Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 4 de 4
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Chin J Integr Med ; 30(4): 311-321, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-37594703

RÉSUMÉ

OBJECTIVE: To investigate the in vivo immunomodulatory and anti-tumor mechanisms of the combined treatment of novel Four-Herb formula (4HF) and doxorubicin in triple-negative breast cancer (TNBC). METHODS: Murine-derived triple-negative mammary carcinoma cell line, 4T1 cells, was cultured and inoculated into mouse mammary glands. Sixty-six mice were randomly assigned into 6 groups (n=11 in ench): naïve, control, LD 4HF (low dose 4HF), HD 4HF (high dose 4HF), LD 4HF + D (low dose and doxorubicin), and D (doxorubicin). Apart from the naïve group, each mouse received subcutaneous inoculation with 5 × 105 4T1 cells resuspended in 100 µL of normal saline in the mammary fat pads. Starting from the day of tumor cell inoculation, tumors were grown for 6 days. The LD and HD groups received daily oral gavage of 658 and 2,630 mg/kg 4HF, respectively. The LD 4HF+D group received daily oral gavage of 658 mg/kg 4HF and weekly intraperitoneal injection of doxorubicin (5 mg/kg). The D group received weekly intraperitoneal injections of doxorubicin (5 mg/kg). The treatment naïve mice received daily oral gavage of 0.2 mL double distilled water and 0.1 mL normal saline via intraperitoneal injection once a week. The control group received daily oral gavage of 0.2 mL double-distilled water. The treatment period was 30 days. At the end of treatment, mice organs were harvested to analyze immunological activities via immunophenotyping, gene and multiplex analysis, histological staining, and gut microbiota analysis. RESULTS: Mice treated with the combination of 4HF and doxorubicin resulted in significantly reduced tumor and spleen burdens (P<0.05), altered the hypoxia and overall immune lymphocyte landscape, and manipulated gut microbiota to favor the anti-tumor immunological activities. Moreover, immunosuppressive genes, cytokines, and chemokines such as C-C motif chemokine 2 and interleukin-10 of tumors were significantly downregulated (P<0.05). 4HF-doxorubicin combination treatment demonstrated synergetic activities and was most effective in activating the anti-tumor immune response (P<0.05). CONCLUSION: The above results provide evidence for evaluating the immune regulating mechanisms of 4HF in breast cancer and support its clinical significance in its potential as an adjunctive therapeutic agent or immune supplement.


Sujet(s)
Tumeurs , Solution physiologique salée , Animaux , Souris , Doxorubicine/pharmacologie , Doxorubicine/usage thérapeutique , Association thérapeutique , Immunité , Eau , Souris de lignée BALB C , Lignée cellulaire tumorale , Tumeurs/traitement médicamenteux
2.
J Leukoc Biol ; 115(1): 177-189, 2024 01 05.
Article de Anglais | MEDLINE | ID: mdl-37713617

RÉSUMÉ

The global pandemic of COVID-19, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been developing all over the world for more than 3 years. In late 2020, several variants of concern of SARS-CoV-2 virus emerged, with increased viral fitness and transmissibility by mutations of the spike proteins of the viral particle, denting hopes of the use of early-generation vaccines for a widespread protective immunity against viral infection. The use of adjuvants may enhance the immune responses of the conventional application of the COVID-19 vaccine. We have shown that the water extract of 2 ß-glucan-enriched immunostimulating natural products, Astragalus membranaceus (Fisch.) Bge. (AM) and Coriolus versicolor (CV), could induce innate immunity-related cytokines from human monocytes (CCL5, interleukin [IL]-6, IL-10, and tumor necrosis factor α) and monocyte-derived dendritic cells (IL-1ß, IL-10, IL-12, and tumor necrosis factor α). Using BALB/c mice, orally administrated AM and CV (1,384 and 742 mg/kg/d) for 4 d after vaccination, respectively, could enhance (1) the immunoglobulin G binding activities of BNT162b2 vaccination against ancestral and Delta SARS-CoV-2 spike proteins by 5.8- and 4.3-fold, respectively; (2) the immunoglobulin G3 subclass production of BNT162b2 vaccination against ancestral and variant SARS-CoV-2 spike proteins; and (3) the in vitro antibody-neutralizing activities of BNT162b2 vaccinated mice. In conclusion, combining AM and CV was effective in acting as an oral adjuvant with the messenger RNA vaccine BNT162b2 to improve the antigen binding activities against SARS-CoV-2 ancestral and variant SARS-CoV-2 spike proteins, probably via trained immunity of macrophages and dendritic cells.


Sujet(s)
Produits biologiques , COVID-19 , Humains , Animaux , Souris , Vaccin BNT162 , COVID-19/prévention et contrôle , Astragalus membranaceus , Interleukine-10 , Glycoprotéine de spicule des coronavirus , Vaccins contre la COVID-19 , Facteur de nécrose tumorale alpha , SARS-CoV-2 , Adjuvants immunologiques/pharmacologie , Vaccination , Anticorps neutralisants , Anticorps antiviraux
3.
Molecules ; 28(13)2023 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-37446658

RÉSUMÉ

Vaccination is the most effective method of combating COVID-19 infection, but people with a psychological fear of needles and side effects are hesitant to receive the current vaccination, and alternative delivery methods may help. Bacillus subtilis, a harmless intestinal commensal, has recently earned a strong reputation as a vaccine production host and delivery vector, with advantages such as low cost, safety for human consumption, and straightforward oral administration. In this study, we have succeeded generating "S spores" by engineering B. subtilis with spore coat proteins resembling the spike (S) protein of the ancestral SARS-CoV-2 coronavirus. With the addition of two immunostimulating natural products as adjuvants, namely Astragalus membranaceus (Fisch.) Bge (AM) and Coriolus versicolor (CV), oral administration of S spores could elicit mild immune responses against COVID-19 infection without toxicity. Mucosal IgA against the S protein was enhanced by co-feeding with AM and CV in an S spores-inoculated mouse model. Faster and stronger IgG responses against the S protein were observed when the mice were fed with S spores prior to vaccination with the commercial COVID-19 vaccine CoronaVac. In vitro studies demonstrated that AM, CV, and B. subtilis spores could dose-dependently activate both macrophages and dendritic cells by secreting innate immunity-related IL-1ß, IL-6, and TNF-α, and some other proinflammatory chemokines and cytokines. In conclusion, the combination of S spores with AM and CV may be helpful in developing a vaccine-like supplement against respiratory infection.


Sujet(s)
Produits biologiques , COVID-19 , Vaccins , Humains , Souris , Animaux , Vaccins contre la COVID-19 , Glycoprotéine de spicule des coronavirus/génétique , Glycoprotéine de spicule des coronavirus/métabolisme , Bacillus subtilis/génétique , Bacillus subtilis/métabolisme , Produits biologiques/métabolisme , Spores bactériens/métabolisme , COVID-19/prévention et contrôle , COVID-19/métabolisme , SARS-CoV-2 , Immunité innée
4.
Molecules ; 27(1)2021 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-35011470

RÉSUMÉ

Allergic rhinitis (AR) is a highly prevalent allergic disease induced by immunoglobulin (Ig) E-mediated hypersensitivity reaction at the nasal epithelium against inhaled allergens. Previous studies have demonstrated that Pentaherbs formula (PHF), a modified herbal formula comprising five herbal medicines (Flos Lonicerae, Herba Menthae, Cortex Phellodendri, Cortex Moutan and Rhizoma Atractylodis), could suppress various immune effector cells to exert anti-inflammatory and anti-allergic effects in allergic asthma and atopic dermatitis. The present study aimed to further determine the anti-inflammatory activities of PHF in an ovalbumin (OVA)-induced AR BALB/c mouse model. Nasal symptoms such as sneezing and nose rubbing were recorded and the serum total IgE and OVA-specific IgG1, as well as interleukin (IL)-4, IL-5, IL-10, IL-13, chemokines CXCL9 CXCL10, and tumor necrosis factor (TNF)-α concentrations in nasal lavage fluid (NALF) were measured during different treatments. Effects of PHF on the expression of inflammatory mediators in the sinonasal mucosa were quantified using real-time QPCR. PHF was found to suppress allergic symptoms, infiltration of inflammatory cells, and hyperplasia of goblet cells in the nasal epithelium of the OVA-induced AR mice. PHF could reduce OVA-specific IgG1 level in serum, and TNF-α and IL-10 in nasal lavage fluid (NALF), significantly up-regulate the splenic regulatory T (Treg) cell level, increase the Type 1 helper T cell (Th1)/Type 2 helper T cell (Th2) ratio, and reduce the Th17 cells (all p < 0.05). PHF could also alleviate in situ inflammation in sinonasal mucosa of OVA-induced AR mice. In conclusion, oral treatment of PHF showed immuno-modulatory activities in the OVA-induced AR mice by regulating the splenic T cell population to suppress the nasal allergy symptoms and modulating inflammatory mediators, implicating that PHF could be a therapeutic strategy for allergic rhinitis.


Sujet(s)
Anti-inflammatoires/pharmacologie , Facteurs immunologiques/pharmacologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Extraits de plantes/pharmacologie , Plantes médicinales/composition chimique , Sous-populations de lymphocytes T/effets des médicaments et des substances chimiques , Sous-populations de lymphocytes T/immunologie , Allergènes/immunologie , Animaux , Anti-inflammatoires/composition chimique , Cytokines/métabolisme , Modèles animaux de maladie humaine , Science des plantes médicinales , Facteurs immunologiques/composition chimique , Immunomodulation/effets des médicaments et des substances chimiques , Souris , Liquide de lavage nasal/immunologie , Muqueuse nasale/effets des médicaments et des substances chimiques , Muqueuse nasale/immunologie , Muqueuse nasale/métabolisme , Ovalbumine/administration et posologie , Extraits de plantes/composition chimique , Rhinite allergique/traitement médicamenteux , Rhinite allergique/étiologie , Rhinite allergique/anatomopathologie , Rate/effets des médicaments et des substances chimiques , Rate/immunologie , Rate/métabolisme , Sous-populations de lymphocytes T/métabolisme , Lymphocytes T régulateurs/effets des médicaments et des substances chimiques , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Lymphocytes auxiliaires Th2/effets des médicaments et des substances chimiques , Lymphocytes auxiliaires Th2/immunologie , Lymphocytes auxiliaires Th2/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...