Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Life Sci ; 335: 122255, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37967792

RESUMO

BACKGROUND: Cancer metastasis is a major cause of cancer-related deaths, emphasizing the urgent need for effective therapies. Although it has been shown that GMI, a fungal protein from Ganoderma microsporum, could suppress primary tumor growth in a wide spectrum of cancer types, it is still unclear whether GMI exhibits anti-metastasis properties, particularly in lung cancers. Further investigation is needed. AIMS AND OBJECTIVES: The objective of this study is to investigate the potential inhibitory effects of GMI on lung cancer metastasis in vivo. Utilizing systematic and comprehensive approaches, our research aims to elucidate the underlying molecular mechanisms responsible for the anti-metastatic effects. MATERIALS AND METHODS: In vitro migration and cell adhesion assays addressed the epithelial-to-mesenchymal transition (EMT)-related phenotype. Proteomic and bioinformatic analyses identified the GMI-regulated proteins and cellular responses. GMI-treated LLC1-bearing mice were analyzed using IVIS Spectrum to assess the anti-metastatic effect. KEY FINDINGS: GMI inhibits EMT as well as cell migration. GMI disrupts cell adhesion and downregulates integrin, resulting in inhibition of phosphorylated FAK. GMI induces macropinocytosis and lysosome-mediated degradation of integrin αv, α5, α6 and ß1. GMI downregulates Slug via inhibition of FAK activity, which in turn enhances expressions of epithelial-related markers and decreases cell mobility. Mechanistically, GMI-induced FAK inhibition engenders MDM2 expression and enhances MDM2/p21/Slug complex formation, leading to Slug degradation. GMI treatment reduces the metastatic pulmonary lesion and prolongs the survival of LLC1-bearing mice. SIGNIFICANCE: Our findings highlight GMI as a promising therapeutic candidate for metastatic lung cancers, offering potential avenues for further research and drug development.


Assuntos
Neoplasias Pulmonares , Animais , Camundongos , Neoplasias Pulmonares/patologia , Adesões Focais/metabolismo , Adesões Focais/patologia , Proteômica , Linhagem Celular Tumoral , Movimento Celular , Transição Epitelial-Mesenquimal , Metástase Neoplásica/patologia
2.
Cancer Lett ; 578: 216458, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37865161

RESUMO

Epidermal growth factor receptor (EGFR) abnormalities relevant to tumor progression. A newly developed strategy for cancer therapy is induction of EGFR degradation. GMI, an immunomodulatory protein from the medicinal mushroom Ganoderma microsporum, exhibits anticancer activity. However, its role in the intracellular trafficking and degradation of EGFR remains unclear. In this study, we discovered that GMI inhibits the phosphorylation of multiple tyrosine kinases. Specifically, GMI was discovered to suppress lung cancer cells harboring both wild-type and mutant EGFR by inhibiting EGFR dimerization and eliminating EGFR-mediated signaling. Functional studies revealed that GMI binds to the extracellular segment of EGFR. GMI interacts with EGFR to induce phosphorylation of EGFR at tyrosine1045, which triggers clathrin-dependent endocytosis and degradation of EGFR. Furthermore, in the mouse models, GMI was discovered to suppress tumor growth. Knockdown of EGFR in lung cancer cells abolishes GMI's anticancer activity in vivo and in vitro. Our results reveal the interaction mechanisms through which GMI induces EGFR degradation and abolishes EGFR-mediated intracellular pathway. Our study indicates that GMI is an EGFR degrader for inhibiting EGFR-expressing tumor growth.


Assuntos
Ganoderma , Neoplasias Pulmonares , Animais , Camundongos , Neoplasias Pulmonares/tratamento farmacológico , Receptores ErbB , Fosforilação , Fator de Crescimento Epidérmico , Linhagem Celular Tumoral
4.
Int J Biol Macromol ; 238: 124144, 2023 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-36958446

RESUMO

Sulfated polysaccharides (Ac-SPSs) of Antrodia cinnamomea present anti-cancer activity. However, the anti-cancer mechanism of Ac-SPSs is not fully understood and remains largely unexplored. In this study, we identify an Ac-SPS with 7.9 kDa, noted ZnF3, and aim to examine the dual anti-cancer functions of ZnF3 on inhibiting cancer cells and activating macrophages. A biological study shows that ZnF3 inhibits lung cancer cells by inducing subG1 population and apoptosis. ZnF3 downregulates the expression of TGFß receptor in lung cancer cells. In parallel, ZnF3 activates macrophages via induction of TNF-α and IL-6 secretion, NO production and phagocytosis. ZnF3 activates AKT/mTOR pathway and induces M1 type macrophage polarization. Cancer cells co-cultured with ZnF3-stimulated macrophages, leading to inhibition of lung cancer cells. This study demonstrates that ZnF3 not only directly inhibits cancer cells but also activates macrophages-mediated cytotoxic effect on cancer cells. Moreover, ZnF3 may be a supplement for suppressing lung cancer cells.


Assuntos
Antrodia , Neoplasias Pulmonares , Humanos , Sulfatos/farmacologia , Polissacarídeos/farmacologia , Apoptose , Morte Celular , Neoplasias Pulmonares/tratamento farmacológico , Macrófagos
5.
Int J Biol Macromol ; 219: 940-948, 2022 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-35952817

RESUMO

Glioblastoma multiforme (GBM), which is a malignant primary brain tumor, is the cancer that spreads most aggressively into the adjacent brain tissue. Patients with metastatic GBM have a poor chance of survival. In this study, we examined the anti-GBM mobility effect of small protein, called GMI, which is cloned and purified from Ganoderma microsporum. Proteomic profiles showed that GMI-mediated proteins were involved in cell motility and cell growth functions. Specifically, we demonstrated that GMI significantly suppressed cell migration and invasion of GBM cells. GMI combined with temozolomide (TMZ), which is a traditional chemotherapeutic agent for GBM treatment, synergistically inhibited motility in GBM cells. Mechanistically, we demonstrated that GMI induced proteasome-dependent degradation of Slug, which is a critical transcription factor, is frequently linked to metastasis and drug resistance in GBM. Knockdown of Slug reduced cell viability and colony formation of GBM cells but enhanced TMZ-suppressed cell migration and viability. The results of this study show that targeting Slug degradation is involved in GMI-suppressed mobility of GBM cells. Moreover, GMI may be a potential supplementary agent for the suppression of GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular , Resistencia a Medicamentos Antineoplásicos , Ganoderma , Glioblastoma/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma , Proteômica , Fatores de Transcrição da Família Snail , Temozolomida/farmacologia , Fatores de Transcrição/genética
6.
Polymers (Basel) ; 13(24)2021 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-34960904

RESUMO

Lung cancer has the highest global mortality rate of any cancer. Although targeted therapeutic drugs are commercially available, the common drug resistance and insensitivity to cisplatin-based chemotherapy, a common clinical treatment for lung cancer, have prompted active research on alternative lung cancer therapies and methods for mitigating cisplatin-related complications. In this study, we investigated the effect of WSG, a glucose-rich, water soluble polysaccharide derived from Ganoderma lucidum, on cisplatin-based treatment for lung cancer. Murine Lewis lung carcinoma (LLC1) cells were injected into C57BL/6 mice subcutaneously and through the tail vein. The combined administration of WSG and cisplatin effectively inhibited tumor growth and the formation of metastatic nodules in the lung tissue of the mice. Moreover, WSG increased the survival rate of mice receiving cisplatin. Co-treatment with WSG and cisplatin induced a synergistic inhibitory effect on the growth of lung cancer cells, enhancing the apoptotic responses mediated by cisplatin. WSG also reduced the cytotoxic effect of cisplatin in both macrophages and normal lung fibroblasts. Our findings suggest that WSG can increase the therapeutic effectiveness of cisplatin. In clinical settings, WSG may be used as an adjuvant or supplementary agent.

7.
Int J Biol Macromol ; 193(Pt B): 1201-1208, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34742847

RESUMO

Tongue cancer, a kind of oral cancer, is common in Southeast Asian countries because of dietary habits. However, there is no specific targeted drug that could effectively inhibit oral cancer. WSG, as a water soluble glucose-enriched polysaccharide from Ganoderma lucidum, exerts excellent pharmacological efficacy of anti-lung cancer. However, its anticancer functions and mechanisms in human tongue cancer need to be further explored. Herein, we showed that WSG dramatically reduced cell viability and colony formation of tongue cancer cells. WSG increased subG1 and G2/M populations as well as induced apoptotic responses. In parallel, WSG enhanced apoptosis-related Bax/Bcl2 ratio. Mechanistic studies showed that WSG reduced phosphorylation of EGFR and AKT. In addition, we found a synergistic effect of WSG with cisplatin in inhibition of cell viability and induction of apoptosis. WSG significantly reduced the inhibition concentration 50% (IC50) of cisplatin. More importantly, WSG ameliorated cisplatin-induced cytotoxicity in normal human oral epithelial SG cells. In conclusion, our findings provided important insights into the anti-tongue cancer effects of WSG via inhibition of EGFR/AKT axis and induction of apoptosis, which indicated that WSG could be a promising supplement for tongue cancer treatment.


Assuntos
Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Polissacarídeos Fúngicos , Proteínas de Neoplasias/metabolismo , Reishi/química , Transdução de Sinais/efeitos dos fármacos , Neoplasias da Língua , Linhagem Celular Tumoral , Receptores ErbB/metabolismo , Polissacarídeos Fúngicos/química , Polissacarídeos Fúngicos/farmacologia , Humanos , Neoplasias da Língua/tratamento farmacológico , Neoplasias da Língua/metabolismo
8.
Phytomedicine ; 80: 153384, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33113507

RESUMO

BACKGROUND: Ling Zhi-8 (LZ-8) and GMI are two fungal immunomodulatory proteins (FIPs) with a similar structure and amino acid sequence and are respectively obtained from the medicinal mushroom Ganoderma lucidum and Ganoderma microsporum. They present the anti-cancer progression and metastasis. We previously demonstrated that LZ-8 reduces the tumor progression in lung cancer LLC1 cell-bearing mouse. However, it is unclear whether these FIPs induce changes in the protein expression profile in cancer cells and the mechanism for such a process is not defined. PURPOSE: This study determines the changes in the proteomic profile for tumor lesions of LLC1 cell-bearing mouse received with LZ-8 and the potential mechanism for FIPs in anti-lung cancer cells. METHODS: The proteomic profile of tumor lesions was determined using two-dimensional electrophoresis and a LTQ-OrbitrapXL mass spectrometer (LC-MS/MS). The biological processes and the signaling pathway enrichment analysis were performed using Ingenuity Pathway Analysis (IPA). The differentially expressed proteins were verified by Western blot. Cell viability was determined by MTT assay. Cell morphology was characterized using electron microscopy. Migration was detected using the Transwell assay. The apoptotic response was determined using Western blot and flow cytometry. RESULTS: Obtained results showed that 21 proteins in the tumor lesions exhibited differential (2-fold change, p < 0.05) expression between PBS and LZ-8 treatment groups. LZ-8-induced changes in the proteomic profile that may relate to protein degradation pathways. Specifically, three heat shock proteins (HSPs), HSP60, 70 and 90, were significantly downregulated in tumor lesions of LLC1-bearing mouse received with LZ-8. Both LZ-8 and GMI reduced the protein levels for these HSPs in lung cancer cells. Functional studies showed that they inhibited cell migration but effectively induced apoptotic response in LLC1 cells in vitro. In addition, the inhibitors of HSP60 and HSP70 effectively inhibited cell migration and decreased cell viability of LLC1 cells. CONCLUSIONS: LZ-8 induced changes in the proteomic profile of tumor lesions which may regulate the HSPs-related cell viability. Moreover, inhibition of HSPs may be related to the anti-lung cancer activity.


Assuntos
Proteínas Fúngicas/farmacologia , Ganoderma/química , Proteínas de Choque Térmico/metabolismo , Fatores Imunológicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida , Regulação para Baixo/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Masculino , Camundongos Endogâmicos C57BL , Proteômica/métodos , Espectrometria de Massas em Tandem , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Int J Biol Macromol ; 165(Pt A): 1604-1613, 2020 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-33011264

RESUMO

WSG is a water soluble polysaccharides isolated from Ganoderma lucidum. In this study, we showed that WSG, a glucose-rich polysaccharide with an average molecular mass of approximately 1000 kDa, effectively inhibited cell viability and mobility of lung cancer cells. Functional studies revealed that WSG reduced phosphorylation of ERK1/2 in cells upon either EGF or TGFß stimulation. WSG also inhibited phosphorylation of multiple intracellular signaling molecules such as FAK, AKT and Smad2. Mechanistically, we demonstrated that WSG induced degradation of TGFß and EGF receptors via proteasome and lysosome, respectively. Moreover, we found that WSG significantly suppressed lung tumor growth, reduced the size of metastatic nodules in the lungs and prolonged the survival of LLC1-bearing mice. Our findings suggested that WSG may have potential as a therapeutic intervention for treatment of lung cancer.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Polissacarídeos/química , Reishi/química , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Proteínas de Neoplasias/genética , Polissacarídeos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Int J Biol Macromol ; 162: 1476-1483, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32739511

RESUMO

Sterols play crucial roles in the physiological functions of organisms. In this study, we examined the chemical and biological effects of sterol type elicitors, including squalene, cholesterol and stigmasterol, on polysaccharides (PSs) of Antrodia cinnamomea. Characteristic studies revealed that squalene and stigmasterol effectively increased the glucose contents in PSs. Specifically, squalene not only induced glucose content but also increased fucose and mannose levels in PSs. However, cholesterol did not induce changes in sugar content in PSs. We further identified that high dose squalene significantly promoted 20% yield (w/w) of PSs as well as significantly increased the glucose, galactose and fucose contents. In addition, the major PSs species had a molecular weight of 21 kDa, and squalene significantly increased its area percentage to 43.54. The biological effects of PSs (squalenePS) from squalene treated A. cinnamomea presented anticancer activities by inhibiting lung cancer cell viability and colony formation. Functional studies revealed that squalenePS reduced the glucose uptake and lactate secretion may correlate to inhibition of AKT activity and downregulation of glucose transporter (GLUT1) expression. Our findings suggested squalene may play vital roles in regulating the PSs assembling and bioactivities of A. cinnamomea. Moreover, squalene may be a potential supplement for adding the culture medium of A. cinnamomea.


Assuntos
Antrodia/química , Fenômenos Químicos , Polissacarídeos Fúngicos/química , Polissacarídeos Fúngicos/farmacologia , Esteróis/química , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Cromatografia em Gel , Polissacarídeos Fúngicos/isolamento & purificação , Glucose/metabolismo , Humanos , Ácido Láctico/biossíntese , Camundongos , Estrutura Molecular
11.
Int J Biol Macromol ; 153: 796-805, 2020 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-32169452

RESUMO

Gefitinib is a first tyrosine kinase inhibitor (TKI) designed with an EGFR tyrosine kinase for lung cancer targeted therapy. However, some lung cancer patients with wild-type EGFR (wtEGFR) or acquired secondary EGFR mutation showed lower response rate of gefitinib. In this study, we examined the efficacy of fucoidan on altering gefitinib-sensitivity on TKI-resistant lung cancer A549 and H1975 cells. We found that the simultaneous administration of fucoidan and gefitinib synergistically inhibited lung cancer cell viability via activating apoptotic response. Moreover, we found that fucoidan effectively downregulated expressions of mesenchymal-like molecules. Mechanistically, we demonstrated that fucoidan altered the gefitinib-inhibitory rate may result from induction of proteasome-dependent Slug degradation. Abolishment of TGFß signaling enhanced gefitinib-inhibited cell viability and reduced N-cadherin, Twist and Slug levels. Moreover, knockdown of Slug contributed the increasing the gefitinib-sensitivity of H1975 cells. Our study is the first to find that fucoidan alters the gefitinib-sensitive of TKI-resistant cells by reduction of TGFß receptor-mediated expressions of mesenchymal-like molecules and induction of Slug degradation. Together, our current results indicate that combination of fucoidan and gefitinib may be a potential and effective therapeutic strategy in gefitinib non-sensitive lung cancer.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Gefitinibe/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares , Proteínas de Neoplasias/metabolismo , Polissacarídeos/farmacologia , Fatores de Transcrição da Família Snail/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Células A549 , Animais , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos
12.
Carbohydr Polym ; 216: 204-212, 2019 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-31047058

RESUMO

Sulfated polysaccharides (SPSs) are polysaccharides (PSs) with high sulfate functionalization and possess bioactivities. This study aimed to increase the sulfate content of SPSs in Antrodia cinnamomea through sulfate feeding. Feeding A. cinnamomea with sodium thiosulfate was found to increase yields of PSs and SPSs in A. cinnamomea. The SPSs thus obtained (ST-SPS) were further isolated, showing enhanced sulfate content of 2.5 mmol/g. Sodium thiosulfate induced changes in molecular weight from 320 kDa to 1342 kDa, and area percentage of low-molecular-weight ST-SPS (< 20 kDa) was decreased. Functional studies revealed that sodium thiosulfate increased the ST-SPS anticancer efficacy in cancer cells via inhibition of EGFR/AKT signaling. Moreover, the ST-SPS enhanced synergistically cisplatin-, gefitinib- and 5 FU-induced cytotoxic effects in lung cancer H1975 cells and colon cancer CT26 cells. This study is the first to demonstrate that sodium thiosulfate induced changes in properties of A. cinnamomea with the anticancer mechanisms of ST-SPS.


Assuntos
Antineoplásicos/farmacologia , Antrodia/química , Antrodia/metabolismo , Polissacarídeos/farmacologia , Ésteres do Ácido Sulfúrico/farmacologia , Tiossulfatos/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Fluoruracila/farmacologia , Gefitinibe/farmacologia , Humanos , Concentração Inibidora 50 , Camundongos , Peso Molecular , Fosforilação/efeitos dos fármacos , Polissacarídeos/biossíntese , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Transdução de Sinais/efeitos dos fármacos , Ésteres do Ácido Sulfúrico/química , Ésteres do Ácido Sulfúrico/isolamento & purificação , Ésteres do Ácido Sulfúrico/metabolismo
13.
Carbohydr Polym ; 210: 175-184, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-30732751

RESUMO

SGA is a sulfated glucan from Antrodia cinnamomea. In this study, we showed that SGA suppressed tumor growth in vitro and in vivo. SGA also potentiated cisplatin-induced cytotoxicity in lung cancer cells. TGFß signaling and overexpression of Slug are regarded as the critical events in lung tumor malignancy. Functional studies revealed that SGA inhibited the TGFß/FAK/AKT axis by inducing lipid-raft-mediated lysosome-dependent TGFß receptor degradation, resulting in suppressing cancer cell viability and migration. Moreover, SGA elimination of TGFß-mediated intracellular signaling promoted Slug degradation in H1975 cells. Mechanistically, we demonstrated that proteasome-dependent Slug degradation was controlled by TGFß-mediated downstream signaling pathways; however, inhibitors of AKT and GSK3 abolished Slug degradation. Our findings suggested that SGA targets of the TGFß/AKT/GSK3ß axis played a key role in enhancing Slug degradation and suppressing lung cancer cells. In addition, SGA may be a potential therapeutic supplement for lung cancer.


Assuntos
Antrodia/química , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucanos/farmacologia , Neoplasias Pulmonares/patologia , Fatores de Transcrição da Família Snail/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Masculino , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Int J Biol Macromol ; 120(Pt A): 952-958, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30144544

RESUMO

Microelements play pivotal roles for fungal/plant development and end-use properties. In this study, we examined the production and characterization of valuable sulfated polysaccharides (SPSs) with biological benefits from Antrodia cinnamomea and fine-tuning of mycelial culture conditions. Using various sulfated salts (e.g. CuSO4, FeSO4 and ZnSO4) to feed A. cinnamomea, we found that CuSO4 and ZnSO4 increased 25% and 20% of mycelium yields, respectively. We further isolated the SPSs from CuSO4, FeSO4 and ZnSO4-feeding of A. cinnamomea (called CuFSPS, FeFSPS and ZnFSPS, respectively) and found that CuSO4 and ZnSO4 significantly promoted SPS production. By contrast, FeSO4 did not change the yields of mycelium and SPS from A. cinnamomea. Characteristic studies have revealed that these sulfated salts did not significantly induce change in the sulfation and the sugar contents of SPS. However, the galactose and glucose contents in ZnFSPS were increased to the value of 249 and 1038 µmol/g, respectively. In addition, in regard to area percentages, while the major SPSs species were low-molecular-weight SPSs (<23 kDa), the sulfated salts increased the area percentages of molecular size in the range of 200-500 kDa. Anticancer function studies showed that those SPSs inhibit the cell viability 35-45% at 800 µg/ml of lung cancer A549 cells via downregulation of EGFR signaling. Our study is the first to identify the efficacy of microelements in the enhancement of mycelia yield and SPS, in which CuSO4 and ZnSO4 enhanced mycelia growth and increased the production of SPS. Our finding suggests that ZnSO4 may play roles in regulating the SPS assembling. Moreover, those SPSs derived from feeding A. cinnamomea with microelements may be useful as a potential agent for inhibition of lung cancer viability.


Assuntos
Antrodia/crescimento & desenvolvimento , Micélio/crescimento & desenvolvimento , Polissacarídeos/biossíntese , Sulfatos/farmacologia , Células A549 , Antrodia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sulfato de Cobre/farmacologia , Galactose/química , Glucose/química , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Micélio/efeitos dos fármacos , Polissacarídeos/química , Polissacarídeos/farmacologia , Sulfato de Zinco/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...