Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Anim Sci Biotechnol ; 13(1): 118, 2022 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-36224643

RESUMO

BACKGROUND: This study aimed to investigate whether the combination of Macleaya cordata extract (MCE) and Bacillus could improve the laying performance and health of laying hens better. METHODS: A total of 360 29-week-old Jingbai laying hens were randomly divided into 4 treatments: control group (basal diet), MCE group (basal diet + MCE), Probiotics Bacillus Compound (PBC) group (basal diet + compound Bacillus), MCE + PBC group (basal diet + MCE + compound Bacillus). The feeding experiment lasted for 42 d. RESULTS: The results showed that the laying rate and the average daily egg mass in the MCE + PBC group were significantly higher than those in the control group (P < 0.05) and better than the MCE and PBC group. Combination of MCE and Bacillus significantly increased the content of follicle-stimulating hormone (FSH) in the serum and up-regulated the expression of related hormone receptor gene (estrogen receptor-ß, FSHR and luteinizing hormone/choriogonadotropin receptor) in the ovary of laying hens (P < 0.05). In the MCE + PBC group, the mRNA expressions of zonula occluden-1, Occludin and mucin-2 in jejunum was increased and the intestinal epithelial barrier detected by transmission electron microscopy was enhanced compared with the control group (P < 0.05). In addition, compared with the control group, combination of MCE and Bacillus significantly increased the total antioxidant capacity and catalase activity (P < 0.05), and down-regulated the mRNA expressions of inflammation-related genes (interleukin-1ß and tumor necrosis factor-α) as well as apoptosis-related genes (Caspase 3, Caspase 8 and P53) (P < 0.05). The concentration of acetic acid and butyric acid in the cecum content of laying hens in the MCE + PBC group was significantly increased compared with the control group (P < 0.05). CONCLUSIONS: Collectively, dietary supplementation of 600 µg/kg MCE and 5 × 108 CFU/kg compound Bacillus can improve laying performance by improving microbiota to enhance antioxidant capacity and intestinal barrier, regulate reproductive hormones and the concentration of cecal short-chain fatty acids of laying hens, and the combined effect of MCE and Bacillus is better than that of single supplementation.

2.
Int J Mol Sci ; 22(22)2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34830269

RESUMO

Clostridium perfringens (C. perfringens) causes intestinal injury through overgrowth and the secretion of multiple toxins, leading to diarrhea and necrotic enteritis in animals, including pigs, chickens, and sheep. This study aimed to investigate the protective effects of Lactobacillus plantarum (L. plantarum) Lac16 on C. perfringens infection-associated injury in intestinal porcine epithelial cell line (IPEC-J2). The results showed that L. plantarum Lac16 significantly inhibited the growth of C. perfringens, which was accompanied by a decrease in pH levels. In addition, L. plantarum Lac16 significantly elevated the mRNA expression levels of host defense peptides (HDPs) in IPEC-J2 cells, decreased the adhesion of C. perfringens to IPEC-J2 cells, and attenuated C. perfringens-induced cellular cytotoxicity and intestinal barrier damage. Furthermore, L. plantarum Lac16 significantly suppressed C. perfringens-induced gene expressions of proinflammatory cytokines and pattern recognition receptors (PRRs) in IPEC-J2 cells. Moreover, L. plantarum Lac16 preincubation effectively inhibited the phosphorylation of p65 caused by C. perfringens infection. Collectively, probiotic L. plantarum Lac16 exerts protective effects against C. perfringens infection-associated injury in IPEC-J2 cells.


Assuntos
Infecções por Clostridium/metabolismo , Clostridium perfringens/crescimento & desenvolvimento , Células Epiteliais/metabolismo , Enteropatias/metabolismo , Enteropatias/veterinária , Mucosa Intestinal/metabolismo , Lactobacillus plantarum/metabolismo , Probióticos/farmacologia , Substâncias Protetoras/farmacologia , Doenças dos Suínos/metabolismo , Animais , Aderência Bacteriana , Linhagem Celular , Infecções por Clostridium/microbiologia , Clostridium perfringens/metabolismo , Técnicas de Cocultura/métodos , Células Epiteliais/microbiologia , Enteropatias/microbiologia , Mucosa Intestinal/microbiologia , Probióticos/metabolismo , Substâncias Protetoras/metabolismo , Suínos , Doenças dos Suínos/microbiologia
3.
Front Vet Sci ; 8: 679368, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34150896

RESUMO

The aim of this study was to evaluate the dietary effects of Bacillus amyloliquefaciens SC06 (SC06) instead of antibiotics on the growth performance, intestinal health, and intestinal microbiota of broilers. A total of 360 30-day-old Lingnan yellow broilers were randomly allocated into two groups with six replicates per group (30 birds per replicate). The broilers were fed either a non-supplemented diet or a diet supplemented with 108 colony-forming units lyophilized SC06 per kilogram feed for 30 days. Results showed that SC06 supplementation had no effect on the growth performance compared with that of the control group. SC06 treatment significantly (P <0.05) increased the total antioxidant capacity (T-AOC), total superoxide dismutase (T-SOD) activity in the liver, and the activities of trypsin, α-amylase (AMS), and Na+K+-ATPase in the ileum, whereas it decreased (P < 0.05) lipase, gamma glutamyl transpeptidase (γ-GT), and maltase activities in the ileum. Meanwhile, SC06 treatment also improved the immune function indicated by the significantly (P < 0.05) increased anti-inflammatory cytokine [interleukin (IL)-10] level and the decreased (P < 0.05) pro-inflammatory cytokine [IL-6 and tumor necrosis factor (TNF)-α] levels in the ileum. Furthermore, we also found that SC06 enhanced the intestinal epithelial intercellular integrity (tight junction and adhesion belt) in the ileum. Microbial analysis showed that SC06 mainly increased the alpha diversity indices in the jejunum, ileum, and cecum. SC06 treatment also significantly (P < 0.05) increased the abundances of Bacteroidetes, Bacteroidales, Bacteroides, Fusobacteria, Clostridiaceae, and Veillonellaceae in the cecum and simultaneously decreased the abundances of Planococcaceae in the duodenum, Microbacteriaceae in the jejunum, and Lachnospiraceae, [Ruminococcus] and Ruminococcus in cecum. In conclusion, these results suggested that B. amyloliquefaciens instead of antibiotics showed a potential beneficial effect on the intestinal health of broilers.

4.
Poult Sci ; 99(11): 5356-5365, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33142452

RESUMO

Macrophages are professional phagocytic cells that play a critical role in initiating immune responses by presenting antigen and phagocytic clearance. The macrophages can be targeted for immunomodulation by beneficial microbes, such as probiotics. The aim of this study is to investigate the protective effect of Saccharomyces boulardii against Clostridium perfringens infection in avian macrophage cell line HD11. In this study, HD11 macrophages were prestimulated with S. boulardii for 6 h and then infected with C. perfringens for 3 h. Results showed that S. boulardii enhanced phagocytosis and bactericidal capacity against C. perfringens by HD11 cells. The S. boulardii effectively promoted the mRNA expression of CD80, CD83, and CD197 cell-surface molecules in C. perfringens-infected HD11 cells. Moreover, we found that prestimulation with S. boulardii reduced the mRNA expression of CD40, toll-like receptor [TLR] 4, and TLR15 induced by C. perfringens and thereby downregulated the mRNA expression of myeloid differentiation primary response 88, TNF receptor associated factor 6, nuclear factor kappa-B p65 subunit, and c-Jun N-terminal kinase genes in HD11 cells. The upregulation of cytokines (interleukin [IL]-6, tumor necrosis factor alpha, and IL-10) and inducible nitric oxide synthase mRNA expression in C. perfringens-infected HD11 cells were noticeably inhibited by S. boulardii pretreatment. Conclusively, these results might provide a new insight into the role of S. boulardii in regulating avian immune defense against C. perfringens invasion and immune escape.


Assuntos
Antibiose , Infecções por Clostridium , Clostridium perfringens , Doenças das Aves Domésticas , Saccharomyces boulardii , Animais , Antibiose/imunologia , Galinhas , Infecções por Clostridium/imunologia , Infecções por Clostridium/microbiologia , Infecções por Clostridium/veterinária , Inflamação/imunologia , Macrófagos/imunologia , Macrófagos/microbiologia , Fator 88 de Diferenciação Mieloide/imunologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia , Saccharomyces boulardii/imunologia , Receptor 4 Toll-Like/imunologia , Receptores Toll-Like/imunologia
5.
Animals (Basel) ; 10(11)2020 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-33202761

RESUMO

At present, the widespread use of high-dose zinc oxide and antibiotics to prevent post-weaning diarrhea (PWD) in piglets has caused serious environmental problems. To solve this problem, we studied the effect of HNa as a substitute for zinc oxide (ZnO) and antibiotics on the growth performance, immune status, and antioxidant capacity of piglets. Seventy-two weaned piglets (body weight = 7.42 ± 0.85 kg, 26-d-old) were distributed in a randomized 2 × 3 factorial design (two sexes and three treatments) with six replicates of four piglets each. The three treatments were the control diet (basic diet), HNa diet (basic diet + 2000 mg/kg sodium humate), and ZoA group (basic diet + 1600 mg/kg zinc oxide + 1000 mg/kg oxytetracycline calcium). ANOVA and Chi-square tests were applied to compare the means (p < 0.05) between treatments. The results showed that body weight at 16 and 30 d and the average daily gain of piglets fed with HNa or ZoA were significantly higher (p < 0.05) than the control group. Supplementing HNa or ZoA significantly increased (p < 0.05) the level of immunoglobulin M and G, and reduced (p < 0.05) the concentration of inflammatory factors such as tumor necrosis factor-alpha (TNF-α), interleukins IL-6 and IL-1ß, myeloperoxidase (MPO), and diamine oxidase (DAO). Furthermore, dietary HNa or ZnO significantly reduced (p < 0.05) the level of total antioxidant capacity (T-AOC) and malondialdehyde (MDA) compared with the control group. ZoA treatment showed an upward trend of IgA level and a downward trend of the concentration of lipopolysaccharide (LPS) and catalase (CAT). Overall, the study demonstrated that the addition of HNa in the diet partially replaced antibiotics and ZnO to improve the growth performance, immune function, and antioxidant capacity of weaned piglets, and maintained a good preventive effect on piglet diarrhea.

6.
Anim Sci J ; 91(1): e13439, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32779289

RESUMO

Lactobacillus rhamnosus GG (LGG) is increasingly applied in functional food products and acts as a probiotic model in nutritious and clinical studies. Increasing evidences have revealed the immune modulation of LGG on macrophages. The aim of this study is to investigate the effect of LGG on macrophage polarization of murine bone marrow-derived macrophages (BMDMs). BMDMs were treated with 108 colony-forming units (CFU)/ml LGG for 1.5, 3, and 6 hr. Results showed that LGG obviously upregulated the mRNA expression of M1-associated cytokines (p < .05), including interleukin-1 beta (IL-1ß), IL-6, tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS), whereas had no effect on the expression of M2-associated markers (p > .05), including arginase 1 (Arg1), mannose receptor, and chitinase-like protein 3 (YM1). Furthermore, LGG markedly increased the expression of pro-inflammatory cytokines (IL-12p40, cyclooxygenase-2 [COX-2], and interferon-γ [IFN-γ]) (p < .05) and anti-inflammatory cytokines (IL-10, IL-4, and transforming growth factor-ß [TGF-ß]) (p < .05). In addition, we also found that TLR2/MyD88/MAPK signaling pathway was required for LGG-induced M1 macrophage polarization and M1-related cytokines expression. Together, these findings demonstrate that probiotic LGG facilitates M1 polarization of BMDMs, suggesting that LGG may have an immunotherapeutic potential in regulating the host defense against pathogen invasion.


Assuntos
Células da Medula Óssea , Lacticaseibacillus rhamnosus/química , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Receptor 2 Toll-Like/imunologia , Animais , Camundongos
7.
Food Funct ; 10(12): 7653-7666, 2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31742290

RESUMO

Bacillus amyloliquefaciens SC06 (BaSC06), a potential probiotic, plays a positive role in animal growth performance and immune function. The aim of the present study was to investigate the protective effect of BaSC06 against Salmonella infection and its association with macrophage polarization. C57BL/6 mice were fed with or without a BaSC06-containing diet before Salmonella enterica Typhimurium (ST) challenge. Results showed that BaSC06 had a protective effect against ST inoculation and induced both M1 and M2 macrophage polarization in the cecum. An in vitro co-culture model demonstrated that BaSC06 promoted M1 polarization directly, and thus increased the phagocytosis and bactericidal activity against ST. In addition, adoptive transfer of bone marrow-derived macrophages (BMDMs) stimulated by BaSC06 significantly decreased the counts of ST in the spleen. Furthermore, 16S rRNA-based analysis of cecal content showed that BaSC06 significantly increased the proportion of Verrucomicrobia and decreased Bacterodetes. Transplantation of the fecal microbiota from BaSC06-treated animals promoted M2 macrophage polarization in the cecum and significantly relieved inflammation caused by ST. In conclusion, BaSC06 polarized macrophages to the M1 type directly resulting in excellent bactericidal activity. Meanwhile, the microbiota modified by BaSC06 can induce M2 polarization which ameliorates the inflammation caused by ST.


Assuntos
Bacillus amyloliquefaciens/fisiologia , Macrófagos/imunologia , Microbiota , Probióticos/administração & dosagem , Infecções por Salmonella/prevenção & controle , Salmonella typhimurium/efeitos dos fármacos , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Polaridade Celular/efeitos dos fármacos , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Microbiota/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/fisiologia
8.
Poult Sci ; 98(8): 3138-3149, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30938807

RESUMO

The aim of this study was to investigate the effect of protocatechuic acid (PCA) on the growth performance, meat quality, and intestinal health of Chinese yellow-feathered broilers. Growing broilers were fed the basal diet or diets supplemented with 300 or 600 mg/kg PCA, or 200 mg/kg enramycin for 52 D. We found that addition of 300 mg/kg PCA significantly increased body weight, live weight, and carcass weight and decreased the feed to gain ratio of broilers; PCA improved meat quality through reducing shear force, and increasing a* (relative redness) and decreasing b* (relative yellowness) at 24 h after slaughter. The activities of alkaline phosphatase and diamine oxidase in plasma were significantly decreased by administration of 300 mg/kg PCA; PCA also significantly increased total antioxidant capability and decreased malondialdehyde content and activity of xanthine oxidase in liver. Meanwhile, it enhanced activities of total superoxide dismutase, glutathione s-transferase, and glutathione peroxidase in the jejunal mucosa. Interleukin-10 and transforming growth factor-ß were significantly increased in jejunal mucosa and plasma of 300 mg/kg PCA diet group, whereas interluekin-2 and interferon-γ dropped dramatically. Moreover, relative expression of apoptosis-related genes decreased in liver, whereas that of intestinal barrier-related and immunity-related genes increased in jejunum. Furthermore, 300 mg/kg PCA treatment significantly changed α-diversity and structure of the cecal microflora in broilers, with increasing relative abundance of Firmicutes and Actinobacteria while reducing Bacteroidetes and Proteobacteria. These results indicated that PCA improved the feed efficiency, growth performance, meat quality of broilers, and antioxidant capacity. It also enhanced intestinal immune function and improved the structure of intestinal flora to favor improved intestinal health in Chinese yellow-feathered broilers.


Assuntos
Ração Animal/análise , Galinhas/crescimento & desenvolvimento , Hidroxibenzoatos/farmacologia , Animais , Peso Corporal , Cor , Dieta/veterinária , Microbioma Gastrointestinal/efeitos dos fármacos , Expressão Gênica , Intestinos/efeitos dos fármacos , Intestinos/fisiologia , Carne/análise , Peptídeos/farmacologia , Resistência ao Cisalhamento/efeitos dos fármacos
9.
J Cell Physiol ; 234(11): 19406-19419, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31020664

RESUMO

The intestinal mucosal barrier is the first line to defense against luminal content penetration and performs numerous biological functions. The intestinal epithelium contains a huge surface that is lined by a monolayer of intestinal epithelial cells (IECs). IECs are dominant mediators in maintaining intestinal homeostasis that drive diverse functions including nutrient absorption, physical segregation, secretion of antibacterial peptides, and modulation of immune responses. Autophagy is a cellular self-protection mechanism in response to various stresses, and accumulating studies have revealed its importance in participating physiological processes of IECs. The regulatory effects of autophagy depend on the specific IEC types. This review aims to elucidate the myriad roles of autophagy in regulating the functions of different IECs (stem cells, enterocytes, goblet cells, and Paneth cells), and present the progress of autophagy-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide new preventive and therapeutic strategies for gastrointestinal dysfunction and diseases.


Assuntos
Autofagia/genética , Células Epiteliais/metabolismo , Gastroenteropatias/genética , Mucosa Intestinal/metabolismo , Peptídeos Catiônicos Antimicrobianos , Colite/genética , Colite/microbiologia , Colite/patologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Gastroenteropatias/metabolismo , Gastroenteropatias/microbiologia , Células Caliciformes/metabolismo , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Intestinos/microbiologia , Intestinos/patologia , Celulas de Paneth/metabolismo , Celulas de Paneth/microbiologia , Celulas de Paneth/patologia
10.
Front Immunol ; 9: 2321, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30459751

RESUMO

Glycyrrhizin (GL), a triterpenoid glycoside, serves important functions in various biological activities, including antiviral and antitumor immune responses. However, the anti-inflammatory effects of GL on Salmonella enterica serovar Typhimurium (ST)-induced injury in mice and the mechanisms underlying the protection of GL are poorly understood. Here, we investigated the effects of GL on host immune responses against ST infection in mice. A phenotypic analysis using hematoxylin and eosin (H&E) staining and transmission electron microscopy showed that GL relieved ST-induced weight loss and intestinal mucosal injury. A colonization assay showed that GL significantly reduced ST colonization in the ileum and colon and translocation to the liver and spleen. An antibacterial activity assay and real-time PCR revealed that GL had no direct inhibitory impact on ST growth or virulence gene expression. ELISA showed that GL pretreatment significantly decreased proinflammatory cytokine (IFN-γ, TNF-α, IL-6) secretion and increased anti-inflammatory cytokine (IL-10) secretion in the ileum, colon and serum of ST-infected mice. Moreover, flora analysis showed that GL reduced Akkermansia, Sutterella, Prevotella and Coprococcus but enriched Parabacteroides and Anaerotruncus in the cecum of ST-infected mice. These results suggest that GL promotes the secretion of immune factors and modulates intestinal flora to prevent further ST infection. We also analyzed the effect of GL on immunocytes and found that GL promoted the phenotypic and functional maturation of murine bone marrow-derived dendritic cells (BMDCs). Flow cytometry and western blotting demonstrated that NF-κB, ERK, and p38 MAPK were required for GL-induced BMDC maturation. The above findings indicate that GL attenuates ST infection by modulating immune function and intestinal flora. This study enriches our current knowledge of GL-mediated immunological function and provides a new perspective on the prevention of Salmonella infection in animals and humans.


Assuntos
Ácido Glicirrízico/farmacologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Infecções por Salmonella/imunologia , Infecções por Salmonella/microbiologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/fisiologia , Animais , Apresentação de Antígeno , Citocinas/biossíntese , Citocinas/sangue , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal , Regulação Bacteriana da Expressão Gênica , Mediadores da Inflamação/sangue , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Infecções por Salmonella/metabolismo , Infecções por Salmonella/patologia , Salmonella typhimurium/patogenicidade , Transdução de Sinais , Virulência/genética
11.
J Zhejiang Univ Sci B ; 19(10): 785-795, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30269446

RESUMO

OBJECTIVE: Salmonella enterica remains a major cause of food-borne disease in humans, and Salmonella Typhimurium (ST) contamination of poultry products is a worldwide problem. Since macrophages play an essential role in controlling Salmonella infection, the aim of this study was to evaluate the effect of glycyrrhizic acid (GA) on immune function of chicken HD11 macrophages. METHODS: Chicken HD11 macrophages were treated with GA (0, 12.5, 25, 50, 100, 200, 400, or 800 µg/ml) and lipopolysaccharide (LPS, 500 ng/ml) for 3, 6, 12, 24, or 48 h. Evaluated responses included phagocytosis, bacteria-killing, gene expression of cell surface molecules (cluster of differentiation 40 (CD40), CD80, CD83, and CD197) and antimicrobial effectors (inducible nitric oxide synthase (iNOS), NADPH oxidase-1 (NOX-1), interferon-γ (IFN-γ), LPS-induced tumor necrosis factor (TNF)-α factor (LITAF), interleukin-6 (IL-6), and IL-10), and production of nitric oxide (NO) and hydrogen peroxide (H2O2). RESULTS: GA increased the internalization of both fluorescein isothiocyanate (FITC)-dextran and ST by HD11 cells and markedly decreased the intracellular survival of ST. We found that the messenger RNA (mRNA) expression of cell surface molecules (CD40, CD80, CD83, and CD197) and cytokines (IFN-γ, IL-6, and IL-10) of HD11 cells was up-regulated following GA exposure. The expression of iNOS and NOX-1 was induced by GA and thereby the productions of NO and H2O2 in HD11 cells were enhanced. Notably, it was verified that nuclear factor-κB (NF-κB) and c-Jun N-terminal kinase (JNK) pathways were responsible for GA-induced synthesis of NO and IFN-γ gene expression. CONCLUSIONS: Taken together, these results suggested that GA exhibits a potent immune regulatory effect to activate chicken macrophages and enhances Salmonella-killing capacity.


Assuntos
Ácido Glicirrízico/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Salmonella/efeitos dos fármacos , Animais , Células Cultivadas , Galinhas , NF-kappa B/fisiologia , Fagocitose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
12.
J Exp Clin Cancer Res ; 37(1): 151, 2018 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-30005681

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the most prevalent malignancies in the world and developed drug resistance has represented one of the most challenging tasks for management. The current therapeutic regimens may select and enrich cancer stem-like cells (CSCs) resulting in the increased resistance against treatment, metastatic potential and mortality. Regorafenib is a multi-kinase inhibitor, an FDA-approved last-of-line treatment for patients with chemo-refractory metastatic CRC. However, regorafenib's potential effects on CSCs have not been fully elucidated. METHODS: Here, we developed two 5-FU resistant CRC cell lines, HCT-116R and DLD-1R and showed the increased CSCs characteristics such as increased side-population cells, tumor sphere formation and expression of stemness markers. These cell lines and CSCs properties were used for evaluating the potential of regorafenib in suppressing CSCs. RESULTS: We showed that regorafenib treatment decreased the stemness phenotypes including tumor sphere formation, and side-population, of both HCT-116R and DLD-1R cells. Additionally, regorafenib suppressed the cell viability in both cell lines synergistically with 5-FU. In vivo, the combination of regorafenib and 5-FU significantly suppressed the tumorigenesis and stemness markers of 5-FU resistant DLD-1R. Mechanistically, regorafenib-mediated effects were associated with the induction of tumor suppressor miR-34a and suppression of WNT/ß-catenin signaling. Our findings demonstrated that regorafenib treatment was associated with the increased level of miR-34a, resulting in reversing drug resistance and cancer-initiating cell phenotypes by degrading WNT/ß-catenin in CRC. CONCLUSION: Regorafenib might be a potential drug for colon cancer stem-like cells and it should be investigated in future clinical trials.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Compostos de Fenilureia/uso terapêutico , Piridinas/uso terapêutico , Carcinogênese , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Compostos de Fenilureia/farmacologia , Piridinas/farmacologia , Transdução de Sinais
13.
Mol Ther ; 26(5): 1385-1393, 2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29567310

RESUMO

Development of biocompatible nanomaterials with multiple functionalities for combination of radiotherapy and chemotherapy has attracted tremendous attention in cancer treatment. Herein, poly(ethylene glycol) (PEG) modified polydopamine (PDA) nanoparticles were successfully developed as a favorable biocompatible nanoplatform for co-loading antitumor drugs and radionuclides to achieve imaging-guided combined radio-chemotherapy. It is demonstrated that PEGylated PDA nanoparticles can effectively load two different drugs including sanguinarine (SAN) and metformin (MET), as well as radionuclides 131I in one system. The loaded SAN and MET could inhibit tumor growth via inducing cell apoptosis and relieving tumor hypoxia, while labeling PDA-PEG with 131I enables in vivo radionuclide imaging and radioisotope therapy. As revealed by the therapeutic efficacy both in cell and animal levels, the multifunctional PDA nanoparticles (131I-PDA-PEG-SAN-MET) can effectively repress the growth of cancer cells in a synergistic manner without significant toxic side effects, exhibiting superior treatment outcome than the respective monotherapy. Therefore, this study provides a promising polymer-based platform to realize imaging-guided radioisotope/chemotherapy combination cancer treatment in future clinical application.


Assuntos
Quimiorradioterapia , Indóis , Radioisótopos do Iodo , Nanopartículas , Polímeros , Cintilografia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Modelos Animais de Doenças , Humanos , Indóis/administração & dosagem , Marcação por Isótopo , Camundongos , Nanopartículas/química , Nanopartículas/ultraestrutura , Neoplasias/patologia , Neoplasias/terapia , Polietilenoglicóis , Polímeros/administração & dosagem , Cintilografia/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Sci Rep ; 8(1): 1621, 2018 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-29374219

RESUMO

Trichostatin A (TSA) possess histone deacetylase (HDAC) inhibitory potential, can reverse the deactivation of tumor suppressor genes and inhibit tumor cell proliferation. We evaluated the effect of TSA on HDAC expression, tumor cell proliferation, and cancer stem cells (CSCs) activities in pancreatic ductal adenocarnoma (PDAC) cells. The PDAC cell lines MiaPaCa-2 and PANC-1 were distinctly sensitive to TSA, with enhanced apoptosis, compared to SAHA. TSA or SAHA inhibited vimentin, HDACs 1, 7 and 8, upregulated E-cadherin mRNA and protein levels in the PDAC cells, and time-dependently downregulated Oct-4, Sox-2, and Nanog, as well as inhibited PDAC tumorsphere formation. TSA also induces accumulation of acetylated histones, while increasing histone 3 lysine 4 or 9 dimethylation levels in PDAC cells and enhancing the epigenetic activity of SAHA. The anti-CSCs effect of TSA was like that obtained by silencing HDAC-1 or 7 using siRNA, and enhances Gemcitabine activity. Our study highlights the molecular targetability of HDACs 1, 7, and 8, confirm their PDAC-CSCs maintaining role, and demonstrate that compared to SAHA, TSA modulates the epigenetically- mediated oncogenic activity of PDAC-CSCs, and potentiate Gemcitabine therapeutic activity, making a case for further exploration of TSA activity alone or in combination with Gemcitabine in PDAC therapy.


Assuntos
Antimetabólitos Antineoplásicos/metabolismo , Desoxicitidina/análogos & derivados , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Ácidos Hidroxâmicos/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias Pancreáticas/patologia , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/metabolismo , Humanos , Células-Tronco Neoplásicas/fisiologia , Gencitabina
15.
Probiotics Antimicrob Proteins ; 9(3): 292-299, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28083809

RESUMO

Previous study showed that dietary Bacillus licheniformis (B. licheniformis) administration contributes to the improvement of laying performance and egg quality in laying hens. In this study, we aimed to further evaluate its underlying mechanisms. Three hundred sixty Hy-Line Variety W-36 hens (28 weeks of age) were randomized into four groups, each group with six replications (n = 15). The control group received the basal diet and the treatment groups received the same basal diets supplemented with 0.01, 0.03, and 0.06% B. licheniformis powder (2 × 1010 cfu/g) for an 8-week trial. The results demonstrate that B. licheniformis significantly enhance the intestinal barrier functions via decreasing gut permeability, promoting mucin-2 transcription, and regulating inflammatory cytokines. The systemic immunity of layers in B. licheniformis treatment groups is improved through modulating the specific and non-specific immunity. In addition, gene expressions of hormone receptors, including estrogen receptor α, estrogen receptor ß, and follicle-stimulating hormone receptor, are also regulated by B. licheniformis. Meanwhile, compared with the control, B. licheniformis significantly increase gonadotropin-releasing hormone level, but markedly reduce ghrelin and inhibin secretions. Overall, our data suggest that dietary inclusion of B. licheniformis can improve the intestinal barrier function and systemic immunity and regulate reproductive hormone secretions, which contribute to better laying performance and egg quality of hens.


Assuntos
Bacillus licheniformis/fisiologia , Microbioma Gastrointestinal , Hormônio Liberador de Gonadotropina/metabolismo , Imunidade , Ração Animal/análise , Ração Animal/microbiologia , Animais , Galinhas , Citocinas/metabolismo , Dieta/veterinária , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Mucina-2/metabolismo , Receptores do FSH/metabolismo
16.
Mediators Inflamm ; 2015: 372931, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26664149

RESUMO

The roots and rhizomes of Glycyrrhiza species (licorice) have been widely used as natural sweeteners and herbal medicines. The aim of this study is to investigate the effect of glycyrrhizic acid (GA) from licorice on macrophage polarization. Both phenotypic and functional activities of murine bone marrow-derived macrophages (BMDMs) treated by GA were assessed. Our results showed that GA obviously increased the cell surface expression of CD80, CD86, and MHCII molecules. Meanwhile, GA upregulated the expression of CCR7 and the production of TNF-α, IL-12, IL-6, and NO (the markers of classically activated (M1) macrophages), whereas it downregulated the expression of MR, Ym1, and Arg1 (the markers of alternatively activated (M2) macrophage). The functional tests showed that GA dramatically enhanced the uptake of FITC-dextran and E. coli K88 by BMDMs and decreased the intracellular survival of E. coli K88 and S. typhimurium. Moreover, we demonstrated that JNK and NF-κB activation are required for GA-induced NO and M1-related cytokines production, while ERK1/2 pathway exhibits a regulatory effect via induction of IL-10. Together, these findings indicated that GA promoted polarization of M1 macrophages and enhanced its phagocytosis and bactericidal capacity. The results expanded our knowledge about the role of GA in macrophage polarization.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Ácido Glicirrízico/farmacologia , MAP Quinase Quinase 4/fisiologia , Macrófagos/efeitos dos fármacos , NF-kappa B/fisiologia , Animais , Células da Medula Óssea/fisiologia , Ativação Enzimática , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Fagocitose/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA