Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Biochem Biophys Res Commun ; 710: 149860, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38604070

RESUMO

Schizophyllan (SPG), a ß-glucan from Schizophyllum commune, is recognized for its antioxidant, immunoregulatory, and anticancer activities. In this study, its effects on bone cells, particularly osteoclasts and osteoblasts, were examined. We demonstrated that SPG dose-dependently inhibited osteoclastogenesis and reduced gene expression associated with osteoclast differentiation. SPG also decreased bone resorption and F-actin ring formation. This inhibition could have been due to the downregulation of transcription factors c-Fos and nuclear factor of activated T cells 1 (NFATc1) via the MAPKs (JNK and p38), IκBα, and PGC1ß/PPARγ pathways. In coculture, SPG lowered osteoclastogenic activity in calvaria-derived osteoblasts by reducing macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) expression. In addition, SPG slightly enhanced osteoblast differentiation, as evidenced by increased differentiation marker gene expression and alizarin red staining. It also exhibited antiresorptive effects in a lipopolysaccharide-induced calvarial bone loss model. These results indicated a dual role of SPG in bone cell regulation by suppressing osteoclastogenesis and promoting osteoblast differentiation. Thus, SPG could be a therapeutic agent for bone resorption-related diseases such as osteoporosis, rheumatoid arthritis, and periodontitis.


Assuntos
Reabsorção Óssea , Sizofirano , Humanos , Osteoclastos/metabolismo , Sizofirano/metabolismo , Sizofirano/farmacologia , Fatores de Transcrição NFATC/metabolismo , Osteoblastos/metabolismo , Diferenciação Celular , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Osteogênese , Ligante RANK/metabolismo
2.
Clin Oral Investig ; 28(1): 116, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38270679

RESUMO

OBJECTIVES: To investigate molecular and clinical background of associations among oral health, muscle and bone metabolism, and frailty incidence in patients with fall and fracture history. MATERIALS AND METHODS: In total, 88 elderly participants (mean age 71.9 ± 5.8 years) with the distal radius fractures were included. Participants were divided into three groups based on an Oral Health Assessment Tool score. Fried criteria and Mini-nutritional assessments were adopted to diagnose frailty and malnutrition, respectively. Blood samples were collected and analyzed for serum levels of bone turnover markers, proteins, insulin-like growth factor-1, 25-hydroxyvitamin D, and inflammatory cytokines. The mRNA levels of markers of inflammation, muscle synthesis and wasting, and muscle homeostasis regulator in the pronator quadratus muscle were analyzed. RESULTS: Patients with deteriorated oral health demonstrated a higher prevalence of frailty and malnutrition. Significantly lower serum levels of total protein and higher concentrations of tumor necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) were detected in patients with poor oral health. Significant interaction effects between oral health and frailty level in gait speed, serum TNF-α, IL-1ß, and total protein levels were exhibited. Significantly different mRNA expression levels in the pronator quadratus muscle of TNF-α, IL-1ß, NF kB, MYOG, and FOXO1 following the oral health were detected. CONCLUSION: This study highlights relationship between oral health, nutritional uptake, systemic inflammation, and their combined impact on muscle and bone metabolism, ultimately affecting frailty development in the aging populations. CLINICAL RELEVANCE: A comprehensive understanding of mutual interactions among oral health, nutrition, and inflammation is essential for managing frailty.


Assuntos
Fragilidade , Desnutrição , Idoso , Humanos , Saúde Bucal , Fator de Necrose Tumoral alfa , Músculos , Inflamação , RNA Mensageiro
3.
J Endocr Soc ; 6(8): bvac084, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35702666

RESUMO

Undercarboxylated osteocalcin (ucOCN) has been considered to be an important endocrine factor, especially to regulate bone and energy metabolism. Even with the mounting evidence showing the consistent inverse correlation of ucOCN levels in chronic inflammatory diseases, however, the mechanism underlying the involvement of ucOCN in the muscular inflammation has not been fully understood. In the present study, we explored 1) the endocrine role of ucOCN in the regulation of inflammation in C2C12 myoblasts and primary myoblasts and the underlying intracellular signaling mechanisms, and 2) whether G protein-coupled receptor family C group 6 member A (GPRC6A) is the ucOCN-sensing receptor associated with the ucOCN-mediated anti-inflammatory signaling pathway in myoblasts. ucOCN suppressed the tumor necrosis factor-α (TNF-α)-induced expressions of major inflammatory cytokines, including interleukin-1ß (IL-1ß) and inhibited the TNF-α-stimulated activities of transcription factors, including NF-κB, in C2C12 and primary myoblasts. Both knockdown and knockout of GPRC6A, by using siRNA or a CRISPR/CAS9 system, respectively, did not reverse the effect of ucOCN on IL-1ß expression in myoblasts. Interestingly, TNF-α-induced IL-1ß expression was inhibited by knockdown or deletion of GPRC6A itself, regardless of the ucOCN treatment. ucOCN was rapidly internalized into the cytoplasmic region via caveolae-mediated endocytosis, suggesting the presence of new target proteins in the cell membrane and/or in the cytoplasm for interaction with ucOCN in myoblasts. Taken together, these findings indicate that ucOCN suppresses the TNF-α-induced inflammatory signaling pathway in myoblasts. GPRC6A is not a sensing receptor associated with the ucOCN-mediated anti-inflammatory signaling pathway in myoblasts.

4.
Cancer Sci ; 113(1): 145-155, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34714604

RESUMO

To improve the poor survival rate of lung cancer patients, we investigated the role of HDGF-related protein 3 (HRP-3) as a potential biomarker for lung cancer. The expression of endogenous HRP-3 in human lung cancer tissues and xenograft tumor models is indicative of its clinical relevance in lung cancer. Additionally, we demonstrated that HRP-3 directly binds to the E2F1 promoter on chromatin. Interestingly, HRP-3 depletion in A549 cells impedes the binding of HRP-3 to the E2F1 promoter; this in turn hampers the interaction between Histone H3/H4 and HDAC1/2 on the E2F1 promoter, while concomitantly inducing Histone H3/H4 acetylation around the E2F1 promoter. The enhanced Histone H3/H4 acetylation on the E2F1 promoter through HRP-3 depletion increases the transcription level of E2F1. Furthermore, the increased E2F1 transcription levels lead to the enhanced transcription of Cyclin E, known as the E2F1-responsive gene, thus inducing S-phase accumulation. Therefore, our study provides evidence for the utility of HRP-3 as a biomarker for the prognosis and treatment of lung cancer. Furthermore, we delineated the capacity of HRP-3 to regulate the E2F1 transcription level via histone deacetylation.


Assuntos
Biomarcadores Tumorais/metabolismo , Ciclina E/metabolismo , Fator de Transcrição E2F1/genética , Histona Desacetilases/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pulmonares/patologia , Células A549 , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Transplante de Neoplasias , Regiões Promotoras Genéticas , Transdução de Sinais
5.
Int J Mol Sci ; 22(23)2021 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-34884637

RESUMO

The adverse effects of radiation are proportional to the total dose and dose rate. We aimed to investigate the effects of radiation dose rate on different organs in mice. The mice were subjected to low dose rate (LDR, ~3.4 mGy/h) and high dose rate (HDR, ~51 Gy/h) radiation. LDR radiation caused severe tissue toxicity, as observed in the histological analysis of testis. It adversely influenced sperm production, including sperm count and motility, and induced greater sperm abnormalities. The expression of markers of early stage spermatogonial stem cells, such as Plzf, c-Kit, and Oct4, decreased significantly after LDR irradiation, compared to that following exposure of HDR radiation, in qPCR analysis. The compositional ratios of all stages of spermatogonia and meiotic cells, except round spermatid, were considerably reduced by LDR in FACS analysis. Therefore, LDR radiation caused more adverse testicular damage than that by HDR radiation, contrary to the response observed in other organs. Therefore, the dose rate of radiation may have differential effects, depending on the organ; it is necessary to evaluate the effect of radiation in terms of radiation dose, dose rate, organ type, and other conditions.


Assuntos
Espermatogênese/efeitos da radiação , Testículo/efeitos da radiação , Animais , Relação Dose-Resposta à Radiação , Raios gama , Masculino , Camundongos , Modelos Animais , Doses de Radiação , Espermátides/citologia , Espermátides/efeitos da radiação , Espermatogônias/citologia , Espermatogônias/efeitos da radiação , Espermatozoides/citologia , Espermatozoides/efeitos da radiação , Testículo/citologia
6.
Sci Rep ; 10(1): 12272, 2020 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-32704022

RESUMO

Glioblastoma is frequently associated with TP53 mutation, which is linked to a worse prognosis and response to conventional treatments (chemoradiotherapy). Therefore, targeting TP53 is a promising strategy to overcome this poor therapeutic response. Tumor-treating fields (TTFields) are a recently approved treatment for newly diagnosed glioblastoma, which involves direct application of low-intensity, intermediate-frequency alternating electric fields to the tumor, thereby offering a local tumor-killing effect. However, the influence of TP53 mutation status on the effectiveness of TTFields is controversial. Here, we identified the key gene signatures and pathways associated with TTFields in four glioblastoma cell lines varying in TP53 mutation status using gene profiling and functional annotation. Overall, genes associated with the cell cycle, cell death, and immune response were significantly altered by TTFields regardless of TP53 status. TTFields appeared to exert enhanced anti-cancer effects by altering the immune system in the inflammatory environment and regulating cell cycle- and cell death-related genes, but the precise genes influenced vary according to TP53 status. These results should facilitate detailed mechanistic studies on the molecular basis of TTFields to further develop this modality as combination therapy, which can improve the therapeutic effect and minimize side effects of chemoradiotherapy.


Assuntos
Perfilação da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Biomarcadores Tumorais , Ciclo Celular/genética , Biologia Computacional/métodos , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Estadiamento de Neoplasias , Prognóstico , Transcriptoma
7.
Cell Death Dis ; 11(5): 398, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32457423

RESUMO

The poor therapeutic efficacy of non-small cell lung cancer (NSCLC) is partly attributed to the acquisition of chemoresistance. To investigate the mechanism underlying this resistance, we examined the potential link between kinesin light chain 4 (KLC4), which we have previously reported to be associated with radioresistance in NSCLC, and sensitivity to chemotherapy in human lung cancer cell lines. KLC4 protein levels in lung cancer cells correlated with the degree of chemoresistance to cisplatin treatment. Furthermore, KLC4 silencing enhanced the cytotoxic effect of cisplatin by promoting DNA double-strand breaks and apoptosis. These effects were mediated by interaction with the checkpoint kinase CHK2, as KLC4 knockdown increased CHK2 activation, which was further enhanced in combination with cisplatin treatment. In addition, KLC4 and CHEK2 expression levels showed negative correlation in lung tumor samples from patients, and KLC4 overexpression correlated negatively with survival. Our results indicate a novel link between the KLC4 and CHK2 pathways regulating DNA damage response in chemoresistance, and highlight KLC4 as a candidate for developing lung cancer-specific drugs and customized targeted molecular therapy.


Assuntos
Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Quinase do Ponto de Checagem 2/antagonistas & inibidores , Reparo do DNA , Resistencia a Medicamentos Antineoplásicos , Cinesinas/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Associadas aos Microtúbulos/metabolismo , Terapia de Alvo Molecular , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem/metabolismo , Quinase do Ponto de Checagem 2/metabolismo , Cisplatino/farmacologia , Neoplasias Colorretais/patologia , Dano ao DNA , Reparo do DNA/efeitos dos fármacos , Proteína Quinase Ativada por DNA/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Etoposídeo/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Inativação Gênica/efeitos dos fármacos , Humanos , Cinesinas/genética , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Transdução de Sinais/efeitos dos fármacos
8.
Oncogene ; 38(39): 6630-6646, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31375748

RESUMO

Tumor-treating fields (TTFs) - a type of electromagnetic field-based therapy using low-intensity electrical fields - has recently been characterized as a potential anticancer therapy for glioblastoma multiforme (GBM). However, the molecular mechanisms involved remain poorly understood. Our results show that the activation of autophagy contributes to the TTF-induced anti-GBM activity in vitro or in vivo and GBM patient stem cells or primary in vivo culture systems. TTF-treatment upregulated several autophagy-related genes (~2-fold) and induced cytomorphological changes. TTF-induced autophagy in GBM was associated with decreased Akt2 expression, not Akt1 or Akt3, via the mTOR/p70S6K pathway. An Affymetrix GeneChip miRNA 4.0 Array analysis revealed that TTFs altered the expression of many microRNAs (miRNAs). TTF-induced autophagy upregulated miR-29b, which subsequently suppressed the Akt signaling pathway. A luciferase reporter assay confirmed that TTFs induced miR-29b to target Akt2, negatively affecting Akt2 expression thereby triggering autophagy. TTF-induced autophagy suppressed tumor growth in GBM mouse models subjected to TTFs as determined by positron emission tomography and computed tomography (PET-CT). GBM patient stem cells and a primary in vivo culture system with high Akt2 levels also showed TTF-induced inhibition. Taken together, our results identified autophagy as a critical cell death pathway triggered by TTFs in GBM and indicate that TTF is a potential treatment option for GBM.


Assuntos
Autofagia/efeitos da radiação , Neoplasias Encefálicas/radioterapia , Glioblastoma/radioterapia , MicroRNAs/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Campos Eletromagnéticos , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Bone ; 127: 220-227, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31216497

RESUMO

Osteocalcin is an osteoblast-specific secreted protein that has been associated with endocrine roles in multiple aspects of energy metabolism. We examined whether undercarboxylated osteocalcin (ucOC) downregulates pancreatic lipase (PNLIP) expression in pancreatic acinar cells and then identified the downstream signaling pathway involved. We previously demonstrated that ß adrenergic blockade attenuates body weight/fat mass gain in high-fat diet-fed mice and that this effect is associated with decreased PNLIP expression in pancreatic acinar cells. In the present study, we first confirmed that the serum ucOC level is inversely correlated with PNLIP expression, i.e., mice exhibiting high serum levels of ucOC showed low PNLIP levels in the pancreas. In in vitro experiments using primary pancreatic acinar and 266-6 cells, ucOC downregulated PNLIP expression. cAMP/PKA signaling inhibitors significantly reversed ucOC-induced downregulation of PNLIP expression. ucOC promoted the phosphorylation of cAMP response element-binding protein 2 (ATF4). Overexpression of ATF4 significantly suppressed PNLIP expression. Knockdown of ATF4 by siRNA reversed the ucOC-induced downregulation of PNLIP expression. A luciferase reporter assay showed that ucOC suppressed PNLIP promoter transactivation. Chromatin immunoprecipitation and a luciferase reporter assay demonstrated that ATF4 directly bound to the CRE on the mouse PNLIP promoter and suppressed PNLIP transactivation. Knockdown of G-protein coupled receptor 6A (Gprc6a), a candidate receptor for mediating the response to ucOC in the bone-pancreas endocrine loop, by siRNA reversed the downregulating effect of ucOC on PNLIP expression. Taken together, ucOC downregulates pancreatic lipase expression in a cAMP/protein kinase A/ATF4-dependent manner. Gprc6a is a potential osteocalcin-sensing receptor that regulates PNLIP expression in pancreatic acinar cells.


Assuntos
Células Acinares/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Regulação para Baixo , Lipase/metabolismo , Osteocalcina/metabolismo , Pâncreas/enzimologia , Animais , Sequência de Bases , Linhagem Celular , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Osteocalcina/sangue , Regiões Promotoras Genéticas/genética , Ligação Proteica , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
10.
Cell Death Dis ; 10(3): 156, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30770789

RESUMO

Current lung cancer treatments are far from satisfactory; thus, finding novel treatment targets is crucial. We recently identified procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3 (PLOD3), which is involved in fibrosis and tissue remodeling as a radioresistance-related protein in lung cancer cells; however, its mechanism is unclear. In this study, we designed human PLOD3-specific short interfering (si)RNAs and tested their effects on tumor growth inhibition in vitro and in vivo. PLOD3 knockdown overcame chemoresistance and decreased radioresistance by inducing caspase-3-dependent apoptosis in lung cancer cells. Furthermore, PLOD3 interacted with PKCδ to activate caspase-2,4-dependent apoptosis through ER-stress-induced IRE1α activation and the downstream unfolded-protein response pathway. In a mouse xenograft model, PLOD3 knockdown promoted radiation-induced tumor growth inhibition, without side effects. Moreover, lung cancer patients with high PLOD3 expression showed poorer prognosis than those with low PLOD3 expression upon radiotherapy, suggesting that PLOD3 promotes tumor growth. Therefore, PLOD3 siRNA suppresses radioresistance and chemoresistance by inducing apoptosis and renders PLOD3 as a candidate lung cancer biomarker. PLOD3 gene therapy might enhance the efficacy of radiotherapy or chemotherapy in lung cancer patients.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pulmonares/metabolismo , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Proteína Quinase C-delta/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/genética , Células A549 , Animais , Apoptose/genética , Biomarcadores Tumorais/genética , Carcinogênese/genética , Proliferação de Células/genética , Dano ao DNA/genética , Estresse do Retículo Endoplasmático/genética , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Transfecção , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação , Resposta a Proteínas não Dobradas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Mol Sci ; 19(11)2018 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-30469352

RESUMO

Glioblastoma, the most common primary brain tumor in adults, is an incurable malignancy with poor short-term survival and is typically treated with radiotherapy along with temozolomide. While the development of tumor-treating fields (TTFields), electric fields with alternating low and intermediate intensity has facilitated glioblastoma treatment, clinical outcomes of TTFields are reportedly inconsistent. However, combinatorial administration of chemotherapy with TTFields has proven effective for glioblastoma patients. Sorafenib, an anti-proliferative and apoptogenic agent, is used as first-line treatment for glioblastoma. This study aimed to investigate the effect of sorafenib on TTFields-induced anti-tumor and anti-angiogenesis responses in glioblastoma cells in vitro and in vivo. Sorafenib sensitized glioblastoma cells to TTFields, as evident from significantly decreased post-TTFields cell viability (p < 0.05), and combinatorial treatment with sorafenib and TTFields accelerated apoptosis via reactive oxygen species (ROS) generation, as evident from Poly (ADP-ribose) polymerase (PARP) cleavage. Furthermore, use of sorafenib plus TTFields increased autophagy, as evident from LC3 upregulation and autophagic vacuole formation. Cell cycle markers accumulated, and cells underwent a G2/M arrest, with an increased G0/G1 cell ratio. In addition, the combinatorial treatment significantly inhibited tumor cell motility and invasiveness, and angiogenesis. Our results suggest that combination therapy with sorafenib and TTFields is slightly better than each individual therapy and could potentially be used to treat glioblastoma in clinic, which requires further studies.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica/métodos , Glioblastoma/terapia , Sorafenibe/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Autofagia , Neoplasias Encefálicas/tratamento farmacológico , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Terapia Combinada/métodos , Glioblastoma/tratamento farmacológico , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sorafenibe/administração & dosagem
12.
Cell Death Dis ; 9(12): 1138, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-30442941

RESUMO

Procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD3), a membrane-bound homodimeric enzyme, hydroxylates lysyl residues in collagen-like peptides; however, its role in lung cancer is unknown. This study aimed to investigate the role of PLOD3 as a pro-metastatic factor and to elucidate the underlying mechanism. First, we experimentally confirmed the release of PLOD3 in circulation in animal models, rendering it a potential serum biomarker for lung cancer in humans. Thereafter, we investigated the effects of PLOD3 overexpression and downregulation on cancer cell invasion and migration in vitro and in vivo, using human lung cancer cell lines and a mouse tumor xenograft model, respectively. Further, PLOD3 levels were determined in lung tissue samples from lung cancer patients. Functional analyses revealed that PLOD3 interacts with STAT3, thereby expressing matrix metalloproteinases (MMP-2 and MMP-9) and with urokinase plasminogen activator (uPA) to enhance tumor metastasis. PLOD3 and the STAT3 pathway were significantly correlated in the metastatic foci of lung cancer patients; PLOD3-STAT3 levels were highly correlated with a poor prognosis. These results indicate that PLOD3 promotes lung cancer metastasis in a RAS-MAP kinase pathway-independent manner. Therefore, secreted PLOD3 serves as a potent inducer of lung cancer metastasis and a potential therapeutic target to enhance survival in lung cancer.


Assuntos
Proliferação de Células/genética , Neoplasias Pulmonares/genética , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Fator de Transcrição STAT3/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/patologia , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Ligação Proteica/genética , Ativador de Plasminogênio Tipo Uroquinase/genética
13.
Int J Mol Sci ; 19(10)2018 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-30322210

RESUMO

Distal-less homeobox 5 (Dlx5) is a negative regulator of adipogenesis. Dlx5 expression is decreased by adipogenic stimuli, but the mechanisms of Dlx5 downregulation by adipogenic stimuli have not yet been determined. Here, we tested the impact of cAMP/PKA (protein kinase A) signaling induced by 3-isobutyl-1 methyl xanthine (IBMX), forskolin, and 8-CPT-cAMP on the expression of Dlx5 in 3T3-L1 preadipocytes. Significant downregulation of Dlx5 mRNA expression and protein production levels were observed via cAMP/PKA-dependent signaling. Forced expression of cAMP-responsive element-binding protein (CREB) and CCAAT/enhancer-binding protein ß (C/EBPß) was sufficient for downregulation of Dlx5 expression and revealed that CREB functions upstream of C/EBPß. In addition, C/EBPß knockdown by siRNA rescued Dlx5 expression in IBMX-treated 3T3-L1 preadipocytes. Luciferase assays using a Dlx5-luc-2935 reporter construct demonstrated the requirement of the Dlx5 promoter region, ranging from -774 to -95 bp that contains two putative C/EBPß binding elements (site-1: -517 to -510 bp and site-2: -164 to -157 bp), in the suppression of Dlx5 transcription. Consequently, chromatin immunoprecipitation analysis confirmed the importance of site-1, but not site-2, in C/EBPß binding and transcriptional suppression of Dlx5. In conclusion, we elucidated the underling mechanism of Dlx5 downregulation in IBMX-induced adipogenesis. IBMX activated cAMP/PKA/CREB signaling and subsequently upregulated C/EBPß, which binds to the Dlx5 promoter to suppress Dlx5 transcription.


Assuntos
Adipócitos/citologia , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Homeodomínio/genética , 1-Metil-3-Isobutilxantina/farmacologia , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia , Animais , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/genética , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Tionucleotídeos/farmacologia
14.
Cell Death Discov ; 4: 46, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30302280

RESUMO

Tumor treating fields (TTFs) are a newly developed cancer therapy technology using an alternating electric field that may be a possible candidate for overcoming the limitations of conventional treatment methods currently used in cancer treatment. Although clinical results using TTFs appear promising, concerns regarding side effects must be clarified to demonstrate the effectiveness of this treatment method. To investigate the side effects of TTF treatment, the damage to normal cell lines and normal tissue of a mouse model was compared with the damage to tumor cells and tumors in a mouse model after TTF treatment. No serious damage was found in the normal cells and normal tissues of the mouse model, suggesting that the side effects of TTF treatment may not be serious. Our evidence based on in vitro and in vivo experiments suggests that TTF may cause selective damage to cancer cells, further demonstrating the potential of TTF as an attractive alternative to conventional cancer treatment modalities.

15.
Cell Death Dis ; 9(5): 496, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717133

RESUMO

Kinesins act as molecular microtubule-dependent motor proteins and have various important cellular functions related to cell division, intracellular transport, and membrane trafficking. However, the function of kinesin light chain 4 (KLC4) in cancer, especially radioresistance, has not been previously described. Thus, we investigated KLC4 function in lung cancer cells and radioresistant R-H460 cells by analyzing alterations in radiosensitivity after gene knockdown with siRNA and by evaluating cellular phenotypes and xenograft tumor growth. KLC4 was upregulated in human lung cancer cell lines. Moreover, in paired clinical specimens of lung cancer patients, KLC4 expression was significantly higher in tumor tissues than in paired adjacent normal tissues. Fluorescence-activated cell sorting (FACS) analysis showed that apoptosis rates and cleaved poly (ADP-ribose) polymerase (PARP) and cleaved caspase-3 levels in KLC4-knockdown lung cancer cells were significantly increased compared with those in control cells. Colony formation decreased as the radiation dose increased in KLC4-knockdown lung cancer cells, demonstrating an essential role for KLC4 in radioresistance. Importantly, KLC4 silencing suppressed tumor growth in an in vivo xenograft model, accompanied by increased apoptosis. Finally, KLC4-knockdown cells exhibited impaired mitochondrial respiration, increased mitochondrial reactive oxygen species production, and enhanced mitochondrial calcium uptake, resulting in mitochondrial dysfunction. Thus, KLC4 as a kinesin superfamily-targeted therapy may represent a novel, effective anticancer strategy, particularly for patients showing radioresistance.


Assuntos
Apoptose/efeitos da radiação , Sinalização do Cálcio/efeitos da radiação , Neoplasias Pulmonares/radioterapia , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/efeitos da radiação , Tolerância a Radiação , Neoplasias do Colo do Útero/radioterapia , Células A549 , Animais , Caspase 3/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Cinesinas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Carga Tumoral/efeitos da radiação , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Int J Mol Sci ; 19(3)2018 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-29538343

RESUMO

We investigated whether ß-adrenergic antagonists attenuates dietary fat absorption through the regulation of pancreatic lipase (PNLIP) expression in pancreatic acinar cells in the context of high fat diet feeding. Male six-week-old C57BL/6 mice were assigned into an ad libitum fed control diet (CON) and a high fat diet (HIGH). Within each diet group, subgroups of mice were treated with vehicle (VEH) or propranolol, a ß-adrenergic antagonist (BB). Over 12 weeks, body weight gain observed in HIGHVEH was mitigated in HIGHBB (+103% vs. +72%). Increase in fecal fat amount observed in HIGHVEH was further increased in HIGHBB. Increase in PNLIP expressions observed in HIGHVEH pancreatic tissues was abolished in HIGHBB. PNLIP expression in mouse primary pancreatic acinar cells and 266-6 cell lines increased with isoproterenol treatment, which was blocked by propranolol. Isoproterenol increased PNLIP expression in a cAMP/protein kinase A/ cyclic AMP response element binding protein (CREB)-dependent manner. CREB directly bound to the CRE on the mouse PNLIP promoter and transactivated PNLIP expression. These results suggest that sympathetic activation increases dietary fat absorption through the upregulation of PNLIP expression and that a ß-adrenergic antagonist attenuates obesity development partly through the downregulation of PNLIP expression and inhibition of dietary fat absorption in the context of high fat diet feeding.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Gorduras na Dieta/metabolismo , Absorção Intestinal/efeitos dos fármacos , Lipase/metabolismo , Obesidade/metabolismo , Propranolol/farmacologia , Células Acinares/metabolismo , Antagonistas Adrenérgicos beta/uso terapêutico , Animais , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dieta Hiperlipídica/efeitos adversos , Células HEK293 , Humanos , Lipase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/prevenção & controle , Pâncreas/citologia , Pâncreas/metabolismo , Propranolol/uso terapêutico , Transdução de Sinais
17.
Oncol Rep ; 39(4): 1565-1572, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29484424

RESUMO

The role of end-binding protein 1 (EB1) in lung cancer tumorigenesis and radiotherapy remains poorly understood. In the present study, we observed that EB1 was highly expressed in lung tumor tissues compared with normal non-tumor tissues based on immunohistochemical analysis of lung cancer tissue samples obtained from human tissue microarrays. EB1 was also highly overexpressed in radioresistant lung and cervical cancer cells, which exhibited increased cell death after EB1 silencing. The cytotoxicity induced by EB1 gene knockdown was due to the activation and generation of reactive oxygen species by p38 mitogen-activated protein kinase. Notably, this signaling cascade, however not nuclear factor-κB-mediated signaling, induced the expression of cyclooxygenase-2, a key effector of apoptotic death. Our results provided new molecular evidence supporting the use of EB1 as a novel target in lung cancer therapy, especially in the case of radioresistance.


Assuntos
Ciclo-Oxigenase 2/genética , Neoplasias Pulmonares/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Células A549 , Apoptose/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/genética , Espécies Reativas de Oxigênio/metabolismo , Análise Serial de Tecidos
18.
Int J Mol Sci ; 19(1)2018 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-29315243

RESUMO

Hyperglycemic conditions in diabetic patients can affect various cellular functions, including the modulation of osteogenic differentiation. However, the molecular mechanisms by which hyperglycemia affects osteogenic differentiation are yet to be clarified. This study aimed to investigate whether the aberrant increase in protein O-linked-ß-N-acetylglucosamine glycosylation (O-GlcNAcylation) contributes to the suppression of osteogenic differentiation due to hyperglycemia. To induce osteogenic differentiation, C2C12 cells were cultured in the presence of recombinant human bone morphogenetic protein 2 (BMP2). Excessive protein O-GlcNAcylation was induced by treating C2C12 cells with high glucose, glucosamine, or N-acetylglucosamine concentrations or by O-GlcNAc transferase (OGT) overexpression. The effect of O-GlcNAcylation on osteoblast differentiation was then confirmed by examining the expression levels of osteogenic marker gene mRNAs, activity of alkaline phosphatase, and transcriptional activity of Runx2, a critical transcription factor for osteoblast differentiation and bone formation. Cell treatment with high glucose, glucosamine or N-acetylglucosamine increased O-GlcNAcylation of Runx2 and the total levels of O-GlcNAcylated proteins, which led to a decrease in the transcriptional activity of Runx2, expression levels of osteogenic marker genes (Runx2, osterix, alkaline phosphatase, and type I collagen), and activity of alkaline phosphatase. These inhibitory effects were rescued by lowering protein O-GlcNAcylation levels by adding STO45849, an OGT inhibitor, or by overexpressing ß-N-acetylglucosaminidase. Our findings suggest that excessive protein O-GlcNAcylation contributes to high glucose-suppressed osteogenic differentiation.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Animais , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Inibidores Enzimáticos/farmacologia , Glucosamina/farmacologia , Glucose/farmacologia , Glicosilação/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , N-Acetilglucosaminiltransferases/antagonistas & inibidores , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Ligamento Periodontal/citologia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Transcrição Sp7/genética , Fator de Transcrição Sp7/metabolismo , Transcrição Gênica/efeitos dos fármacos
19.
Int J Mol Sci ; 18(10)2017 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-29053621

RESUMO

Sympathetic nervous system stimulation-induced ß-adrenergic signal transduction is known to induce bone loss and increase of osteoclast activity. Although isoproterenol, a nonspecific ß-adrenergic receptor agonist, has been shown to increase receptor activator of NF-κB ligand (RANKL), the details of the regulatory mechanisms remain unclear. In the present study, we investigated the role of the nuclear factor of activated T-cells (NFAT) in isoproterenol-induced RANKL expression in C2C12 and in primary cultured mouse calvarial cells. Isoproterenol increased nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) and RANKL expressions at both mRNA and protein levels and increased NFAT reporter activity. NFATc1 knockdown blocked isoproterenol-mediated RANKL expression. Isoproterenol also promoted cAMP response element-binding protein 1 (CREB1) and activating transcription factor 4 (ATF4) phosphorylation. Isoproterenol-mediated transcriptional activation of NFAT was blocked by protein kinase A (PKA) inhibitor H89. Isoproterenol-induced CREB1, ATF4, NFATc1, and RANKL expressions were suppressed by H89. Mutations in cAMP response element-like or NFAT-binding element suppressed isoproterenol-induced RANKL promoter activity. Chromatin immunoprecipitation analysis demonstrated that isoproterenol increased NFAT-binding and ATF4-binding activities on the mouse RANKL promoter, but did not increase CREB1-binding activity. Association of NFATc1 and ATF4 was not observed in a co-immunoprecipitation study. ATF4 knockdown suppressed isoproterenol-induced NFAT binding to the RANKL promoter, whereas NFATc1 knockdown did not suppress isoproterenol-induced ATF4 binding to the RANKL promoter. ATF4 knockdown suppressed isoproterenol-induced expressions of NFATc1 and RANKL. These results suggest that isoproterenol increases RANKL expression in an ATF4/NFATc1-dependent manner.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Isoproterenol/farmacologia , Fatores de Transcrição NFATC/genética , Ligante RANK/genética , Animais , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Camundongos , Mutação , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Fosforilação , Regiões Promotoras Genéticas , Ligante RANK/metabolismo , Transdução de Sinais
20.
Biomaterials ; 149: 77-87, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29017079

RESUMO

Host responses to a biomaterial critically influence its in vivo performance. Biomaterial architectures that can recruit endogenous host stem cells could be beneficial in tissue regeneration or integration. Here, we report that the fibrous topography of biomaterials promotes the recruitment of host mesenchymal stem cells (MSCs) by facilitating the macrophage phenotype transition from M1-to-M2. Electrospun poly (ε-caprolactone) fiber (PCL-fiber) films were implanted into the subcutaneous tissues of rats, and the response of host cells to the PCL-fiber was evaluated and compared with those of solid ones (PCL-solid). During the initial post-implantation period, greater numbers of cells were recruited and adhered to the PCL-fiber compared to the PCL-solid, and the cells exhibited the M1 phenotype, which was supported by the enhanced adsorption of complement C3a to the implanted PCL-fiber. Subsequently, the PCL-fiber supported the macrophage phenotype transition from M1-to-M2, which was confirmed by the ratio of M2/M1 marker (CD163/CCR7)-positive cells and by the expression of M2/M1 markers (arginase-1/iNOS). The PCL-fiber also reduced the formation of foreign body giant cells. MSC marker (CD29, CD44, and CD90)-positive cells began to appear as early as day 4 on the PCL-fiber, while few MSCs were observed on the PCL-solid. The MSCs migration ex vivo assay showed that MSCs substantially migrated across the trans-wells toward the implanted PCL-fiber. The cells on the implanted PCL-fiber expressed and secreted substantial levels of SDF-1 (CXCL-12), while anti-SDF-1 neutralizing antibody abrogated the MSCs migration. Taken together, these results provide evidence that the fibrous topography of biomaterials enhances the recruitment of MSCs by promoting macrophage recruitment, facilitating M1-to-M2 transition, and enhancing SDF-1 secretion.


Assuntos
Macrófagos/citologia , Células-Tronco Mesenquimais/fisiologia , Poliésteres/química , Animais , Biomarcadores/metabolismo , Adesão Celular , Movimento Celular , Quimiocina CXCL12/metabolismo , Humanos , Macrófagos/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Fenótipo , Ratos Sprague-Dawley , Alicerces Teciduais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA