Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 19.759
Filtrar
1.
Mol Omics ; 19(7): 585-597, 2023 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-37345535

RESUMO

Photochemical internalization (PCI) is a promising new technology for site-specific drug delivery, developed from photodynamic therapy (PDT). In PCI, light-induced activation of a photosensitizer trapped inside endosomes together with e.g. chemotherapeutics, nucleic acids or immunotoxins, allows cytosolic delivery and enhanced local therapeutic effect. Here we have evaluated the photosensitizer meso-tetraphenyl chlorine disulphonate (TPCS2a/fimaporfin) in a proteome analysis of AY-27 rat bladder cancer cells in combination with the chemotherapeutic drug bleomycin (BML). We find that BLMPCI attenuates oxidative stress responses induced by BLM alone, while concomitantly increasing transcriptional repression and DNA damage responses. BLMPCI also mediates downregulation of bleomycin hydrolase (Blmh), which is responsible for cellular degradation of BLM, as well as several factors known to be involved in fibrotic responses. PCI-mediated delivery might thus allow reduced dosage of BLM and alleviate unwanted side effects from treatment, including pulmonary fibrosis.


Assuntos
Bleomicina , Fotoquímica , Proteômica , Neoplasias da Bexiga Urinária , Bleomicina/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Proteínas Supressoras de Tumor/metabolismo , Regulação para Baixo/efeitos dos fármacos , Animais , Ratos , Linhagem Celular Tumoral , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética
2.
Sci Rep ; 12(1): 10423, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35729230

RESUMO

Blocking CD73 ectonucleotidase has been proposed as a potential therapeutic approach for cancer treatment. The present study aimed to investigate the antitumor effect of a novel EGFR-Targeted liposomal CD73 siRNA formulation in combination therapy with liposomal doxorubicin in the 4T1 mouse model. CD73 siRNA was encapsulated into nanoliposomes by the ethanol injection method. After preparation, characterization, morphology, and stability evaluation of formulations, the toxicity was measured by MTT assay. Uptake assay and efficiency of the liposomal formulations were investigated on the 4T1 cell line. The liposomal formulation containing CD73 siRNA was targeted with GE11 peptide for in vivo evaluations. Following biodistribution analysis, the antitumor activity of prepared formulations in combination with liposomal doxorubicin was studied in mice bearing 4T1 metastatic breast cancer cells. Finally, the induction of immune response of formulations in concomitant treatment with liposomal doxorubicin was evaluated in the tumor microenvironment of a mouse model of breast cancer. The size of prepared liposomal formulations at N/P = 16 for the liposomal CD73 siRNA and GE11-liposomal CD73 siRNA groups were 89 nm ± 4.4 and 95 nm ± 6.6, respectively. The nanoparticle's PDI was less than 0.3 and their surface charge was below 10 mV. The results demonstrated that N/P = 16 yielded the best encapsulation efficiency which was 94% ± 3.3. AFM results showed that the liposomes were spherical in shape and were less than 100 nm in size. The results of the MTT assay showed significant toxicity of the liposomes containing CD73 siRNA during the 48-h cell culture. Real-time PCR and flow cytometry results showed that liposomes containing CD73 siRNA could effectively downregulate CD73 expression. Liposomal formulations were able to significantly downregulate CD73 gene expression, in vivo. However, CD73 downregulation efficiency was significantly higher for the targeted form compared to the non-targeted formulation (P value < 0.01). The combination showed maximum tumor growth delay with remarkable survival improvement compared to the control group. Studying the immune responses in the treatment groups which received doxorubicin, showed decreased number of lymphocytes in the tumor environment. However, this decrease was lower in the combination therapy group. Finally, our results clearly showed that CD73 downregulation increases the activity of CD8+ lymphocytes (IFN-ℽ production) and also significantly decreases the Foxp3 in the CD25+ lymphocytes compared to the control group. GE11-Lipo CD73 siRNA formulation can efficiently knockdown CD73 ectonucleotidase. Also, the efficacy of liposomal doxorubicin is significantly enhanced via the downregulation of CD73 ectonucleotidase.


Assuntos
Neoplasias da Mama , Doxorrubicina , Receptores ErbB , Lipossomos , RNA Interferente Pequeno , 5'-Nucleotidase/genética , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Doxorrubicina/análogos & derivados , Doxorrubicina/uso terapêutico , Receptores ErbB/metabolismo , Feminino , Proteínas Ligadas por GPI/genética , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Camundongos , Terapia de Alvo Molecular , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , RNA Interferente Pequeno/metabolismo , Distribuição Tecidual , Microambiente Tumoral
3.
Phytomedicine ; 102: 154183, 2022 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-35636176

RESUMO

BACKGROUND: Esophageal cancer (EC) is highly prevalent in Eastern Asia (including China) with high rates of mortality. The metastatic tendency in EC is associated with a poor prognosis. Our previous studies have demonstrated the suppressive effects of Andrographis paniculata water extract (APW) on metastatic esophageal cancer in vitro and in tumor-bearing mice models, as well as illustrated the potential underlying mechanism by transcriptome analysis. HYPOTHESIS: High expressions of several membrane protein tetraspanins were reported to lead to a high risk of metastasis in esophageal cancer in patients. We hypothesized that APW could downregulate the expression of tetraspanin CD81 in esophageal cancer cells and xenografts. METHODS: Human esophageal cancer cells EC109 and KYSE520 were incubated with APW for 24 hours in cell culture, while mice bearing EC109 xenograft tumors were treated with APW for 21 days. The expressions of CD81 in cancer cells and in tumors from mice were evaluated. Molecular docking and microscale thermophoresis analyses were applied to identify the components in APW interacting with CD81. The influence of the identified components on CD81 expression was further evaluated in EC109 cells. RESULTS: APW could significantly suppress the expressions of CD81 in both EC109 and KYSE520 cells in a concentration-dependent manner. Treatment of APW in xenograft-bearing mice reduces the metastasis in lungs, livers, and lymph nodes. The expression of CD81 in xenograft tumors of APW-treated mice was significantly lower than those of untreated control mice. The binding of andrographolide, bisandrographolide A, and bisandrographolide C with CD81 were elucidated by microscale thermophoresis. The suppressive effects of these compounds on the motility of EC109 cells, as well as CD81 protein and mRNA expressions, were further confirmed. CONCLUSION: This is the first time to demonstrate that andrographolide, bisandrographolide A, and bisandrographolide C, which are present in APW, bind to CD81 and suppress its function. These compounds are likely to be responsible for the anti-metastatic activities of APW in esophageal cancer.


Assuntos
Andrographis paniculata , Diterpenos , Neoplasias Esofágicas , Extratos Vegetais/química , Tetraspanina 28 , Andrographis paniculata/química , Animais , Linhagem Celular Tumoral , Diterpenos/química , Regulação para Baixo/efeitos dos fármacos , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Humanos , Camundongos , Simulação de Acoplamento Molecular , Terapia de Alvo Molecular , Extratos Vegetais/farmacologia
4.
Cell Oncol (Dordr) ; 45(3): 355-365, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35579750

RESUMO

PURPOSE: N6-methyladenosine (m6A), the most prevalent mRNA modification, plays an essential role in tumorigenesis. Notably, increasing interest has been directed to bioactive peptides (BPs) with antitumor activities. Here, we set out to investigate the potential of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis on prevention and treatment of acute myeloid leukemia (AML). METHODS: The biological effects of BP on AML cells were detected by MTT and ApoLive-Glo™ multiplex assays. The role of BP in tumor growth was determined by a subcutaneous xenograft model. The ALKBH5/MLST8/EIF4EBP1 axis was identified as a potential BP target in AML via methylated RNA immunoprecipitation sequencing (MeRIP-seq) combined with RNA sequencing (RNA-seq). Western blot, RT-qPCR, MeRIP-qPCR, dual-luciferase reporter and RNA stability assays were performed to validate the function and mode of action of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis. The clinical relevance of the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis in AML was confirmed by TCGA data analysis. RESULTS: We found that BP can inhibit AML cell proliferation and promote apoptosis in vitro, and repress AML tumor growth in vivo. Mechanistically, we found that BP downregulated ALKBH5 expression, which in turn repressed m6A demethylation of MLST8 and EIF4EBP1 mRNAs. Reduction of the m6A levels of MLST8 and EIF4EBP1 facilitated MLST8 and EIF4EBP1 mRNA decay, resulting in inhibition of AML cell proliferation. Furthermore, we found that the BP-regulated ALKBH5/MLST8/EIF4EBP1 axis closely correlates with AML patient prognosis. CONCLUSIONS: Our data indicate that BP can inhibit acute myeloid leukemia cell proliferation by downregulating ALKBH5-mediated m6A demethylation of EIF4EBP1 and MLST8 mRNAs, which may have potential to prevent and treat this disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Homólogo AlkB 5 da RNA Desmetilase , Proteínas de Ciclo Celular , Leucemia Mieloide Aguda , Peptídeos , Homólogo LST8 da Proteína Associada a mTOR , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Homólogo AlkB 5 da RNA Desmetilase/genética , Homólogo AlkB 5 da RNA Desmetilase/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Desmetilação/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Peptídeos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homólogo LST8 da Proteína Associada a mTOR/genética , Homólogo LST8 da Proteína Associada a mTOR/metabolismo
5.
Front Immunol ; 13: 820131, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251001

RESUMO

Coronavirus disease 2019 (COVID-19) is currently a worldwide emergency caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). In observational clinical studies, statins have been identified as beneficial to hospitalized patients with COVID-19. However, experimental evidence of underlying statins protection against SARS-CoV-2 remains elusive. Here we reported for the first-time experimental evidence of the protective effects of simvastatin treatment both in vitro and in vivo. We found that treatment with simvastatin significantly reduced the viral replication and lung damage in vivo, delaying SARS-CoV-2-associated physiopathology and mortality in the K18-hACE2-transgenic mice model. Moreover, simvastatin also downregulated the inflammation triggered by SARS-CoV-2 infection in pulmonary tissue and in human neutrophils, peripheral blood monocytes, and lung epithelial Calu-3 cells in vitro, showing its potential to modulate the inflammatory response both at the site of infection and systemically. Additionally, we also observed that simvastatin affected the course of SARS-CoV-2 infection through displacing ACE2 on cell membrane lipid rafts. In conclusion, our results show that simvastatin exhibits early protective effects on SARS-CoV-2 infection by inhibiting virus cell entry and inflammatory cytokine production, through mechanisms at least in part dependent on lipid rafts disruption.


Assuntos
Tratamento Farmacológico da COVID-19 , Regulação para Baixo/efeitos dos fármacos , Inflamação/tratamento farmacológico , Microdomínios da Membrana/efeitos dos fármacos , SARS-CoV-2/patogenicidade , Sinvastatina/farmacologia , Animais , COVID-19/virologia , Modelos Animais de Doenças , Humanos , Inflamação/virologia , Pulmão/virologia , Camundongos , Camundongos Transgênicos , Replicação Viral/efeitos dos fármacos
6.
BMC Cancer ; 22(1): 238, 2022 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-35241028

RESUMO

BACKGROUND: Circular RNAs (circRNAs) are well-known regulators of cancer progression and chemoresistance in various types of cancers. This study was performed to investigate the function of hsa_circ_0000277 in esophageal squamous cell carcinoma (ESCC). METHODS: RNA levels were analyzed via the reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8 (CCK-8) assay was applied to determine cell proliferation and half maximal inhibitory concentration (IC50) of cisplatin (DDP). Colony formation ability was evaluated by colony formation assay. Cell cycle and apoptosis were measured using flow cytometry. RNA immunoprecipitation (RIP), pull-down assay and dual-luciferase reporter assays were performed for target interaction analysis. The protein levels were determined through western blot. Xenograft models were established for researching hsa_circ_0000277 function in vivo. RESULTS: Hsa_circ_0000277 expression was increased in ESCC cells and tissues, and it had important clinical significance. Downregulation of hsa_circ_0000277 repressed ESCC cell proliferation, colony formation, cell cycle, and DDP resistance. Hsa_circ_0000277 acted as a microRNA-873-5p (miR-873-5p) sponge and Sry-related high-mobility group box 4 (SOX4) was validated as a target of miR-873-5p. Moreover, hsa_circ_0000277/miR-873-5p axis and miR-873-5p/SOX4 axis regulated ESCC cell progression and DDP resistance. Hsa_circ_0000277/miR-873-5p axis activated SOX4/Wnt/ß-catenin signaling pathway. Hsa_circ_0000277 facilitated tumorigenesis and DDP resistance by miR-873-5p/SOX4 axis in vivo. CONCLUSION: These findings unraveled that hsa_circ_0000277 promoted ESCC progression and DDP resistance via miR-873-5p/SOX4/Wnt/ß-catenin axis, showing a specific molecular mechanism of carcinogenesis and chemoresistance in ESCC.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , RNA Circular/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Esofágicas/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Humanos , MicroRNAs/efeitos dos fármacos , Fatores de Transcrição SOXC/efeitos dos fármacos , Proteínas Wnt/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/efeitos dos fármacos
7.
J Orthop Surg Res ; 17(1): 133, 2022 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241114

RESUMO

BACKGROUND: Mitochondrial calcium uniporter (MCU) is an important ion channel regulating calcium transport across the mitochondrial membrane. Calcium signaling, particularly via the Ca2+/NFATc1 pathway, has been identified as an important mediator of the osteoclast differentiation that leads to osteolysis around implants. The present study aimed to investigate whether down-regulation of MCU using microRNA-25 (miR-25) mimics could reduce osteoclast differentiation induced upon exposure to titanium (Ti) particles. METHODS: Ti particles were prepared. Osteoclast differentiation of RAW264.7 cells was induced by adding Ti particles and determined by TRAP staining. Calcium oscillation was determined using a dual-wavelength technique. After exposure of the cells in each group to Ti particles or control medium for 5 days, relative MCU and NFATc1 mRNA expression levels were determined by RT-qPCR. MCU and NFATc1 protein expression was determined by western blotting. NFATc1 activation was determined by immunofluorescence staining. Comparisons among multiple groups were conducted using one-way analysis of variance followed by Tukey test, and differences were considered significant if p < 0.05. RESULTS: MCU expression was reduced in response to miR-25 overexpression during the process of RAW 264.7 cell differentiation induced by Ti particles. Furthermore, osteoclast formation was inhibited, as evidenced by the low amplitude of calcium ion oscillation, reduced NFATc1 activation, and decreased mRNA and protein expression levels of nuclear factor-κB p65 and calmodulin kinases II/IV. CONCLUSIONS: Regulation of MCU expression can impact osteoclast differentiation, and the underlying mechanism likely involves the Ca2+/NFATc1 signal pathway. Therefore, MCU may be a promising target in the development of new strategies to prevent and treat periprosthetic osteolysis.


Assuntos
Canais de Cálcio , MicroRNAs , Osteoclastos/metabolismo , Osteólise , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , MicroRNAs/metabolismo , Osteólise/induzido quimicamente , Ligante RANK/genética , RNA Mensageiro/metabolismo , Titânio/efeitos adversos
8.
Oxid Med Cell Longev ; 2022: 1269747, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35136484

RESUMO

Sepsis is a systemic inflammatory response syndrome caused by a dysregulated host response to infection. Peroxisome proliferator-activated receptor gamma (PPARγ) exerts anti-inflammatory and antioxidative properties. To investigate the potential effects of PPARγ on sepsis-induced liver injury and determine the related mechanisms, C57BL/6 male mice were subjected to cecal ligation and puncture (CLP) to create a sepsis model which was treated with GW1929 or GW9662 to upregulate or downregulate the expression of PPARγ. We found that upregulation of PPARγ decreased the serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (TBIL), and liver pathological damage and improved the 5-day survival rate. Increased expression of PPARγ also decreased sepsis-induced reactive oxygen species (ROS) by promoting the expression of Nrf2. In addition, upregulated PPARγ inhibited the expression of the TXNIP/NLRP3 signaling pathway by reducing ROS-induced injury in the liver during sepsis, which further reduced NLRP3-mediated pyroptosis and the inflammatory response. The role of PPARγ was further examined in in vitro experiments, where lipopolysaccharide- (LPS-) treated HepG2 and Hep3B cells were incubated with GW1929 or GW9662 to upregulate or downregulate the expression of PPARγ. We found that upregulated PPARγ ameliorated LDH release and improved cell viability. Our results indicated that increased expression of PPARγ reduced ROS levels and inhibited the TXNIP/NLRP3 signaling pathway, resulting in decreased pyroptosis and reduced liver dysfunction during sepsis.


Assuntos
Proteínas de Transporte/metabolismo , Hepatócitos/metabolismo , Hepatopatias/etiologia , Hepatopatias/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , PPAR gama/metabolismo , Piroptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Sepse/complicações , Transdução de Sinais/efeitos dos fármacos , Tiorredoxinas/metabolismo , Anilidas/administração & dosagem , Animais , Benzofenonas/administração & dosagem , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Células Hep G2 , Humanos , Hepatopatias/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , Resultado do Tratamento , Tirosina/administração & dosagem , Tirosina/análogos & derivados , Regulação para Cima/efeitos dos fármacos
9.
Chem Biol Interact ; 355: 109804, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-35123994

RESUMO

Recent reports have challenged the notion that the lens is immune-privileged. However, these studies have not fully identified the molecular mechanism(s) that promote immune surveillance of the lens. Using a mouse model of targeted glutathione (GSH) deficiency in ocular surface tissues, we have investigated the role of oxidative stress in upregulating cytokine expression and promoting immune surveillance of the eye. RNA-sequencing of lenses from postnatal day (P) 1-aged Gclcf/f;Le-CreTg/- (KO) and Gclcf/f;Le-Cre-/- control (CON) mice revealed upregulation of many cytokines (e.g., CCL4, GDF15, CSF1) and immune response genes in the lenses of KO mice. The eyes of KO mice had a greater number of cells in the aqueous and vitreous humors at P1, P20 and P50 than age-matched CON and Gclcw/w;Le-CreTg/- (CRE) mice. Histological analyses revealed the presence of innate immune cells (i.e., macrophages, leukocytes) in ocular structures of the KO mice. At P20, the expression of cytokines and ROS content was higher in the lenses of KO mice than in those from age-matched CRE and CON mice, suggesting that oxidative stress may induce cytokine expression. In vitro administration of the oxidant, hydrogen peroxide, and the depletion of GSH (using buthionine sulfoximine (BSO)) in 21EM15 lens epithelial cells induced cytokine expression, an effect that was prevented by co-treatment of the cells with N-acetyl-l-cysteine (NAC), a antioxidant. The in vivo and ex vivo induction of cytokine expression by oxidative stress was associated with the expression of markers of epithelial-to-mesenchymal transition (EMT), α-SMA, in lens cells. Given that EMT of lens epithelial cells causes posterior capsule opacification (PCO), we propose that oxidative stress induces cytokine expression, EMT and the development of PCO in a positive feedback loop. Collectively these data indicate that oxidative stress induces inflammation of lens cells which promotes immune surveillance of ocular structures.


Assuntos
Olho/anatomia & histologia , Imunidade Inata , Cristalino/metabolismo , Estresse Oxidativo , Acetilcisteína/farmacologia , Animais , Butionina Sulfoximina/farmacologia , Linhagem Celular , Quimiocina CCL7/genética , Quimiocina CCL7/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Olho/metabolismo , Glutamato-Cisteína Ligase/deficiência , Glutamato-Cisteína Ligase/genética , Cristalino/citologia , Leucócitos/citologia , Leucócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Biomed Pharmacother ; 147: 112666, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35124384

RESUMO

Naturally occurring phytochemicals especially polyphenolic compounds have received increasing attention as chemopreventive agents. The chemopreventive potential of the ethanolic extract of Salvadora persica L. fruits SP, (the arak tree or miswak) on 7,12-dimethylbenz (a) anthracene (DMBA)-induced mammary carcinogenesis in female albino rats was investigated in this work. Ethanolic extract of SP fruits was supplemented to the experimental groups at a concentration of 500 mg/kg body weight for 22 weeks. Administration of SP extract suppressed DMBA-induced mammary carcinogenesis as revealed by incidence of tumors in histological investigation. There was a significant reduction in cell proliferation and an increase in apoptosis with downregulation of estrogen receptor expression in the mammary tissue of SP-treated animals. Additionally, SP extract prevented the oxidative damage induced in breast tissues of DMBA-treated rats. SP treatment also decreased the viability of MCF-7 breast cancer cells and induced early and late apoptosis and induced S cell cycle arrest. The chemo-preventive properties and anticancer effects of SP could be attributed to its anti-oxidative and a high percentage of phenolic compounds and esters which were detected here in the SP fruit extract.


Assuntos
Neoplasias Mamárias Experimentais/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Receptores de Estrogênio/efeitos dos fármacos , Salvadoraceae , 9,10-Dimetil-1,2-benzantraceno/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Células MCF-7 , Distribuição Aleatória , Ratos , Ratos Wistar , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Sci Rep ; 12(1): 1975, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-35132122

RESUMO

Aphelenchoides besseyi could cause great yield losses of rice and many economically important crops. Acetylcholinesterase (AChE) inhibitors were commonly used to manage plant-parasitic nematodes. However, nematodes resistant to AChE inhibitors have been increasingly reported due to the extensive use of these chemicals. The current study was aimed to establish the correlation between fenamiphos (an AChE-inhibitor) sensitivities and acetylcholinesterase genes (ace) by analyzing two isolates of A. besseyi (designated Rl and HSF), which displayed differential sensitivities to fenamiphos. The concentrations of fenamiphos that led to the death of 50% (LD50) of Rl and HSF were 572.2 ppm and 129.4 ppm, respectively. Three ace genes were cloned from A. besseyi and sequenced. Sequence searching and phylogenic analyses revealed that AChEs of R1 and HSF shared strong similarities with those of various vertebrate and invertebrate species. Molecular docking analysis indicated that AChEs-HSF had much higher affinities to fenamiphos than AChEs-R1. Quantitative reverse transcriptase-PCR analyses revealed that expression of three ace genes were downregulated in HSF but were upregulated in Rl after exposure to 100 ppm fenamiphos for 12 h. The results indicated that the expression of the ace genes was modulated in response to fenamiphos in different nematode strains. An increased expression of the ace genes might contribute to fenamiphos-insensitivity as seen in the Rl isolate.


Assuntos
Acetilcolinesterase/genética , Inibidores da Colinesterase/farmacologia , Expressão Gênica , Nematoides/efeitos dos fármacos , Nematoides/genética , Compostos Organofosforados/farmacologia , Acetilcolinesterase/metabolismo , Animais , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Dose Letal Mediana , Simulação de Acoplamento Molecular , Regulação para Cima/efeitos dos fármacos
12.
Oxid Med Cell Longev ; 2022: 6227330, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35126818

RESUMO

BACKGROUND: Heart failure is characterized by activation of the renin-angiotensin-aldosterone system, which is involved in the regulation of cardiac hypertrophy and hypertension. Recently, we reported that Hdac8 inhibition alleviates isoproterenol-induced and angiotensin II-induced cardiac hypertrophy or hypertension in mice. Here, the effect and regulatory mechanisms of the Hdac8 selective inhibitor PCI34051 on pressure overload-induced heart failure were examined. METHODS AND RESULTS: At week 6 posttransverse aortic constriction (TAC), mice were administered with PCI34051 (3, 10, or 30 mg/kg bodyweight/day) for 2 weeks. The therapeutic effects of PCI34051 on TAC-induced cardiac and lung hypertrophy were determined by examining the heart weight-to-bodyweight and lung weight-to-bodyweight ratios and the cross-sectional cardiomyocyte area. Echocardiography analysis revealed that PCI34051 mitigated TAC-induced decreased ejection fraction and fractional shortening. Additionally, the expression of Hdac8 was upregulated in the cardiac and pulmonary tissues of TAC mice. The expression levels of Ace1 and Agtr1 were upregulated, whereas those of Ace2 and Agtr2 were downregulated in TAC mice. PCI34051 treatment or Hdac8 knockdown alleviated inflammation as evidenced by Rela downregulation and Nfkbia upregulation in mice, as well as in cardiomyocytes, but not in cardiac fibroblasts. Hdac8 overexpression-induced Rela pathway activation was downregulated in Ace1 knockdown cells. Picrosirius red staining, real-time polymerase chain reaction, and western blotting analyses revealed that PCI34051 alleviated fibrosis and downregulated fibrosis-related genes. Moreover, PCI34051 or Hdac8 knockdown in rat cardiac fibroblasts alleviated cardiac fibrosis through the Tgfb1-Smad2/3 pathway. The results of overexpression and knockdown experiments revealed that Hdac8 and Ace1 promote inflammation and fibrosis. CONCLUSIONS: Treatment with PCI34051 enhanced cardiac and lung functions in the TAC-induced heart failure mouse model. These data suggest that HDAC8 is a potential novel therapeutic target for heart failure accompanied by pathological lung diseases.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Insuficiência Cardíaca/patologia , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/farmacologia , Indóis/farmacologia , Peptidil Dipeptidase A/metabolismo , Animais , Aorta Torácica/cirurgia , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Histona Desacetilases/química , Histona Desacetilases/genética , Ácidos Hidroxâmicos/uso terapêutico , Indóis/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos ICR , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia
13.
Int J Oncol ; 60(3)2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35179222

RESUMO

Regulator of ribosome synthesis 1 (RRS1) is a key factor in ribosome biosynthesis and other cellular functions. High level of RRS1 in breast cancer cell lines is associated with increased cell proliferation, invasion and migration. RRS1 controls the assembly of the 60s subunit and maturation of 25S rRNA during ribosome biosynthesis. In this study, lentiviral transfection of sh­RNA was used to knock down the level of RRS1, to detect the effect of RRS1 on cell function and to explore the specific mechanism of RRS1 affecting cell invasion and metastasis by COIP and dual­luciferase reporter gene assays. The present study found that RRS1 knockdown reduced the accumulation of ribosome protein L11 (RPL11) in the nucleolus, which then migrated to the nucleoplasm and bound to c­Myc. This inhibited trans­activation of SNAIL by c­Myc and eventually decreased the invasion and metastasis capacity of the human breast cancer cell line BT549. Taken together, RRS1 regulates invasion and metastasis of human breast cancer cells through the RPL11­c­Myc­SNAIL axis. The findings are of great significance for exploring the mechanism of breast cancer invasion and metastasis and the corresponding regulatory factors.


Assuntos
Regulação para Baixo/genética , Metástase Neoplásica/genética , Proteínas de Ligação a RNA/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Proliferação de Células/genética , Proteínas de Ligação a DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Humanos , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Fatores de Transcrição da Família Snail/efeitos dos fármacos , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/genética
14.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35163281

RESUMO

Calycosin, a bioactive isoflavonoid isolated from root extracts of Astragalus membranaceus, has been reported to inhibit melanogenesis, the mechanism of which remains undefined. In this study, we interrogated the mechanistic basis by which calycosin inhibits melanin production in two model systems, i.e., B16F10 melanoma cells and zebrafish embryos. Calycosin was effective in protecting B16F10 cells from α-melanocyte-stimulating hormone (α-MSH)-induced melanogenesis and tyrosinase activity. This anti-melanogenic effect was accompanied by decreased expression levels of microphthalmia-associated transcription factor (MITF), a key protein controlling melanin synthesis, and its target genes tyrosinase and tyrosinase-related protein-2 (TRP-2) in calycosin-treated cells. Mechanistically, we obtained the first evidence that calycosin-mediated MITF downregulation was attributable to its ability to block signaling pathways mediated by cAMP response element-binding protein (CREB) and p38 MAP kinase. The protein kinase A (PKA) inhibitor H-89 and p38 inhibitor SB203580 validated the premise that calycosin inhibits melanin synthesis and tyrosinase activity by regulating the PKA/CREB and p38 MAPK signaling pathways. Moreover, the in vivo anti-melanogenic efficacy of calycosin was manifested by its ability to suppress body pigmentation and tyrosinase activity in zebrafish embryos. Together, these data suggested the translational potential of calycosin to be developed as skin-lightening cosmeceuticals.


Assuntos
Isoflavonas/farmacologia , Melaninas/metabolismo , Animais , Astragalus propinquus/metabolismo , Linhagem Celular Tumoral , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/efeitos dos fármacos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Isoflavonas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Fator de Transcrição Associado à Microftalmia/metabolismo , Fosforilação/efeitos dos fármacos , Extratos Vegetais/farmacologia , Raízes de Plantas , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/metabolismo , alfa-MSH/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Int J Mol Sci ; 23(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35163437

RESUMO

Insulin-like growth factor 1 (IGF-1) not only regulates neuronal function and development but also is neuroprotective in the setting of acute ischemic stroke. G-protein-coupled receptor 17 (GPR17) expression in brain tissue serves as an indicator of brain damage. As whether IGF-1 regulates GPR17 expression remains unknown, the aim of this study is to investigate how IGF-1 regulates GPR17 expression in vitro. Human neuroblastoma SK-N-SH cells were used. Lentivirus-mediated short hairpin RNA (shRNA) was constructed to mediate the silencing of FoxO1, while adenoviral vectors were used for its overexpression. Verification of the relevant signaling cascade was performed using a FoxO1 inhibitor (AS1842856), a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002), and a GPR17 antagonist (cangrelor). Cell proliferation was analyzed using EdU staining; immunofluorescence staining was used to detect the expression and subcellular localization of FoxO1. Chromatin immunoprecipitation was used to analyze the binding of FoxO1 to the GPR17 promoter in SK-N-SH cells. The expression of FoxO1, GPR17, and protein kinase B (also known as Akt) mRNA and protein as well as the levels of FoxO1 and Akt phosphorylation were investigated in this study. IGF-1 was found to downregulate FoxO1 and GPR17 expression in SK-N-SH cells while promoting cell viability and proliferation. Inhibition of FoxO1 and antagonism of GPR17 were found to play a role similar to that of IGF-1. Silencing of FoxO1 by lentivirus-mediated shRNA resulted in the downregulation of FoxO1 and GPR17 expression. The overexpression of FoxO1 via adenoviral vectors resulted in the upregulation of FoxO1 and GPR17 expression. Blocking of PI3K signaling by LY294002 inhibited the effect of IGF-1 on GPR17 suppression. Results from chromatin immunoprecipitation revealed that IGF-1 promotes FoxO1 nuclear export and reduces FoxO1 binding to the GPR17 promoter in SK-N-SH cells. Here, we conclude that IGF-1 enhances cell viability and proliferation in SK-N-SH cells via the promotion of FoxO1 nuclear export and reduction of FoxO1 binding to the GPR17 promoter via PI3K/Akt signaling. Our findings suggest that the enhancement of IGF-1 signaling to antagonize GPR17 serves as a potential therapeutic strategy in the management of acute ischemic stroke.


Assuntos
Regulação para Baixo , Proteína Forkhead Box O1/genética , Fator de Crescimento Insulin-Like I/metabolismo , Neurônios/citologia , Receptores Acoplados a Proteínas G/genética , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Cromonas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Técnicas de Inativação de Genes , Humanos , Lentivirus/fisiologia , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolonas/farmacologia , RNA Interferente Pequeno/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos
16.
Chem Biol Drug Des ; 99(6): 828-838, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35184389

RESUMO

Infantile hemangioma (IH) is a common benign endothelial cell tumor in infants and young children, sometimes accompanied by potential complications, and may develop into malignant tumors. Hemangioma endothelial cells (HemECs) are one of the main components of IH. Podophyllotoxin (PPT) has been reported to have many pharmacological activities, especially anti-tumor, but its high toxicity, poor water solubility, and serious gastrointestinal side effects limit its clinical application. In this study, we have designed and synthesized 20 ester derivatives by introducing Boc-amino acids or organic acids at the C-4 position of podophyllotoxin through esterification reactions. The cytotoxicity of these compounds was evaluated on HemECs. Changes in cell proliferation and apoptotic signaling pathways were studied by DAPI staining, colony formation assay, migration assay, measurement of reactive oxygen species (ROS) levels flow cytometry, and Western blot analysis. We found that eight of the compounds were more potent than PPT. Of these, compound V-31 was the most active (IC50  = 0.079 ± 0.0049 µM). Further research indicated that compound V-31 inhibited its proliferation and migration, increased the level of ROS in cells, and induced apoptosis by downregulating p-PI3K, p-Akt, and Bcl-2, and upregulating cleaved caspase-3 and Bax. Our research provides the first insight into the application of PPT derivatives in HemECs, may provide an effective medicine for IH treatment.


Assuntos
Células Endoteliais , Hemangioma , Fosfatidilinositol 3-Quinases , Podofilotoxina , Proteínas Proto-Oncogênicas c-akt , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Criança , Pré-Escolar , Regulação para Baixo/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Ésteres/metabolismo , Hemangioma/tratamento farmacológico , Hemangioma/metabolismo , Hemangioma/patologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Podofilotoxina/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Int J Mol Sci ; 23(3)2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35163660

RESUMO

Induced neural stem cells (iNSCs) reprogrammed from somatic cells hold great potentials for drug discovery, disease modelling and the treatment of neurological diseases. Although studies have shown that human somatic cells can be converted into iNSCs by introducing transcription factors, these iNSCs are unlikely to be used for clinical application due to the safety concern of using exogenous genes and viral transduction vectors. Here, we report the successful conversion of human fibroblasts into iNSCs using a cocktail of small molecules. Furthermore, our results demonstrate that these human iNSCs (hiNSCs) have similar gene expression profiles to bona fide NSCs, can proliferate, and are capable of differentiating into glial cells and functional neurons. This study collectively describes a novel approach based on small molecules to produce hiNSCs from human fibroblasts, which may be useful for both research and therapeutic purposes.


Assuntos
Diferenciação Celular , Fibroblastos/citologia , Células-Tronco Neurais/citologia , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fenômenos Eletrofisiológicos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
18.
PLoS One ; 17(2): e0262942, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35171916

RESUMO

This study was designed to explore the therapeutics and the mechanisms of a patented and marked gastric acid and intestine juice-resistant probiotics Bifidobacterium lactis BL-99 (B. lactis BL-99) on the intestinal inflammation and functions in the zebrafish models. After feeding for 6 hours, B. lactis BL-99 was fully retained in the larval zebrafish intestinal tract and stayed for over 24 hours. B. lactis BL-99 promoted the intestinal motility and effectively alleviated aluminum sulfate-induced larval zebrafish constipation (p < 0.01). Irregular high glucose diet induced adult zebrafish intestinal functional and metabolic disorders. After fed with B. lactis BL-99, IL-1ß gene expression was significantly down-regulated, and IL-10 and IL-12 gene levels were markedly up-regulated in this model (p < 0.05). The intestinal lipase activity was elevated in the adult zebrafish intestinal functional disorder model after B. lactis BL-99 treatment (p < 0.05), but tryptase content had no statistical changes (p > 0.05). B. lactis BL-99 improved the histopathology of the adult zebrafish intestinal inflammation, increased the goblet cell numbers, and up-and-down metabolites were markedly recovered after treatment of B. lactis BL-99 (p < 0.05). These results suggest that B. lactis BL-99 could relieve intestinal inflammation and promote intestinal functions, at least in part, through modulating intestinal and microbial metabolism to maintain intestinal health.


Assuntos
Bifidobacterium/fisiologia , Inflamação/terapia , Intestinos/metabolismo , Probióticos/uso terapêutico , Compostos de Alúmen/toxicidade , Animais , Constipação Intestinal/induzido quimicamente , Constipação Intestinal/patologia , Constipação Intestinal/terapia , Análise Discriminante , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Motilidade Gastrointestinal/efeitos dos fármacos , Glucose/farmacologia , Inflamação/induzido quimicamente , Inflamação/patologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Intestinos/microbiologia , Intestinos/patologia , Larva/efeitos dos fármacos , Larva/metabolismo , Probióticos/farmacologia , Regulação para Cima/efeitos dos fármacos , Peixe-Zebra/crescimento & desenvolvimento
19.
Molecules ; 27(4)2022 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-35208960

RESUMO

Melanoma is the most dangerous skin malignancy due to its strong metastatic potential with high mortality. Activation of crucial signaling pathways enforcing melanoma progression depends on phosphorylation of distinct tyrosine kinases and oxidative stress. We here investigated the effect of a bis-coumarin derivative [3, 3'- ((3″, 5'-Dichlorophenyl) methylene) bis (4-hydroxy-2H-chromen-2-one)] [3, 3'- (3, 5-DCPBC)] on human melanoma cell survival, growth, proliferation, migration, intracellular redox state, and deciphered associated signaling pathways. This derivative is toxic for melanoma cells and non-toxic for melanocytes, their benign counterpart, and fibroblasts. 3, 3'- (3, 5-DCPBC) inhibits cell survival, migration, and proliferation of different metastatic and non-metastatic melanoma cell lines through profound suppression of the phosphorylation of Epidermal Growth Factor receptor (EGFR) and proto-oncogene cellular sarcoma (c-SRC) related downstream pathways. Thus, 3, 3'- (3, 5-DCPBC) endowed with the unique property to simultaneously suppress phosphorylation of multiple downstream kinases, such as EGFR/JAK/STAT and EGFR/SRC and their corresponding transcription factors.


Assuntos
Cumarínicos , Regulação para Baixo/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Melanoma , Proteínas de Neoplasias/biossíntese , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Cumarínicos/química , Cumarínicos/farmacologia , Receptores ErbB/biossíntese , Receptores ErbB/genética , Humanos , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Proteínas de Neoplasias/genética , Fosforilação/efeitos dos fármacos , Fosfotransferases (Aceptor do Grupo Álcool)/genética
20.
Acta Biochim Pol ; 69(1): 85-89, 2022 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-35225439

RESUMO

Oral carcinoma is a lethal type of cancer associated with huge morbidity and mortality. Oral cancer patients show a very low survival chances even if diagnosed at early stages. The need for novel naturally occurring chemotherapeutic drugs increases due to high cost and toxicity of currently used clinical drugs. Present study was designed to investigate anticancer property of oroselol, keeping in view the medicinal potential showed by coumarin subclass furanocoumarins. MTT assay and clonogenic assays were implemented for viability assessments. Transmission electron microscopy was used for autophagic studies. The transwell chambers assay was used to investigate the migration and invasion. Western blotting was performed to determine the expressions levels of autophagy and PI3K/AKT signalling related proteins. Results showed that oroselol could potentially inhibit viability of oral cancer SSC-4 cells in time- and dose-reliant fashion. The antiproliferative effects were mediated through autophagy induction as indicated by formation of autophagosomes and enhanced LC3-l expressions and reduced LC3-II and p62 expressions. Cancer cell migration and invasion was potentially supressed by oroselol cell treatment. The PI3K/AKT signalling pathway was blocked potentially by oroselol in SSC-4 cells which showed reduced phosphorylation of PI3K and AKT. In conclusion, oroselol holds a significant potential to induce autophagy-related antiproliferative effects along with inhibition of cell migration, cell invasion, and PI3K/AKT signalling pathway. Therefore, oroselol may prove to be a lead molecule in oral cancer chemotherapy provided further in vivo and toxicological studies are performed on it.


Assuntos
Autofagia/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cumarínicos/farmacologia , Neoplasias Bucais/tratamento farmacológico , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Furocumarinas/farmacologia , Humanos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...