic potential of targeting p32 for cancer treatment.


Asunto(s)
Proteínas Portadoras/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Mitocondriales/metabolismo , Neoplasias/metabolismo , Multimerización de Proteína , Elementos de Respuesta , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Humanos , Proteínas Mitocondriales/genética , Neoplasias/genética , Neoplasias/patología , Proteína p53 Supresora de Tumor/genética
14.
Epigenetics Chromatin ; 12(1): 25, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992059

RESUMEN

BACKGROUND: MMP-9-dependent proteolysis of histone H3 N-terminal tail (H3NT) is an important mechanism for activation of gene expression during osteoclast differentiation. Like other enzymes targeting their substrates within chromatin structure, MMP-9 enzymatic activity toward H3NT is tightly controlled by histone modifications such as H3K18 acetylation (H3K18ac) and H3K27 monomethylation (H3K27me1). Growing evidence indicates that DNA methylation is another epigenetic mechanism controlling osteoclastogenesis, but whether DNA methylation is also critical for regulating MMP-9-dependent H3NT proteolysis and gene expression remains unknown. RESULTS: We show here that treating RANKL-induced osteoclast progenitor (OCP) cells with the DNMT inhibitor 5-Aza-2'-deoxycytidine (5-Aza-CdR) induces CpG island hypomethylation and facilitates MMP-9 transcription. This increase in MMP-9 expression results in a significant enhancement of H3NT proteolysis and OCP cell differentiation. On the other hand, despite an increase in levels of H3K18ac, treatment with the HDAC inhibitor trichostatin A (TSA) leads to impairment of osteoclastogenic gene expression. Mechanistically, TSA treatment of OCP-induced cells stimulates H3K27ac with accompanying reduction in H3K27me1, which is a key modification to facilitate stable interaction of MMP-9 with nucleosomes for H3NT proteolysis. Moreover, hypomethylated osteoclastogenic genes in 5-Aza-CdR-treated cells remain transcriptionally inactive after TSA treatment, because H3K27 is highly acetylated and cannot be modified by G9a. CONCLUSIONS: These findings clearly indicate that DNA methylation and histone modification are important mechanisms in regulating osteoclastogenic gene expression and that their inhibitors can be used as potential therapeutic tools for treating bone disorders.


Asunto(s)
Diferenciación Celular , Histonas/metabolismo , Osteoclastos/citología , Osteogénesis , Proteolisis , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Histonas/química , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Osteoclastos/metabolismo
15.
Cell Rep ; 24(1): 224-237, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29972783

RESUMEN

Breast cancer cells relocate to bone and activate osteoclast-induced bone resorption. Soluble factors secreted by breast cancer cells trigger a cascade of events that stimulate osteoclast differentiation in the bone microenvironment. MacroH2A is a unique histone variant with a C-terminal non-histone domain and plays a crucial role in modulating chromatin organization and gene transcription. Here, we show that macroH2A1.2, one of the macroH2A isoforms, has an intrinsic ability to inhibit breast cancer-derived osteoclastogenesis. This repressive effect requires macroH2A1.2-dependent attenuation of expression and secretion of lysyl oxidase (LOX) in breast cancer cells. Furthermore, our mechanistic studies reveal that macroH2A1.2 physically and functionally interacts with the histone methyltransferase EZH2 and elevates H3K27me3 levels to keep LOX gene in a repressed state. Collectively, this study unravels a role for macroH2A1.2 in regulating osteoclastogenic potential of breast cancer cells, suggesting possibilities for developing therapeutic tools to treat osteolytic bone destruction.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Osteogénesis , Animales , Resorción Ósea/patología , Neoplasias de la Mama/genética , Diferenciación Celular , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metilación , Ratones Endogámicos C57BL , Nucleosomas/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Fosforilación , Unión Proteica , Proteína-Lisina 6-Oxidasa/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo
16.
Oncogene ; 37(43): 5749-5765, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29925860

RESUMEN

Osteoclasts are multinuclear bone-resorbing cells that differentiate from hematopoietic precursor cells. Prostate cancer cells frequently spread to bone and secrete soluble signaling factors to accelerate osteoclast differentiation and bone resorption. However, processes and mechanisms that govern the expression of osteoclastogenic soluble factors secreted by prostate cancer cells are largely unknown. MacroH2A (mH2A) is a histone variant that replaces canonical H2A at designated genomic loci and establishes functionally distinct chromatin regions. Here, we report that mH2A1.2, one of the mH2A isoforms, attenuates prostate cancer-induced osteoclastogenesis by maintaining the inactive state of genes encoding soluble factors in prostate cancer cells. Our functional analyses of soluble factors identify lymphotoxin beta (LTß) as a major stimulator of osteoclastogenesis and an essential mH2A1.2 target for its anti-osteoclastogenic activity. Mechanistically, mH2A1.2 directly interacts with HP1α and H1.2 and requires them to inactivate LTß gene in prostate cancer cells. Consistently, HP1α and H1.2 have an intrinsic ability to inhibit osteoclast differentiation in a mH2A1.2-dependent manner. Together, our data uncover a new and specific role for mH2A1.2 in modulating osteoclastogenic potential of prostate cancer cells and demonstrate how this signaling pathway can be exploited to treat osteolytic bone metastases at the molecular level.


Asunto(s)
Neoplasias Óseas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoclastos/metabolismo , Osteólisis/metabolismo , Neoplasias de la Próstata/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Línea Celular Tumoral , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Histonas/genética , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Osteoclastos/patología , Osteólisis/genética , Osteólisis/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología
17.
Epigenetics Chromatin ; 11(1): 23, 2018 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-29807539

RESUMEN

BACKGROUND: MMP-9 plays a direct role in the activation of pro-osteoclastogenic genes by cleaving histone H3N-terminal tail (H3NT) and altering chromatin architecture. Although H3 acetylation at K18 has been shown to stimulate MMP-9 enzymatic activity toward H3NT, nothing is known about the influence of other H3NT modifications on this epigenetic reaction. RESULTS: We show that H3 monomethylation at lysine 27 (H3K27me1) is essential for MMP-9-dependent H3NT proteolysis during RANKL-induced osteoclast differentiation. Through the recognition of H3K27me1 mark, MMP-9 localizes and generates H3NT proteolysis at the genes encoding osteoclast differentiation factors. By using RNAi and small molecule inhibitor approaches, we also confirmed that G9a is the major methyltransferase to catalyze H3K27me1 for MMP-9-dependent H3NT proteolysis and trigger the expression of osteoclast-specific genes. CONCLUSIONS: Our data establish new functions for G9a-mediated H3K27me1 in MMP-9-dependent H3NT proteolysis and demonstrate how histone modification can be exploited to regulate osteoclastogenic gene expression at the molecular level. Further studies are warranted to investigate the detailed mechanism by which G9a overexpression with concomitant dysregulation of osteoclastogenesis contributes to the pathogenesis of bone disorders.


Asunto(s)
Antígenos de Histocompatibilidad/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Lisina/química , Metaloproteinasa 9 de la Matriz/metabolismo , Osteogénesis , Ligando RANK/farmacología , Cromatina/química , Epigénesis Genética , Células HEK293 , Humanos , Metilación , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Proteolisis
18.
Neurospine ; 15(1): 86-90, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29656628

RESUMEN

OBJECTIVE: We evaluated the efficacy of diffusion-weighted magnetic resonance imaging (DW-MRI) in detecting perioperative cerebral thromboembolic events after anterior cervical discectomy and fusion (ACDF). METHODS: This retrospective case series included 51 consecutive patients that underwent ACDF. To assess baseline presence of plaque or stenosis, all patients were assessed for risk factors of cerebrovascular events and underwent carotid ultrasound preoperatively and cerebral DW-MRI was performed in all patients postoperatively, with areas of high signal intensity interpreted as a cerebral thromboembolic event. RESULTS: One male patient who underwent a C4/5 ACDF had a focal diffusion abnormality on DW-MRI concerning for cerebral thromboembolic events in the right posterior cerebral artery territory. He remained asymptomatic and did not display related neurological symptoms, such as visual deficits. CONCLUSION: Asymptomatic cerebral thromboembolic events can be detected by DW-MRI after ACDF. The incidence of such events remains very rare despite the direct manipulation and associated alteration of common carotid artery flow dynamics.

19.
Sci Rep ; 7(1): 13278, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29038508

RESUMEN

Wnt signaling controls critical developmental processes including tissue/body patterning. Here we report the identification of a novel regulator of Wnt signaling, OTTOGI (OTG), isolated from a large-scale expression screening of human cDNAs in zebrafish embryos. Overexpression of OTG in zebrafish embryos caused dorso-anteriorized phenotype, inhibited the expression of Wnt target genes, and prevented nuclear accumulation of ß-catenin. Conversely, knockdown of zebrafish otg using specific antisense morpholino promoted nuclear accumulation of ß-catenin and caused ventralization. However, OTG failed to rescue headless-like phenotype induced by inhibition of GSK-3ß activity, suggesting that OTG acts upstream of GSK-3ß. OTG bound specifically to Frizzled8 (Fz8) receptor and caused retention of Fz8 in the endoplasmic reticulum possibly by preventing N-linked glycosylation of Fz8. Taken together, our data indicate that OTG functions as a novel negative regulator of Wnt signaling during development by the modulation of cell surface expression of Fz receptor.


Asunto(s)
Membrana Celular/metabolismo , Receptores de Superficie Celular/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Animales , ADN Complementario/genética , Desarrollo Embrionario/genética , Retículo Endoplásmico/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Glicosilación , Humanos , Fenotipo , Unión Proteica , Transporte de Proteínas , Transcriptoma , Proteínas de Pez Cebra/genética
20.
Oncotarget ; 8(30): 48972-48982, 2017 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-28430665

RESUMEN

In this study, we investigated purinergic receptor P2X7 and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome expressions, and their role in head and neck cancer. We found upregulation of purinergic receptor P2X7 and all NLRP3 inflammasome components in biopsied head and neck squamous cell carcinoma tissues. Similarly, the expression of purinergic receptor P2X7, apoptosis-associated speck-like protein containing CARD, and pro-form caspase 1 in A253 cells derived from epidermoid carcinoma were highly upregulated in comparison to normal Human Salivary Gland cell line. Active caspase-1 and its final product, active interleukin-1ß, both increased in primed A253 cells stimulated with purinergic receptor P2X7 agonists, while this elevated NLRP3 inflammasome activity was suppressed by purinergic receptor P2X7 antagonists. However, we observed none of these effects in Human Salivary Gland cells. Inhibition of both NLRP3 inflammasome and purinergic receptor P2X7 led to the significant cell death of primed A253 cells, but had no effect on the viability of primed HSG cells or the primary cultured human fibroblast cells. Furthermore, inhibition of either purinergic receptor P2X7 or NLRP3 inflammasome decreased invasiveness of A253, and this effect became more evident when both purinergic receptor P2X7 and NLRP3 inflammasome were simultaneously blocked. Therefore, it is concluded that the purinergic receptor P2X7 and the activation of NLRP3 inflammasome play important roles in the survival and invasiveness of head and neck squamous cell carcinoma in humans.


Asunto(s)
Neoplasias de Cabeza y Cuello/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Anciano , Biomarcadores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Femenino , Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Masculino , Persona de Mediana Edad , Agonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Purinérgicos P2X7/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...