Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 671
Filtrar
1.
Cells ; 10(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34943938

RESUMO

TMEM176B is a member of the membrane spanning 4-domains (MS4) family of transmembrane proteins, and a putative ion channel that is expressed in immune cells and certain cancers. We aimed to understand the role of TMEM176B in cancer cell signaling, gene expression, cell proliferation, and migration in vitro, as well as tumor growth in vivo. We generated breast cancer cell lines with overexpressed and silenced TMEM176B, and a therapeutic antibody targeting TMEM176B. Proliferation and migration assays were performed in vitro, and tumor growth was evaluated in vivo. We performed gene expression and Western blot analyses to identify the most differentially regulated genes and signaling pathways in cells with TMEM176B overexpression and silencing. Silencing TMEM176B or inhibiting it with a therapeutic antibody impaired cell proliferation, while overexpression increased proliferation in vitro. Syngeneic and xenograft tumor studies revealed the attenuated growth of tumors with TMEM176B gene silencing compared with controls. We found that the AKT/mTOR signaling pathway was activated or repressed in cells overexpressing or silenced for TMEM176B, respectively. Overall, our results suggest that TMEM176B expression in breast cancer cells regulates key signaling pathways and genes that contribute to cancer cell growth and progression, and is a potential target for therapeutic antibodies.


Assuntos
Proteínas de Membrana/genética , Proteína Oncogênica v-akt/genética , Serina-Treonina Quinases TOR/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Animais , Antígeno CD24/genética , Antígeno CD24/imunologia , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Xenoenxertos , Humanos , Camundongos , RNA-Seq , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Neoplasias de Mama Triplo Negativas/patologia
2.
Cancer Lett ; 520: 409-421, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34419501

RESUMO

Bcl2-associated athanogene 4 (BAG4) has been found to be aberrantly expressed in several types of human cancers. However, little is known about its expression, role, and clinical significance in gastric cancer (GC). In this study, we aimed to address these issues and to explore the underlying mechanisms. The expression level of BAG4, measured by immunohistochemistry, was significantly higher in GC tissues than in paired normal tissues. Elevated BAG4 expression was positively correlated with T stage, lymph node metastasis, and tumor size of GC and was associated with unfavorable outcomes of the patients. The overexpression of BAG4 promoted the in vitro invasion and in vivo metastasis of GC cells, and opposite results were observed after silencing of BAG4. Silencing of BAG4 significantly reduced the phosphorylation of PI3K, AKT, and p65, whereas overexpression of BAG4 markedly enhanced the phosphorylation of these molecules. At the same time, manipulating BAG4 expression resulted in the corresponding changes in p65 nuclear translocation and ZEB1 expression. Luciferase reporter and chromatin immunoprecipitation assays verified that p65 binds to the promoter of ZEB1 to upregulate its transcription. Our results demonstrate that BAG4 plays an oncogenic role in the invasion and metastasis of GC cells by activating the PI3K/AKT/NF-κB/ZEB1 axis to induce epithelial-mesenchymal transition.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Neoplasias Gástricas/genética , Fator de Transcrição RelA/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , NF-kappa B/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Neoplasias Gástricas/patologia
3.
Exp Mol Pathol ; 122: 104673, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34371011

RESUMO

Recent findings have demonstrated a synergic crosstalk between SHH/GLI and PI3K/Akt/mTORC1 signaling in glioblastoma progression cells in vitro and in tumors in mice, but it is not known if this also occurs in human gliomas. We then aimed to investigate the expression of key proteins of these pathways in different human gliomas. The expression of PTEN, phospho-Akt (Ser473), phospho-S6K1 (Thr389), SHH, GLI1, GLI2 and GLI3 was assessed by immunohistochemistry in gliomas and in control brain tissues. The pattern of expression of each protein was established according to glioma type, glioma grade and to cell type; the relative expression of each protein was used to perform statistical analyses. We found that the expression of proteins of both signaling pathways differs between normal brain and glioma tissues. For instance, normal astrocytes had a different protein expression pattern compared with reactive and tumoral astrocytes. Interestingly, we detected a recurrent pattern of expression of GLI3 in oligodendrocytes and of phospho-S6K1 in mitotic neoplastic cells. We also identified differences of cell signaling according to glioma type: oligodendrogliomas and ependymomas are related with the expression of SHH/GLI proteins. Finally, we detected that high grade gliomas statistically correlate with the expression of GLI1 and GLI2, and that GLI1, GLI2, phospho-Akt and phospho-S6K1 are more expressed in patients with less survival, suggesting that activation of these cell signaling influences glioma outcome and patient survival. In summary, our results show that proteins of PI3K/Akt/mTORC1 and SHH/GLI pathways are differentially expressed in human gliomas according to tumor type and grade, and suggest that the activation of these signaling networks is associated with glioma progression.


Assuntos
Glioma/genética , Proteínas Hedgehog/genética , PTEN Fosfo-Hidrolase/genética , Proteína GLI1 em Dedos de Zinco/genética , Adulto , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Glioma/patologia , Humanos , Fatores de Transcrição Kruppel-Like/genética , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Pessoa de Meia-Idade , Gradação de Tumores , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética
4.
CNS Neurosci Ther ; 27(11): 1300-1312, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34346167

RESUMO

AIMS: The neurotropic growth factor PDGF-BB was shown to have vital neurorestorative functions in various animal models of Parkinson's disease (PD). Previous studies indicated that the regenerative property of PDGF-BB contributes to the increased intensity of tyrosine hydroxylase (TH) fibers in vivo. However, whether PDGF-BB directly modulates the expression of TH, and the underlying mechanism is still unknown. We will carefully examine this in our current study. METHOD: MPTP-lesion mice received PDGF-BB treatment via intracerebroventricular (i.c.v) administration, and the expression of TH in different brain regions was assessed by RT-PCR, Western blot, and immunohistochemistry staining. The molecular mechanisms of PDGF-BB-mediated TH upregulation were examined by RT-PCR, Western blot, ChIP assay, luciferase reporter assay, and immunocytochemistry. RESULTS: We validated a reversal expression of TH in MPTP-lesion mice upon i.c.v administration of PDGF-BB for seven days. Similar effects of PDGF-BB-mediated TH upregulation were also observed in MPP+ -treated primary neuronal culture and dopaminergic neuronal cell line SH-SY5Y cells. We next demonstrated that PDGF-BB rapidly activated the pro-survival PI3K/Akt and MAPK/ERK signaling pathways, as well as the downstream CREB in SH-SY5Y cells. We further confirmed the significant induction of p-CREB in PDGF-BB-treated animals in vivo. Using a genetic approach, we demonstrated that the transcription factor CREB is critical for PDGF-BB-mediated TH expression. The activation and nucleus translocation of CREB were promoted in PDGF-BB-treated SH-SY5Y cells, and the enrichment of CREB on the promoter region of TH gene was also increased upon PDGF-BB treatment. CONCLUSION: Our data demonstrated that PDGF-BB directly regulated the expression of TH via activating the downstream Akt/ERK/CREB signaling pathways. Our finding will further support the therapeutic potential of PDGF-BB in PD, and provide the possibility that targeting PDGF signaling can be harnessed as an adjunctive therapy in PD in the future.


Assuntos
Becaplermina/farmacologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Becaplermina/administração & dosagem , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Feminino , Humanos , Imuno-Histoquímica , Injeções Intraventriculares , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Intoxicação por MPTP/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/administração & dosagem , Proteína Oncogênica v-akt/genética , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/patologia , Gravidez
5.
Genes (Basel) ; 12(7)2021 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203273

RESUMO

Circular RNAs (circRNAs), a class of covalently closed RNAs formed by a back-splicing reaction, have been involved in the regulation of diverse oncogenic processes. In this article we describe circVAMP3, a novel circular RNA overexpressed in RH4, a representative cell line of alveolar rhabdomyosarcoma. We demonstrated that circVAMP3 has a differential m6A pattern opposed to its linear counterpart, suggesting that the two isoforms can be differently regulated by such RNA modification. Moreover, we show how circVAMP3 depletion in alveolar rhabdomyosarcoma cells can impair cell cycle progression, through the alteration of the AKT-related pathways, pointing to this non-coding RNA as a novel regulator of the alveolar rhabdomyosarcoma progression and as a putative future therapeutic target.


Assuntos
Ciclo Celular/genética , RNA Circular/genética , RNA/genética , Rabdomiossarcoma Alveolar/genética , Divisão Celular/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Mioblastos/metabolismo , Proteína Oncogênica v-akt/genética , Splicing de RNA/genética , Rabdomiossarcoma Alveolar/patologia
6.
Neurochem Int ; 148: 105120, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34197898

RESUMO

Oxidative stress and mitochondrial dysfunction are now widely accepted as the major factors involved in the pathogenesis of Parkinson's disease (PD). Rotenone, a commonly used environmental toxin also reproduces these principle pathological features of PD. Hence, it is used frequently to induce experimental PD in cells and animals. In this study, we evaluated the neuroprotective effects of metformin against rotenone-induced toxicity in SH-SY5Y cells. Metformin treatment clearly rescued these cells from rotenone-mediated cell death via the reduction of the cytosolic and mitochondrial levels of reactive oxygen species and restoration of mitochondrial function. Furthermore, metformin upregulated PGC-1α, the master regulator of mitochondrial biogenesis and key antioxidant molecules, including glutathione and superoxide dismutase. We demonstrated that the drug exerted its cytoprotective effects by activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme-oxygenase (HO)-1 pathway, which in turn, is dependent on AKT activation by metformin. Thus, our results implicate that metformin provides neuroprotection against rotenone by inhibiting oxidative stress in the cells by inducing antioxidant system via upregulation of transcription mediated by Nrf2, thereby restoring the rotenone-induced mitochondrial dysfunction and energy deficit in the cells.


Assuntos
Hipoglicemiantes/farmacologia , Metformina/farmacologia , Doenças Mitocondriais/prevenção & controle , Fator 2 Relacionado a NF-E2/genética , Proteína Oncogênica v-akt/genética , Estresse Oxidativo/efeitos dos fármacos , Rotenona/antagonistas & inibidores , Rotenona/toxicidade , Transdução de Sinais/efeitos dos fármacos , Desacopladores/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Humanos , Espécies Reativas de Oxigênio/metabolismo
7.
Mamm Genome ; 32(3): 183-194, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33956176

RESUMO

The miR-302s/367 family has the ability to induce mouse and human somatic cell reprogramming into induced pluripotent stem cells (iPSCs), inhibit the proliferation of several types of cancer cells, and even cause cancer cell apoptosis. However, the functions of the miR-302s/367 family in other mammals have not been explored. In the present study, the effects of miR-302s/367 on reprogramming, proliferation, and apoptosis in sheep fetal fibroblasts (SFFs) were evaluated by the delivery of a plasmid vector containing synthetic precursor miRNAs into cells, followed by the induction of mature miR-302s/367 expression. The results showed that miR-302s/367 could not reprogram SFFs into iPSCs; however, they could inhibit both the proliferation and apoptosis of SFFs by targeting CDK2, E2F1, E2F2, and PTEN in the cell cycle and PI3K-Akt pathways. Based on our findings, a novel mechanism was proposed in which the miR-302s/367 family functions in both the proliferation and apoptosis of somatic cells in mammals, suggesting that caution is needed when using miR-302s/367 as therapeutic agent.


Assuntos
Apoptose/genética , Ciclo Celular/genética , Proliferação de Células/genética , Fibroblastos/metabolismo , Ovinos/genética , Animais , Feto/metabolismo , Fibroblastos/patologia , Humanos , Camundongos , MicroRNAs/genética , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/imunologia
8.
Neurochem Int ; 148: 105082, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34052296

RESUMO

Since the role of estrogen in postmenauposal-associated dementia is still debatable, this issue urges the search for other medications. Dimethyl fumarate (DMF) is a drug used for the treatment of multiple sclerosis and has shown a neuroprotective effect against other neurodegenerative diseases. Accordingly, the present study aimed to evaluate the effect of DMF on an experimental model of Alzheimer disease (AD) using D-galactose (D-Gal) administered to ovariectomized (OVX) rats, resembling a postmenopausal dementia paradigm. Adult 18-month old female Wistar rats were allocated into sham-operated and OVX/D-Gal groups that were either left untreated or treated with DMF for 56 days starting three weeks after sham-operation or ovariectomy. DMF succeeded to ameliorate cognitive (learning/short- and long-term memory) deficits and to enhance the dampened overall activity (NOR, Barnes-/Y-maze tests). These behavioral upturns were associated with increased intact neurons (Nissl stain) and a reduction in OVX/D-Gal-mediated hippocampal CA1 neurodegeneration and astrocyte activation assessed as GFAP immunoreactivity. Mechanistically, DMF suppressed the hippocampal contents of AD-surrogate markers; viz., apolipoprotein (APO)-E1, BACE1, Aß42, and hyperphosphorylated Tau. Additionally, DMF has augmented the neuroprotective parameters p-AKT, its downstream target CREB and BDNF. Besides, it activated AMPK, and enhanced SIRT-1, as well as antioxidant defenses (SOD, GSH). On the other hand, DMF inhibited the transcription factor NF-κB, IL-1ß, adiponectin/adiponectin receptor type (AdipoR)1, GSK-3ß, and MDA. Accordingly, in this postmenopausal AD model, DMF treatment by pursuing the adiponectin/AdipoR1, AMPK/SIRT-1, AKT/CREB/BDNF, AKT/GSK-3ß, and APO-E1 quartet hampered the associated tauo-/amyloidopathy and NF-κB-mediated oxidative/inflammatory responses to advance insights into its anti-amnesic effect.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Amiloidose/tratamento farmacológico , Fumarato de Dimetilo/farmacologia , Fármacos Neuroprotetores/farmacologia , Ovariectomia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tauopatias/tratamento farmacológico , Adiponectina/genética , Doença de Alzheimer/induzido quimicamente , Amiloidose/induzido quimicamente , Amiloidose/psicologia , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Feminino , Galactose , Glicogênio Sintase Quinase 3 beta/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/genética , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , Proteína Oncogênica v-akt/efeitos dos fármacos , Proteína Oncogênica v-akt/genética , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio , Tauopatias/induzido quimicamente , Tauopatias/psicologia
9.
Int J Biol Sci ; 17(7): 1808-1820, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33994864

RESUMO

Increasing evidence reveals that the Rho GTPase-activating protein is a crucial negative regulator of Rho family GTPase involved in tumorigenesis. The Rho GTPase-activating protein 25 (ARHGAP25) has been shown to specifically inactivate the Rho family GTPase Rac1, which plays an important role in pancreatic adenocarcinoma (PAAD) progression. Therefore, here we aimed to clarify the expression and functional role of ARHGAP25 in PAAD. The ARHGAP25 expression was lower in PAAD tissues than that in normal pancreatic tissues based on bioinformatics analysis and immunohistochemistry staining. Overexpression of ARHGAP25 inhibited cell growth of AsPC-1 human pancreatic cancer cells in vitro, while opposite results were observed in BxPC-3 human pancreatic cancer cells with ARHGAP25 knockdown. Consistently, in vivo tumorigenicity assays also confirmed that ARHGAP25 overexpression suppressed tumor growth. Mechanically, overexpression of ARHGAP25 inactivated AKT/mTOR signaling pathway by regulating Rac1/PAK1 signaling, which was in line with the results from the Gene set enrichment analysis on The Cancer Genome Atlas dataset. Furthermore, we found that ARHGAP25 reduced HIF-1α-mediated glycolysis in PAAD cells. Treatment with PF-04691502, a dual PI3K/mTOR inhibitor, hampered the increased cell growth and glycolysis due to ARHGAP25 knockdown in PAAD cells. Altogether, these results conclude that ARHGAP25 acts as a tumor suppressor by inhibiting the AKT/mTOR signaling pathway, which might provide a therapeutic target for PAAD.


Assuntos
Adenocarcinoma/genética , Carcinogênese/genética , Proteínas Ativadoras de GTPase/genética , Regulação Neoplásica da Expressão Gênica , Proteína Oncogênica v-akt/genética , Neoplasias Pancreáticas/genética , Serina-Treonina Quinases TOR/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Feminino , Proteínas Ativadoras de GTPase/biossíntese , Glicólise/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Experimentais , Proteína Oncogênica v-akt/biossíntese , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/biossíntese
10.
Brain Res ; 1764: 147465, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33811842

RESUMO

BACKGROUND: Circular RNAs (circRNAs) are found to regulate glioblastoma evolution. However, the role of circ-ATP binding cassette subfamily C member 3 (circABCC3) in glioblastoma process is still unknown. In this study, the effects of circABCC3 on glioblastoma tumorigenesis and underlying mechanism were revealed. METHODS: The expression levels of circABCC3, microRNA-770-5p (miR-770-5p) and sex determining region Y-box protein 2 (SOX2) mRNA were determined by quantitative real-time polymerase chain reaction (qRT-PCR). The expression of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway-related proteins and SOX2 protein was detected by western blot analysis. Cell proliferation and invasion were severally investigated by cell colony formation and transwell invasion assays. Cell migration was demonstrated by transwell migration and wound-healing assays. Cell apoptosis was revealed by flow cytometry analysis. Tube formation was investigated by tube formation assay. The associated relationship between miR-770-5p and circABCC3 or SOX2 was predicted by starbase or targetscan online database, and identified by dual-luciferase reporter assay, RNA immunoprecipitation assay or RNA pull-down assay. The impacts of circABCC3 knockdown on glioblastoma growth in vivo were revealed by in vivo assay. RESULTS: CircABCC3 and SOX2 expression were dramatically upregulated, while miR-770-5p expression was apparently downregulated in glioblastoma tissues and cells compared with control groups. CircABCC3 expression was higher in stage III glioblastoma tissues than in stage I + II glioblastoma tissues with close correlation with tumor-node-metastasis (TNM) stage. CircABCC3 absence inhibited cell proliferation, migration, invasion, tube formation and the activation of PI3K/AKT pathway, whereas induced cell apoptosis in glioblastoma. Additionally, circABCC3 acted as a sponge for miR-770-5p, and miR-770-5p targeted SOX2. MiR-770-5p inhibitors impaired the impacts of circABCC3 silencing on glioblastoma progression, angiogenesis and PI3K/AKT pathway. Furthermore, circABCC3 knockdown repressed tumor growth in vivo. CONCLUSION: CircABCC3 regulated glioblastoma development via miR-770-5p/SOX2 axis through PI3K/AKT pathway. This finding lays a theoretical foundation for studying circRNA-directed therapy for glioblastoma.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , MicroRNAs/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Fatores de Transcrição SOXB1/genética , Transdução de Sinais/genética , Apoptose/genética , Neoplasias Encefálicas/patologia , Movimento Celular/genética , Proliferação de Células , Glioblastoma/patologia , Humanos , Invasividade Neoplásica/genética , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Ensaio Tumoral de Célula-Tronco
11.
FEBS J ; 288(19): 5650-5667, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33837641

RESUMO

Three-dimensional (3D) cultures of cancer cells in liquid without extracellular matrix (ECM) offer in vitro models for metastasising conditions such as those in vessels and effusion. However, liquid culturing is often hindered by cell adhesiveness, which causes large cell clumps. We previously described a liquid culture material, LA717, which prevents nonclonal cell adhesion and subsequent clumping, thus allowing clonal growth of spheroids in an anchorage-independent manner. Here, we examined such liquid culture cancer spheroids for the acquisition of apical-basal polarity, sensitivity to an Akt inhibitor (anticancer drug MK-2206) and interaction with ECM. The spheroids present apical plasma membrane on the surface, which originated from the failure of polarisation at the single-cell stage and subsequent defects in phosphorylated ezrin accumulation at the cell boundary during the first cleavage, failing internal lumen formation. At the multicellular stage, liquid culture spheroids presented bleb-like protrusion on the surface, which was enhanced by the activation of the PI3K/Akt pathway and reduced by PI3K/Akt inhibitors. Liquid culture spheroids exhibited slow proliferation speed and low endogenous pAkt levels compared with gel-cultured spheroids and 2D-cultured cells, explaining the susceptibility to the Akt-inhibiting anticancer drug. Subcutaneous xenografting and in vitro analysis demonstrated low viability and adhesive property of liquid culture spheroids to ECM, while migratory and invasive capacities were comparable with gel-cultured spheroids. These features agree with the low efficacy of circulating tumour spheroids in the settling step of metastasis. This study demonstrates the feature of anchorage-independent spheroids and validates liquid cultures as a useful method in cancer spheroid research.


Assuntos
Adesão Celular/genética , Técnicas de Cultura de Células , Neoplasias/genética , Esferoides Celulares/patologia , Animais , Linhagem Celular Tumoral , Polaridade Celular/genética , Matriz Extracelular/genética , Humanos , Camundongos , Metástase Neoplásica , Neoplasias/patologia , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Esferoides Celulares/metabolismo , Transplante Heterólogo
12.
Genes (Basel) ; 12(2)2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671468

RESUMO

Systemic treatment of hormone receptor-positive (HR+) breast cancer is undergoing a renaissance, with a number of targeted therapies including CDK4/6, mTOR, and PI3K inhibitors now approved for use in combination with endocrine therapies. The increased use of targeted therapies has changed the natural history of HR+ breast cancers, with the emergence of new escape mechanisms leading to the inevitable progression of disease in patients with advanced cancers. The identification of new predictive and pharmacodynamic biomarkers to current standard-of-care therapies and discovery of new therapies is an evolving and urgent clinical challenge in this setting. While traditional, routinely measured biomarkers such as estrogen receptors (ERs), progesterone receptors (PRs), and human epidermal growth factor receptor 2 (HER2) still represent the best prognostic and predictive biomarkers for HR+ breast cancer, a significant proportion of patients either do not respond to endocrine therapy or develop endocrine resistant disease. Genomic tests have emerged as a useful adjunct prognostication tool and guide the addition of chemotherapy to endocrine therapy. In the treatment-resistant setting, mutational profiling has been used to identify ESR1, PIK3CA, and AKT mutations as predictive molecular biomarkers to newer therapies. Additionally, pharmacodynamic biomarkers are being increasingly used and considered in the metastatic setting. In this review, we summarise the current state-of-the-art therapies; prognostic, predictive, and pharmacodynamic molecular biomarkers; and how these are impacted by emerging therapies for HR+ breast cancer.


Assuntos
Neoplasias da Mama/genética , Classe I de Fosfatidilinositol 3-Quinases/genética , Receptor alfa de Estrogênio/genética , Proteína Oncogênica v-akt/genética , Biomarcadores Tumorais/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Feminino , Hormônios/uso terapêutico , Humanos , Metástase Neoplásica , Prognóstico , Receptor ErbB-2/genética , Receptores de Estrogênio/genética , Receptores de Progesterona/genética
13.
Mol Med Rep ; 23(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33760180

RESUMO

Nasopharyngeal carcinoma (NPC) is an epithelial carcinoma originating from the nasopharyngeal mucosal tissue and is highly prevalent in southeast Asia. Galectin­3 (gal­3) serves crucial roles in many cancers but its role in NPC remains to be elucidated. The aim of the present study was to investigate the role of gal­3 in NPC. Immunohistochemistry and ELISA were used to determine the expression level of gal­3 in patients with NPC or chronic rhinitis (CR). Gal­3 short hairpin (sh)RNA was established to knockdown gal­3 in 5­8F and 6­10B cells, allowing for the evaluation of the roles of gal­3 in proliferation, migration and apoptosis in NPC cell lines. Immunohistochemistry staining of IL­6 and IL­8 was applied to access the inflammatory state of tumor tissues, and the correlation between the inflammatory state and gal­3 was analyzed. The results demonstrated that gal­3 was upregulated in patients with NPC compared with patients with CR. Knockdown of gal­3 inhibited proliferation and migration in 5­8F and 6­10B cells, as well as promoted apoptosis in these cells. The expression levels of MMP­9 and IL­8 were also decreased in 5­8F and 6­10B cells after transfection with gal­3 shRNA. A positive correlation was identified between the expression level of gal­3 and the inflammatory state of NPC. The phosphorylation levels of ERK1/2 and Akt were downregulated after knockdown of gal­3 in 5­8F and 6­10B cells. In conclusion, the expression level of gal­3 was upregulated in patients with NPC and was positively correlated with the inflammatory state of NPC. The results suggested that gal­3 promoted the proliferation and migration of 5­8F and 6­10B cells, while inhibiting the apoptosis of these cells. Moreover, activation of ERK1/2 and Akt may be the underlying mechanism of the effects of gal­3 on NPC.


Assuntos
Galectina 3/genética , Inflamação/genética , Interleucina-8/genética , Metaloproteinase 9 da Matriz/genética , Carcinoma Nasofaríngeo/genética , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Galectina 3/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Inflamação/patologia , Sistema de Sinalização das MAP Quinases/genética , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/terapia , Proteína Oncogênica v-akt/genética , RNA Interferente Pequeno/farmacologia
14.
J Biosci ; 462021.
Artigo em Inglês | MEDLINE | ID: mdl-33576342

RESUMO

The silence of lncRNA small nucleolar RNA host gene 16 (SNHG16) suppressed acute lymphoblastic leukemia (ALL) cell proliferation and migration, whereas its role in acute myeloid leukemia (AML) still lacks clarity. This study showed that SNHG16 was upregulated in AML patients and cells. And SNHG16 overexpression remarkably enhanced the proliferation and migration capacities of HL60 and AML-193 cells, while SNHG16 knockdown acted the opposite way. Subsequently, we revealed that SNHG16 directly bound to CELF2 (CUGBP Elav-like family member 2) protein, and caused CELF2 mRNA unstably and proteins reducing. CELF2 was decreased both in AML patients and cells. CELF2 overexpression or interference weakened the effect of overexpressing or silencing SNHG16 on proliferation and migration. Moreover, the transfection of pcDNA-CELF2 elevated PTEN (phosphatase and tensin homolog) activity and hindered the phosphoinositide 3-kinase (PI3K)/AKT signaling. And SNHG16 reduced PTEN activity and promoted the PI3K/AKT pathway activation by restraining CELF2. Furthermore, GDC-0941 (a specific inhibitor of the PI3K/AKT pathway) impeded the effect of SNHG16 increase, and bpV(pic) (a specific PTEN inhibitor) declined the effect of SNHG16 decrease on cell proliferation and migration. Taken together, the present study indicated that SNHG16 promoted proliferation and migration of AML cells via PTEN/PI3K/AKT axis through suppressing CELF2 protein.


Assuntos
Proteínas CELF/genética , Leucemia Mieloide Aguda/sangue , Proteínas do Tecido Nervoso/genética , PTEN Fosfo-Hidrolase/genética , RNA Longo não Codificante/genética , Proteínas CELF/sangue , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Leucêmica da Expressão Gênica/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Masculino , Proteínas do Tecido Nervoso/sangue , Proteína Oncogênica v-akt/sangue , Proteína Oncogênica v-akt/genética , PTEN Fosfo-Hidrolase/sangue , Fosfatidilinositol 3-Quinases/sangue , Fosfatidilinositol 3-Quinases/genética , RNA Longo não Codificante/sangue , Transdução de Sinais/genética
15.
Oncogene ; 40(9): 1578-1594, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33452458

RESUMO

Aberrant Notch activation has been implicated in multiple malignancies and the identification of NOTCH receptors and related pathways is critical for targeted therapy. In this study, we aim to delineate the most prominent dysregulated NOTCH receptor and comprehensively reveal its deregulation in gastric cancer (GC). In the four Notch members, NOTCH3 was found uniformly upregulated and associated with poor clinical outcomes in multiple GC datasets. siRNA-mediated NOTCH3 knockdown demonstrated antitumor effects by suppressing cell proliferation, inhibiting monolayer formation, and impairing cell invasion abilities. Its depletion also induced early and late apoptosis. NOTCH3 was confirmed to be a direct target of two tumor suppressor microRNAs (miRNAs), namely miR-491-5p and miR-875-5p. The activation of NOTCH3 is partly due to the silence of these two miRNAs. Through RNA-seq profiling and functional validation, PHLDB2 was identified as a potent functional downstream modulator for NOTCH3 in gastric carcinogenesis. PHLDB2 expression demonstrated a positive correlation with NOTCH3, but was negatively correlated with miR-491-5p. Akt-mTOR was revealed as the downstream signaling of PHLDB2. The NOTCH3-PHLDB2-Akt co-activation was found in 33.7% GC patients and the activation of this axis predicted poor clinical outcome. GC cells treated with siNOTCH3, siPHLDB2, miR-491-5p, miR-875-5p, were more sensitive to Cisplatin and 5-FU. Taken together, the NOTCH3-PHLDB2-Akt cascade plays oncogenic role in gastric carcinogenesis and serves as a therapeutic target. Our study provided insights into Notch-mediated underlying molecular mechanisms and implied translational potential.


Assuntos
Proteínas de Transporte/genética , MicroRNAs/genética , Receptor Notch3/genética , Neoplasias Gástricas/genética , Apoptose/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteína Oncogênica v-akt/genética , RNA Interferente Pequeno/genética , Neoplasias Gástricas/patologia , Serina-Treonina Quinases TOR/genética
16.
Oncogene ; 40(9): 1595-1608, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33452459

RESUMO

KNSTRN is a component of the mitotic spindle, which was rarely investigated in tumorigenesis. AKT plays an essential role in tumorigenesis by modulating the phosphorylation of various substrates. The activation of AKT is regulated by PTEN and PIP3. Here, we prove KNSTRN is positively correlated with malignancy of bladder cancer and KNSTRN activates AKT phosphorylation at Thr308 and Ser473. More importantly, our study reveals that both KNSTRN and PTEN interact with PH domain of AKT at cell membrane. The amount of KNSTRN interacted with AKT is negatively related to PTEN. Furthermore, PIP3 pull-down assay proves that KNSTRN promoted AKT movement to PIP3. These data suggest KNSTRN may activate AKT phosphorylation by promoting AKT movement to PIP3 and alleviating PTEN suppression. Based on the activation of AKT phosphorylation, our study demonstrates that KNSTRN promotes bladder cancer metastasis and gemcitabine resistance in vitro and in vivo. Meanwhile, the effect of KNSTRN on tumorigenesis and gemcitabine resistance could be restored by AKT specific inhibitor MK2206 or AKT overexpression. In conclusion, we identify an oncogene KNSTRN that promotes tumorigenesis and gemcitabine resistance by activating AKT phosphorylation and may serve as a therapeutic target in bladder cancer.


Assuntos
Carcinogênese/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Desoxicitidina/análogos & derivados , Proteínas Associadas aos Microtúbulos/genética , Proteína Oncogênica v-akt/genética , Neoplasias da Bexiga Urinária/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Proteína Oncogênica v-akt/antagonistas & inibidores , Fosforilação/efeitos dos fármacos , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Gencitabina
17.
Oncogene ; 40(7): 1269-1283, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33420362

RESUMO

Distant metastasis is the leading cause of treatment failure in patients with hepatocellular carcinoma (HCC). However, the underlying mechanisms have not been fully elucidated. Here, we report that Leucine zipper tumor suppressor 2 (LZTS2) is downregulated and correlated with poor prognosis in HCC. Furthermore, we provide evidence that LZTS2 associates with p85 to inhibit the activation of PI3K/AKT signaling and impairs HCC tumorigenesis and metastasis in vitro and in vivo. Moreover, we identify LZTS2 as a bona fide substrate of the E3 ligase ß-Trcp and protein kinase CK1δ, which are responsible for the ubiquitination and degradation of LZTS2. Importantly, we show that the ß-Trcp and CK1δ-mediated degradation of LZTS2 promotes HCC progression and metastasis by activating PI3K/AKT signaling. Collectively, our study not only illustrates the roles of LZTS2 in regulating HCC tumorigenesis and metastasis but also reveals a novel posttranslational modification of LZTS2 by ß-Trcp and CK1δ, indicating that the ß-Trcp/CK1δ/LZTS2/PI3K axis may be a novel oncogenic driver involved in HCC progression and metastasis.


Assuntos
Carcinoma Hepatocelular/genética , Caseína Quinase Idelta/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Neoplasias Hepáticas/genética , Proteínas Supressoras de Tumor/genética , Proteínas Contendo Repetições de beta-Transducina/genética , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Metástase Neoplásica , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética
18.
J Recept Signal Transduct Res ; 41(1): 19-31, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32586178

RESUMO

PURPOSE: MiR-199a-3p is low expressed in diabetic retinopathy (DR). In the current study, we investigated the effects of miR-199a-3p on DR and the potential mechanisms. METHODS: A DR rat model was established, and endothelial cells (ECs) and retinal pericytes (RPs) were extracted from the DR model rats to detect miR-199a-3p expression. Bioinformatics analysis predicted that fibroblast growth factor 7 (FGF7) was a target gene for miR-199a-3p, which was confirmed by dual-luciferase assay. Cell proliferation, migration, and invasion were detected by cell counting kit-8 (CCK-8), colony formation assay, wound-healing, and Transwell assay. Quantitative real-time polymerase chain reaction (q-PCR) and Western blot were performed to detect the expressions of mRNAs and proteins. RESULTS: MiR-199a-3p was low expressed and FGF7 was high-expressed in ECs and RPs. Overexpressed miR-199a-3p suppressed the proliferation, migration, and invasion, and FGF7 expression of ECs and RPs. However, overexpression of FGF7 effectively eliminated the suppressive effects of miR-199a-3p overexpression malignant behaviors of the cells. Meanwhile, up-regulation of FGF7 noticeably reversed the phosphorylation of phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT) and the expression of epidermal growth factor receptor (EGFR) reduced by miR-199a-3p. CONCLUSION: Our findings revealed that in the DR rat model, miR-199a-3p inhibited cell proliferation, migration, and invasion of EC and RP through targeting FGF7 and inhibiting the activation of the EGFR/PI3K/AKT pathway. This study may provide a new direction for the search for the treatment of DR.


Assuntos
Retinopatia Diabética/genética , Células Endoteliais/metabolismo , Receptores ErbB/genética , Fator 7 de Crescimento de Fibroblastos/genética , MicroRNAs/genética , Animais , Movimento Celular/genética , Proliferação de Células/genética , Retinopatia Diabética/patologia , Células Endoteliais/patologia , Humanos , Proteína Oncogênica v-akt/genética , Pericitos/metabolismo , Pericitos/patologia , Fosfatidilinositol 3-Quinases/genética , Ratos , Retina/metabolismo , Retina/patologia , Transdução de Sinais/genética
19.
Cancer Gene Ther ; 28(6): 649-666, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33093643

RESUMO

Combined antiretroviral therapy (cART) for HIV-1 dramatically slows disease progression among HIV+ individuals. Currently, lymphoma represents the main cause of death among HIV-1-infected patients. Detection of p17 variants (vp17s) endowed with B-cell clonogenic activity in HIV-1-seropositive patients with lymphoma suggests their possible role in lymphomagenesis. Here, we demonstrate that the clonogenic activity of vp17s is mediated by their binding to PAR1 and to PAR1-mediated EGFR transactivation through Gq protein. The entire vp17s-triggered clonogenic process is MMPs dependent. Moreover, phosphoproteomic and bioinformatic analysis highlighted the crucial role of EGFR/PI3K/Akt pathway in modulating several molecules promoting cancer progression, including RAC1, ABL1, p53, CDK1, NPM, Rb, PTP-1B, and STAT1. Finally, we show that a peptide (F1) corresponding to the vp17s functional epitope is sufficient to trigger the PAR1/EGFR/PI3K/Akt pathway and bind PAR1. Our findings suggest novel potential therapeutic targets to counteract vp17-driven lymphomagenesis in HIV+ patients.


Assuntos
Carcinogênese/genética , Antígenos HIV/genética , HIV-1/genética , Linfoma/genética , Receptor PAR-1/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Fator de Crescimento Epidérmico/genética , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/patogenicidade , Humanos , Ativação Linfocitária/genética , Linfoma/patologia , Linfoma/virologia , Proteína Oncogênica v-akt/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Ativação Transcricional/genética
20.
Vet Microbiol ; 252: 108926, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33223234

RESUMO

Our present study aimed to identify host cell proteins that may interact with avian reovirus (ARV) σA protein and their potential effect on ARV replication. The ARV structural protein σA has been demonstrated to suppress interferon production and confirmed to activate the PI3K/Akt pathway. However, host cell factors interacting with σA to affect ARV replication remain unknown. In current study, a cDNA library of chicken embryo fibroblasts (CEFs) was constructed, and host cell proteins interacting with σA were screened by a yeast two-hybrid system. We identified four candidate cellular proteins that interact with ARV σA protein. Among them, Gallus NME/NM23 nucleoside diphosphate kinase 2 (NME2) was further validated as a σA-binding protein through co-immunoprecipitation. The key interaction domain was identified at amino acids (aa) 121-416 in NME2 and at aa 71-139 in σA, respectively. We demonstrated that overexpression of NME2 substantially inhibited ARV replication. In addition silencing NME2 by small interfering RNAs (siRNAs) resulted in marked enhancement of ARV replication. Our work has demonstrated that NME2 is a σA-binding protein that may affect ARV replication in CEF cells.


Assuntos
Nucleosídeo NM23 Difosfato Quinases/metabolismo , Orthoreovirus Aviário/enzimologia , Proteínas de Ligação a RNA/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral , Animais , Embrião de Galinha , Fibroblastos/fisiologia , Nucleosídeo NM23 Difosfato Quinases/genética , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Orthoreovirus Aviário/genética , Orthoreovirus Aviário/fisiologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Mapeamento de Interação de Proteínas/veterinária , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Proteínas do Core Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA