Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Am J Cancer Res ; 13(4): 1209-1239, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37168336

RESUMO

Nuclear epidermal growth factor receptor (EGFR) has been shown to be correlated with drug resistance and a poor prognosis in patients with cancer. Previously, we have identified a tripartite nuclear localization signal (NLS) within EGFR. To comprehensively determine the functions and underlying mechanism of nuclear EGFR and its clinical implications, we aimed to explore the nuclear export signal (NES) sequence of EGFR that is responsible for interacting with the exportins. We combined in silico prediction with site-directed mutagenesis approaches and identified a putative NES motif of EGFR, which is located in amino acid residues 736-749. Mutation at leucine 747 (L747) in the EGFR NES led to increased nuclear accumulation of the protein via a less efficient release of the exportin CRM1. Interestingly, L747 with serine (L747S) and with proline (L747P) mutations were found in both tyrosine kinase inhibitor (TKI)-treated and -naïve patients with lung cancer who had acquired or de novo TKI resistance and a poor outcome. Reconstituted expression of the single NES mutant EGFRL747P or EGFRL747S, but not the dual mutant along with the internalization-defective or NLS mutation, in lung cancer cells promoted malignant phenotypes, including cell migration, invasiveness, TKI resistance, and tumor initiation, supporting an oncogenic role of nuclear EGFR. Intriguingly, cells with germline expression of the NES L747 mutant developed into B cell lymphoma. Mechanistically, nuclear EGFR signaling is required for sustaining nuclear activated STAT3, but not for Erk. These findings suggest that EGFR functions are compartmentalized and that nuclear EGFR signaling plays a crucial role in tumor malignant phenotypes, leading to tumorigenesis in human cancer.

2.
J Biomed Sci ; 29(1): 86, 2022 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-36284291

RESUMO

Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) exploit the concept of synthetic lethality and offer great promise in the treatment of tumors with deficiencies in homologous recombination (HR) repair. PARPi exert antitumor activity by blocking Poly(ADP-ribosyl)ation (PARylation) and trapping PARP1 on damaged DNA. To date, the U.S. Food and Drug Administration (FDA) has approved four PARPi for the treatment of several cancer types including ovarian, breast, pancreatic and prostate cancer. Although patients with HR-deficient tumors benefit from PARPi, majority of tumors ultimately develop acquired resistance to PARPi. Furthermore, even though BRCA1/2 mutations are commonly used as markers of PARPi sensitivity in current clinical practice, not all patients with BRCA1/2 mutations have PARPi-sensitive disease. Thus, there is an urgent need to elucidate the molecular mechanisms of PARPi resistance to support the development of rational effective treatment strategies aimed at overcoming resistance to PARPi, as well as reliable biomarkers to accurately identify patients who will most likely benefit from treatment with PARPi, either as monotherapy or in combination with other agents, so called marker-guided effective therapy (Mget). In this review, we summarize the molecular mechanisms driving the efficacy of and resistance to PARPi as well as emerging therapeutic strategies to overcome PARPi resistance. We also highlight the identification of potential markers to predict PARPi resistance and guide promising PARPi-based combination strategies.


Assuntos
Neoplasias , Neoplasias Ovarianas , Humanos , Feminino , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico , Proteína BRCA1/genética , Ribose/uso terapêutico , Neoplasias/tratamento farmacológico , Biomarcadores , Difosfato de Adenosina/uso terapêutico , DNA/uso terapêutico , Neoplasias Ovarianas/genética
3.
Nat Cancer ; 3(10): 1211-1227, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36253486

RESUMO

Poly(ADP-ribose) polymerase (PARP) inhibitors have demonstrated promising clinical activity in multiple cancers. However, resistance to PARP inhibitors remains a substantial clinical challenge. In the present study, we report that anaplastic lymphoma kinase (ALK) directly phosphorylates CDK9 at tyrosine-19 to promote homologous recombination (HR) repair and PARP inhibitor resistance. Phospho-CDK9-Tyr19 increases its kinase activity and nuclear localization to stabilize positive transcriptional elongation factor b and activate polymerase II-dependent transcription of HR-repair genes. Conversely, ALK inhibition increases ubiquitination and degradation of CDK9 by Skp2, an E3 ligase. Notably, combination of US Food and Drug Administration-approved ALK and PARP inhibitors markedly reduce tumor growth and improve survival of mice in PARP inhibitor-/platinum-resistant tumor xenograft models. Using human tumor biospecimens, we further demonstrate that phosphorylated ALK (p-ALK) expression is associated with resistance to PARP inhibitors and positively correlated with p-Tyr19-CDK9 expression. Together, our findings support a biomarker-driven, combinatorial treatment strategy involving ALK and PARP inhibitors to induce synthetic lethality in PARP inhibitor-/platinum-resistant tumors with high p-ALK-p-Tyr19-CDK9 expression.


Assuntos
Quinase do Linfoma Anaplásico , Antineoplásicos , Neoplasias da Mama , Quinase 9 Dependente de Ciclina , Animais , Feminino , Humanos , Camundongos , Quinase do Linfoma Anaplásico/metabolismo , Antineoplásicos/farmacologia , Biomarcadores , Neoplasias da Mama/tratamento farmacológico , Quinase 9 Dependente de Ciclina/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Fator B de Elongação Transcricional Positiva , Tirosina/química , Tirosina/metabolismo , Ubiquitina-Proteína Ligases/efeitos dos fármacos , Ubiquitina-Proteína Ligases/metabolismo , Estados Unidos
4.
Cancer Res ; 82(11): 2185-2195, 2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35385574

RESUMO

Targeting immune checkpoints such as programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) has transformed cancer treatment, with durable clinical responses across a wide range of tumor types. However, a high percentage of patients fail to respond to anti-PD-1/PD-L1 treatment. A greater understanding of PD-L1 regulation is critical to improving the clinical response rate of PD-1/PD-L1 blockade. Here, we demonstrate that PD-L1 is phosphorylated and stabilized by casein kinase 2 (CK2) in cancer and dendritic cells (DC). Phosphorylation of PD-L1 at Thr285 and Thr290 by CK2 disrupted PD-L1 binding with speckle-type POZ protein, an adaptor protein of the cullin 3 (CUL3) ubiquitin E3 ligase complex, protecting PD-L1 from CUL3-mediated proteasomal degradation. Inhibition of CK2 decreased PD-L1 protein levels by promoting its degradation and resulted in the release of CD80 from DC to reactivate T-cell function. In a syngeneic mouse model, combined treatment with a CK2 inhibitor and an antibody against T-cell immunoglobulin mucin-3 (Tim-3) suppressed tumor growth and prolonged survival. These findings uncover a mechanism by which PD-L1 is regulated and suggest a potential antitumor treatment option to activate DC function by blocking the CK2-PD-L1 pathway and inhibiting Tim-3. SIGNIFICANCE: This work identifies a role for CK2 in immunosuppression by phosphorylation and stabilization of PD-L1, identifying CK2 inhibition as an immunotherapeutic approach for treating cancer.


Assuntos
Antígeno B7-H1 , Caseína Quinase II , Neoplasias , Animais , Caseína Quinase II/metabolismo , Células Dendríticas/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Camundongos , Fosforilação , Receptor de Morte Celular Programada 1/metabolismo
5.
J Biol Chem ; 298(4): 101817, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35278434

RESUMO

Expression of the receptor tyrosine kinase ephrin receptor A10 (EphA10), which is undetectable in most normal tissues except for the male testis, has been shown to correlate with tumor progression and poor prognosis in several malignancies, including triple-negative breast cancer (TNBC). Therefore, EphA10 could be a potential therapeutic target, likely with minimal adverse effects. However, no effective clinical drugs against EphA10 are currently available. Here, we report high expression levels of EphA10 in tumor regions of breast, lung, and ovarian cancers as well as in immunosuppressive myeloid cells in the tumor microenvironment. Furthermore, we developed anti-EphA10 monoclonal antibodies (mAbs) that specifically recognize cell surface EphA10, but not other EphA family isoforms, and target tumor regions precisely in vivo with no apparent accumulation in other organs. In syngeneic TNBC mouse models, we found that anti-EphA10 mAb clone #4 enhanced tumor regression, therapeutic response rate, and T cell-mediated antitumor immunity. Notably, the chimeric antigen receptor T cells derived from clone #4 significantly inhibited TNBC cell viability in vitro and tumor growth in vivo. Together, our findings suggest that targeting EphA10 via EphA10 mAbs and EphA10-specific chimeric antigen receptor-T cell therapy may represent a promising strategy for patients with EphA10-positive tumors.


Assuntos
Anticorpos Monoclonais , Receptores de Antígenos Quiméricos , Receptores da Família Eph , Linfócitos T , Neoplasias de Mama Triplo Negativas , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Camundongos , Receptores da Família Eph/imunologia , Linfócitos T/metabolismo , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Biol Chem ; 298(4): 101821, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35283189

RESUMO

Antibodies that target immune checkpoint proteins such as programmed cell death protein 1, programmed death ligand 1, and cytotoxic T-lymphocyte-associated antigen 4 in human cancers have achieved impressive clinical success; however, a significant proportion of patients fail to respond to these treatments. Galectin-9 (Gal-9), a ß-galactoside-binding protein, has been shown to induce T-cell death and facilitate immunosuppression in the tumor microenvironment by binding to immunomodulatory receptors such as T-cell immunoglobulin and mucin domain-containing molecule 3 and the innate immune receptor dectin-1, suggesting that it may have potential as a target for cancer immunotherapy. Here, we report the development of two novel Gal-9-neutralizing antibodies that specifically react with the N-carbohydrate-recognition domain of human Gal-9 with high affinity. We also show using cell-based functional assays that these antibodies efficiently protected human T cells from Gal-9-induced cell death. Notably, in a T-cell/tumor cell coculture assay of cytotoxicity, these antibodies significantly promoted T cell-mediated killing of tumor cells. Taken together, our findings demonstrate potent inhibition of human Gal-9 by neutralizing antibodies, which may open new avenues for cancer immunotherapy.


Assuntos
Anticorpos Neutralizantes , Morte Celular , Galectinas , Linfócitos T , Anticorpos Neutralizantes/metabolismo , Anticorpos Neutralizantes/farmacologia , Morte Celular/efeitos dos fármacos , Galectinas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/terapia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Microambiente Tumoral
7.
STAR Protoc ; 3(1): 101198, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35243381

RESUMO

Immunotherapy via PD-1/PD-L1 blockade is a promising strategy to eradicate cancer cells. However, the PD-L1 pathological level is inconsistent with the therapeutic response and is not a reliable biomarker to stratify patients for anti-PD-1/PD-L1 therapy. Here, we describe patient sample deglycosylation in an immunohistochemistry (IHC) assay to resolve this challenge. This protocol facilitates antigen retrieval by removing N-glycans from surface antigens on formalin-fixed paraffin-embedded (FFPE) tissue slides and can be applied in medical pathology for multiple cancer types. For complete details on the use and execution of this profile, please refer to Lee et al. (2019).


Assuntos
Antígeno B7-H1 , Neoplasias , Humanos , Imuno-Histoquímica , Imunoterapia , Neoplasias/terapia
8.
Am J Cancer Res ; 11(1): 236-250, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33520371

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer that lack effective therapeutic strategies. The response rate of PDAC for treatment with gemcitabine, a current first-line chemotherapeutic for this tumor, is lower than 20%. Identifying key targetable molecules that mediate gemcitabine resistance and developing novel strategies for precision PDAC medicine are urgently needed. Most PDACs have either intratumoral hypoxia or high reactive oxygen species (ROS) production; cytotoxic chemotherapy can elevate ROS production in PDACs. Although excessive ROS production leads to oxidative damage of macromolecules such as DNA, pancreatic cancer cells can survive high DNA damage stress levels. Therefore, identifying molecular mechanisms of overcoming ROS-induced stress in pancreatic cancer cells is important for developing novel therapeutic strategies. ROS-induced DNA damage is predominantly repaired via poly (ADP-ribose) polymerase 1 (PARP1)-mediated DNA repair mechanisms. A recent clinical trial reported that PARP inhibitors are effective in treating pancreatic patients carrying BRCA mutations. However, only less than 10% of pancreatic cancer patients bearing BRCA mutated tumors. Activation of the receptor tyrosine kinase c-MET positively correlates with poor prognosis for PDAC, and our previous study showed that nuclear c-MET can phosphorylate PARP1 at tyrosine 907 under ROS stimulation to promote DNA repair. As described herein, we proposed to expand PARP inhibitor-targeted therapy to more pancreatic cancer patients regardless of BRCA mutation status by combining olaparib, a PARP inhibitor, with c-MET inhibitors as we demonstrated in our previous studies in breast cancer. In this prospective study, we found that ROS-inducing chemotherapeutic drugs such as gemcitabine and doxorubicin stimulated nuclear accumulation of c-MET in BxPC-3 and L3.6pl pancreatic cancer cells. We further showed that combining a c-MET inhibitor with gemcitabine or a PARP inhibitor induced more DNA damage than monotherapy did. Moreover, we demonstrated the synergistic antitumor effects of c-MET inhibitors combined with a PARP inhibitor or gemcitabine in eliminating pancreatic cancer cells. These data suggested that accumulation of ROS in pancreatic cancer cells promotes nuclear localization of c-MET, resulting in resistance to both chemotherapy and PARP inhibitors. Our findings suggest that combining c-MET inhibitors with PARP inhibitors or gemcitabine is a novel, rational therapeutic strategy for advanced pancreatic cancer.

9.
Am J Cancer Res ; 10(4): 1194-1206, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32368395

RESUMO

The cyclin-dependent kinase 2 (CDK2) inhibitor dinaciclib, a potential anti-cancer drug, has been tested in clinical trials and reported to suppress tumor initiating cells. Our recent study demonstrated that pharmacological inhibition of CDK2 or enhancer of zeste homolog 2 (EZH2) allows re-expression of ERα and converts triple-negative breast cancers (TNBC) to luminal ERα-positive, rendering TNBC cells targetable by tamoxifen. Like TNBC, EZH2 is also commonly overexpressed in ovarian cancers, and overexpression of cyclin E1 gene (CCNE1) and/or amplification of its associated kinase CDK2 gene is present in ovarian tumor specimens, both of which are associated with primary treatment resistance and poor outcome in high-grade serous ovarian cancer (HGSC). We determined whether inhibition of CDK2-mediated phosphorylation of EZH2 activates ERα expression in ERα-negative HGSOC cells, rendering them targetable by hormonal therapy. The specific CDK2 inhibitor repressed phosphorylation of EZH2 at T416, and in turn activated the expression of its downstream target ERα gene (ESR1). We tested the efficacy of the combination of CDK2 inhibitor and tamoxifen and found significant synergistic inhibition. We further demonstrated that CDK2 inhibitor is a more promising agent than EZH2 inhibitor in repressing TNBC and HGSOC due to a feedback increase in CDK2 activity by EZH2 inhibitor. Our results indicated that the combination treatment of CDK2 inhibitor and tamoxifen has the potential to benefit patients with ERα-negative HGSOC.

10.
Am J Cancer Res ; 10(2): 648-661, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32195033

RESUMO

The limited treatment options and therapeutic failure due to acquired resistance for patients with triple-negative breast cancer (TNBC) represent a significant challenge. Inhibitors against poly (ADP-ribose) polymerase (PARP), olaparib and talazoparib, were recently approved for the treatment of metastatic breast cancer (including TNBC) in patients with germline BRCA1/2 mutations. Despite impressive response rates of ~60%, the prolongation in median progression-free survival with a PARPi is modest, suggesting the emergence of resistance. Several studies have reported that receptor tyrosine kinases (RTKs), such as c-MET (also known as hepatocyte growth factor receptor), are involved in resistance to various anti-neoplastic agents, including PARPi. However, the mechanism by which c-MET contributes to acquired resistance to PARPi in TNBC is not fully understood. In this study, we show that hyperactivated c-Met is detected in TNBC cells with acquired resistance to PARPi, and the combination of talazoparib and crizotinib (a multi-kinase inhibitor that inhibits c-MET) synergistically inhibits proliferation in these cells. Unexpectedly, depleting c-MET had limited effect on talazoparib sensitivity in PARPi-resistant cells. Interestingly, we found evidence of epidermal growth factor receptor (EGFR) hyperactivation and interaction of EGFR/c-Met in these cells. Notably, combining EGFR and PARP inhibitors resulted in greater inhibition of proliferation in c-MET-depleted TNBC cells, and combined c-MET and EGFR inhibition increased sensitivity to talazoparib in TNBC cells with acquired resistance to PARPi. Our findings suggest that combined inhibition of c-MET and EGFR could potentially re-sensitize TNBC to the cytotoxic effects of PARPi.

12.
Nat Commun ; 10(1): 5114, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31704972

RESUMO

Triple-negative breast cancer (TNBC), which lacks estrogen receptor α (ERα), progesterone receptor, and human epidermal growth factor receptor 2 (HER2) expression, is closely related to basal-like breast cancer. Previously, we and others report that cyclin E/cyclin-dependent kinase 2 (CDK2) phosphorylates enhancer of zeste homolog 2 (EZH2) at T416 (pT416-EZH2). Here, we show that transgenic expression of phospho-mimicking EZH2 mutant EZH2T416D in mammary glands leads to tumors with TNBC phenotype. Coexpression of EZH2T416D in mammary epithelia of HER2/Neu transgenic mice reprograms HER2-driven luminal tumors into basal-like tumors. Pharmacological inhibition of CDK2 or EZH2 allows re-expression of ERα and converts TNBC to luminal ERα-positive, rendering TNBC cells targetable by tamoxifen. Furthermore, the combination of either CDK2 or EZH2 inhibitor with tamoxifen effectively suppresses tumor growth and markedly improves the survival of the mice bearing TNBC tumors, suggesting that the mechanism-based combination therapy may be an alternative approach to treat TNBC.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Receptor alfa de Estrogênio/efeitos dos fármacos , Neoplasias Mamárias Experimentais/genética , Neoplasias de Mama Triplo Negativas/genética , Animais , Benzamidas/farmacologia , Compostos de Bifenilo , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Óxidos N-Cíclicos , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Indolizinas , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Transgênicos , Morfolinas , Fosforilação , Compostos de Piridínio/farmacologia , Piridonas/farmacologia , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Progesterona/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo
13.
Am J Cancer Res ; 9(3): 608-618, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949414

RESUMO

Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) are promising targeted therapeutics for breast and ovarian cancers bearing a germline BRCA1/2 mutation (BRCA m), and several have already received regulatory approval in the United States. In patients with a BRCA m cancer, PARPi can increase the burden of unrepaired DNA double-strand breaks by blocking PARP activity and trapping PARP1 onto damaged DNA. Resistance to PARP inhibitors can block the formation of DNA double-strand breaks through BRCA-related DNA repair pathway. MET is a hyper-activated receptor tyrosine kinase expressed in multiple cancer types and the activation contributes to resistance to DNA damage-inducing therapeutic drugs. Our previous study showed that MET inhibition by pan-kinase inhibitors has synergism with PARPi in suppressing growth of breast cancer in vitro and in xenograft tumor models. In this study, we validated the inhibitory effect of novel inhibitors, HS10241 (selective MET inhibitor) and HS10160 (PARPi), to their target respectively in triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSOC) cells. We further demonstrated that these two inhibitors function synergistically in eliminating TNBC and HGSOC cells; combining with HS10241 increased DNA double-strand breaks induced by HS10160 in cancer cells; and PARP1 tyrosine (Y)-907 phosphorylation (PARP1 p-Y907) can be an effective biomarker as an indicator of MET-mediated PARPi in HGSOC. Our results suggest that the combination of HS10241 and HS10160 may benefit patients bearing tumors overexpressing MET as well as those resistant to single-agent PARPi treatment.

14.
Cancer Res ; 79(4): 819-829, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30573522

RESUMO

PARP1 inhibitors (PARPi) are currently used in the clinic for the treatment of ovarian and breast cancers, yet their therapeutic efficacy against hepatocellular carcinoma (HCC) has been disappointing. To ensure therapeutic efficacy of PARPi against HCC, a disease often diagnosed at intermediate to advanced stages with no effective treatment options, it is critical to identify not only biomarkers to predict PARPi resistance but also rational treatments to overcome this. Here, we report that a heterodimer of EGFR and MET interacts with and phosphorylates Y907 of PARP1 in the nucleus, which contributes to PARPi resistance. Inhibition of both EGFR and MET sensitized HCC cells to PARPi, and both EGFR and MET are known to be overexpressed in HCC. This report provides an explanation for the poor efficacy of PARPi against HCC and suggests combinatorial treatment consisting of EGFR, MET, and PARP inhibitors may be an effective therapeutic strategy in HCC. SIGNIFICANCE: Regulation of PARP by the c-MET and EGFR heterodimer suggests a potentially effective combination therapy to sensitize HCC to PARPi.


Assuntos
Carcinoma Hepatocelular/patologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Hepáticas/patologia , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Animais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Proto-Oncogênicas c-met/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cell Death Dis ; 8(11): e3167, 2017 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-29120412

RESUMO

MicroRNAs (miRNAs) have been suggested to repress transcription via binding the 3'-untranslated regions of mRNAs. However, the involvement and details of miRNA-mediated epigenetic regulation, particularly in targeting genomic DNA and mediating epigenetic regulation, remain largely uninvestigated. In the present study, transcription factor CCAAT/enhancer binding protein delta (CEBPD) was responsive to the anticancer drug bortezomib, a clinical and highly selective drug for leukemia treatment, and contributed to bortezomib-induced cell death. Interestingly, following the identification of CEBPD-induced miRNAs, we found that miR-744, miR-3154 and miR-3162 could target CpG islands in the 5'-flanking region of the CEBPD gene. We previously demonstrated that the Yin Yang 1 (YY1)/polycomb group (PcG) protein/DNA methyltransferase (DNMT) complex is important for CCAAT/enhancer binding protein delta (CEBPD) gene inactivation; we further found that Argonaute 2 (Ago2) interacts with YY1 and binds to the CEBPD promoter. The miRNA/Ago2/YY1/PcG group protein/DNMT complex linked the inactivation of CEBPD and genes adjacent to its 5'-flanking region, including protein kinase DNA-activated catalytic polypeptide (PRKDC), minichromosome maintenance-deficient 4 (MCM4) and ubiquitin-conjugating enzyme E2 variant 2 (UBE2V2), upon bortezomib treatment. Moreover, we revealed that miRNA binding is necessary for YY1/PcG group protein/DNMT complex-mediated epigenetic gene silencing and is associated with bortezomib-induced methylation on genomic DNA. The present study successfully characterized the interactions of the miRNA/Ago2/YY1/PcG group protein/DNMT complex and provided new insights for miRNA-mediated epigenetic regulation in bortezomib-induced leukemic cell arrest and cell death.


Assuntos
Apoptose/efeitos dos fármacos , Bortezomib/farmacologia , Leucemia/fisiopatologia , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Antineoplásicos/farmacologia , Proteínas Argonautas/química , Proteínas Argonautas/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/genética , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA/efeitos dos fármacos , Proteína Quinase Ativada por DNA/genética , Proteína Quinase Ativada por DNA/metabolismo , Inativação Gênica , Humanos , Leucemia/metabolismo , Ligases/genética , Ligases/metabolismo , MicroRNAs/genética , Componente 4 do Complexo de Manutenção de Minicromossomo/genética , Componente 4 do Complexo de Manutenção de Minicromossomo/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Transcrição Gênica/efeitos dos fármacos , Enzimas de Conjugação de Ubiquitina , Fator de Transcrição YY1/química , Fator de Transcrição YY1/metabolismo
16.
Clin Cancer Res ; 23(2): 503-513, 2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27435393

RESUMO

PURPOSE: Cisplatin (CDDP) is frequently used in combination chemotherapy with paclitaxel for treating urothelial carcinoma of the urinary bladder (UCUB). CDDP cross-resistance has been suggested to develop with paclitaxel, thus hindering successful UCUB treatment. Therefore, elucidating the mechanisms underlying CDDP-induced anticancer drug resistance is imperative and may provide an insight in developing novel therapeutic strategy. EXPERIMENTAL DESIGN: Loss-of-function assays were performed to elucidate the role of the EGFR and STAT3 in CDDP-induced CCAAT/enhancer-binding protein delta (CEBPD) expression in UCUB cells. Reporter and in vivo DNA-binding assays were employed to determine whether CEBPD directly regulates ATP binding cassette subfamily B member 1 (ABCB1) and ATP binding cassette subfamily C member 2 (ABCC2) activation. Finally, a xenograft animal assay was used to examine the abilities of gefitinib and S3I-201 (a STAT3 inhibitor) to reverse CDDP and paclitaxel sensitivity. RESULTS: CEBPD expression was maintained in postoperative chemotherapy patients, and this expression was induced by CDDP even in CDDP-resistant UCUB cells. Upon CDDP treatment, CEBPD activated ABCB1 and ABCC2. Furthermore, the EGFR/STAT3 pathway contributed to CDDP-induced CEBPD expression in UCUB cells. Gefitinib and S3I-201 treatment significantly reduced the expression of CEBPD and enhanced the sensitivity of CDDP-resistant UCUB cells to CDDP and paclitaxel. CONCLUSIONS: Our results revealed the risk of CEBPD activation in CDDP-resistant UCUB cells and suggested a therapeutic strategy for patients with UCUB or UCUB resisted to CDDP and paclitaxel by combination with either gefitinib or S3I-201. Clin Cancer Res; 23(2); 503-13. ©2016 AACR.


Assuntos
Proteína delta de Ligação ao Facilitador CCAAT/genética , Carcinoma/tratamento farmacológico , Receptores ErbB/genética , Fator de Transcrição STAT3/genética , Neoplasias da Bexiga Urinária/tratamento farmacológico , Ácidos Aminossalicílicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Benzenossulfonatos/administração & dosagem , Carcinoma/genética , Carcinoma/patologia , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Cisplatino/efeitos adversos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Gefitinibe , Regulação Neoplásica da Expressão Gênica , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Paclitaxel/administração & dosagem , Quinazolinas/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Urotélio/efeitos dos fármacos , Urotélio/patologia
17.
Sci Rep ; 6: 32000, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27561946

RESUMO

Infection by the dengue virus (DENV) threatens global public health due to its high prevalence and the lack of effective treatments. Host factors may contribute to the pathogenesis of DENV; herein, we investigated the role of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), which is activated by DENV in mononuclear phagocytes. DENV infection selectively activates Nrf2 following nuclear translocation. Following endoplasmic reticular (ER) stress, protein kinase R-like ER kinase (PERK) facilitated Nrf2-mediated transcriptional activation of C-type lectin domain family 5, member A (CLEC5A) to increase CLEC5A expression. Signaling downstream of the Nrf2-CLEC5A interaction enhances Toll-like receptor 3 (TLR3)-independent tumor necrosis factor (TNF)-α production following DENV infection. Forced expression of the NS2B3 viral protein induces Nrf2 nuclear translocation/activation and CLEC5A expression which increases DENV-induced TNF-α production. Animal studies confirmed Nrf2-induced CLEC5A and TNF-α in brains of DENV-infected mice. These results demonstrate that DENV infection causes Nrf2-regulated TNF-α production by increasing levels of CLEC5A.


Assuntos
Vírus da Dengue/fisiologia , Lectinas Tipo C/metabolismo , Monócitos/virologia , Fator 2 Relacionado a NF-E2/fisiologia , Receptores de Superfície Celular/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Cricetinae , Humanos , Camundongos , Monócitos/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo , Ativação Transcricional , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
18.
Mol Neurobiol ; 53(6): 4173-4188, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26208701

RESUMO

The progression of Alzheimer's disease (AD) has been associated with astrocytes-induced neuroinflammation. However, the detailed mechanism of astrocytes associated with learning impairments and neuronal loss in AD is poorly defined. Here, we provide novel evidences that astrocytic miR-135a is critical for neuronal viability and spatial learning ability in vivo. The AppTg/Cebpd (-/-) mice showed a spatial learning improvement compared with the APPswe/PS1/E9 bigenic (AppTg) mice. miR-135a was found to be a CCAAT/enhancer binding protein δ (CEBPD) responsive miRNA and can repress the transcription of thrombospondin 1 (THBS1) / Thbs1 (mouse) via its 3'-untranslated region (3'UTR). We used different experimental approaches to attenuate the expression of CEBPD/Cebpd (mouse) or miR-135a in astrocytes and found the following results: increase in THBS1/Thbs1 expression, decrease in neuronal apoptosis, and increase in growth of neurites. Importantly, injection of miR-135a antagonist (AM135a) into the brain of AppTg mice was found to prevent neuronal apoptosis and improved the spatial learning ability. Together, our findings demonstrate a critical function for the astrocytic CEBPD, and point to miR-135a antagonist as an attractive therapeutic target for the treatment of Alzheimer's disease.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Proteína delta de Ligação ao Facilitador CCAAT/metabolismo , MicroRNAs/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Aprendizagem Espacial , Regiões 3' não Traduzidas/genética , Animais , Antagomirs/farmacologia , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , MicroRNAs/genética , Crescimento Neuronal/efeitos dos fármacos , Aprendizagem Espacial/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos
19.
J Leukoc Biol ; 99(1): 131-41, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26342105

RESUMO

Acute myeloid leukemia is the majority type presented in leukemia patients. Forcing malignant cells to undergo differentiation is 1 strategy for acute myeloid leukemia therapy. However, the failure of acute myeloid leukemia patients to achieve remission as a result of drug resistance remains a challenge. In this study, we found that the abundances of the proinflammatory cytokine IL-18 and its receptor (IL-18R) correlated with the occurrence of drug resistance in AML patients during standard treatment. Cyclooxygenase 2 (COX-2) has been suggested to have an antiapoptotic role in chemoresistant cancer cells. IL-18 treatment resulted in an increase in COX-2 expression through the post-transcriptional regulation of COX-2 mRNA in differentiated U937 cells and showed antiapoptotic activity in U937 and THP-1 cells. Two RNA-binding proteins, human antigen R and insulin-like growth factor mRNA-binding protein 3, mediated the stabilization of COX-2 mRNA. IL-18 induced the shuttling of human antigen R and insulin-like growth factor mRNA-binding protein 3 from the nucleus to the cytoplasm and facilitated their interaction; subsequently, this complex bound to the 3' untranslated region of COX-2 mRNA and affected its stability. We demonstrated further that JNK and/or ERK1/2 regulated human antigen R nucleocytoplasmic shuttling, mediating IL-18 stabilization of cyclooxygenase 2 mRNA.


Assuntos
Ciclo-Oxigenase 2/genética , Proteína Semelhante a ELAV 1/metabolismo , Interleucina-18/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular Tumoral , Humanos , Interleucina-18/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Ligação Proteica , Processamento Pós-Transcricional do RNA , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ativação Transcricional
20.
Sci Rep ; 5: 15659, 2015 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-26493689

RESUMO

TIM-3 functions to enforce CD8+ T cell exhaustion, a dysfunctional state associated with the tolerization of tumor microenvironment. Here we report apoptosis of IFN-γ competent TIM-3+ population of tumor-infiltrating CD8+ T cells in colon cancer. In humans suffering from colorectal cancer, TIM-3+ population is higher in cancer tissue-resident relative to peripheral blood CD8+ T cells. Both the TIM-3+ and TIM-3- cancer tissue-resident CD8+ T cells secrete IFN-γ of comparable levels, although apoptotic cells are more in TIM-3+ compared to TIM-3- population. In mouse CT26 colon tumor model, majority of tumor-infiltrating CD8+ T cells express TIM-3 and execute cytolysis function with higher effector cytokine secretion and apoptosis in TIM-3+ compared to TIM-3- population. The tumor cells secrete galectin-9, which increases apoptosis of tumor-infiltrating CD8+ T cells. Galectin-9/TIM-3 signaling blockade with anti-TIM-3 antibody reduces the apoptosis and in addition, inhibits tumor growth in mice. The blockade increases therapeutic efficacy of cyclophosphamide to treat tumor in mice as well. These results reveal a previously unexplored role of TIM-3 on tumor-infiltrating CD8+ T cells in vivo.


Assuntos
Apoptose , Linfócitos T CD8-Positivos/imunologia , Neoplasias do Colo/imunologia , Proteínas de Membrana/imunologia , Neoplasias do Colo/patologia , Galectinas/fisiologia , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Proteínas de Membrana/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA