Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Agric Food Chem ; 72(13): 7089-7099, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38512774

ABSTRACT

Breast cancer patients undergoing chemotherapy often experience muscle wasting and weakness, which impact their quality of life. A potential solution lies in customizing amino acid compositions based on exome-derived formulations (ExAAs). The study hypothesized that tailoring dietary amino acids using ExAAs could enhance muscle health. Theoretical amino acid requirements were calculated from the genome's exome region, and a breast cancer mouse model undergoing paclitaxel treatment was established. The mice were supplemented with a cancer-specific nutritional formula (QJS), and the effects of QJS and amino acid-adjusted QJS (adjQJS) were compared. Both formulations improved the nutritional status without compromising tumor growth. Notably, adjQJS significantly enhanced muscle strength compared to QJS (1.51 ± 0.25 vs. 1.30 ± 0.08 fold change, p < 0.05). Transcriptome analysis revealed alterations in complement and coagulation cascades, with an observed upregulation of C3 gene expression in adjQJS. Immune regulation also changed, showing a decrease in B cells and an increase in monocytes in skeletal muscle with adjQJS. Importantly, adjQJS resulted in a notable increase in Alistipes abundance compared to QJS (10.19 ± 0.04% vs. 5.03 ± 1.75%). This study highlights the potential of ExAAs as valuable guide for optimizing amino acid composition in diets for breast cancer patients undergoing chemotherapy.


Subject(s)
Breast Neoplasms , Exome , Humans , Animals , Mice , Female , Exome/genetics , Quality of Life , Amino Acids/metabolism , Diet , Muscle Strength , Muscle, Skeletal/metabolism , Dietary Supplements , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism
2.
Cell Metab ; 35(4): 620-632.e5, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36812915

ABSTRACT

How exercise elicits systemic metabolic benefits in both muscles and non-contractile tissues is unclear. Autophagy is a stress-induced lysosomal degradation pathway that mediates protein and organelle turnover and metabolic adaptation. Exercise activates autophagy in not only contracting muscles but also non-contractile tissues including the liver. However, the role and mechanism of exercise-activated autophagy in non-contractile tissues remain mysterious. Here, we show that hepatic autophagy activation is essential for exercise-induced metabolic benefits. Plasma or serum from exercised mice is sufficient to activate autophagy in cells. By proteomic studies, we identify fibronectin (FN1), which was previously considered as an extracellular matrix protein, as an exercise-induced, muscle-secreted, autophagy-inducing circulating factor. Muscle-secreted FN1 mediates exercise-induced hepatic autophagy and systemic insulin sensitization via the hepatic receptor α5ß1 integrin and the downstream IKKα/ß-JNK1-BECN1 pathway. Thus, we demonstrate that hepatic autophagy activation drives exercise-induced metabolic benefits against diabetes via muscle-secreted soluble FN1 and hepatic α5ß1 integrin signaling.


Subject(s)
Fibronectins , Proteomics , Mice , Animals , Fibronectins/metabolism , Liver/metabolism , Autophagy , Integrins
3.
Front Pharmacol ; 13: 1035101, 2022.
Article in English | MEDLINE | ID: mdl-36313318

ABSTRACT

Background: Gouty arthritis (GA) is a common inflammatory disease that causes pain due to the deposition of monosodium urate (MSU) crystals into joints and surrounding tissues. Anti-inflammatory drugs have significant clinical anti-inflammatory and analgesic effects, but they have many side effects. Cordyceps militaris is an edible and medicinal fungus, and its extract (CME) has good anti-inflammatory and analgesic effects. This study aimed to investigate the anti-inflammatory effect of CME on GA and its underlying mechanism. Methods: The effect of CME on the expression of related inflammatory factors and histopathological changes in the MSU-induced acute inflammatory gout model in rats was studied by ELISA and HE, and its anti-inflammatory mechanism was analyzed by transcriptome combined with RT-qPCR. Results: CME significantly improved gait scores and joint swelling in GA rats, and reduced MSU-induced inflammatory cell infiltration. CME inhibited MSU-induced inflammatory responses by reducing the levels of pro-inflammatory factors TNF-α, IL-1ß, IL-6, and Caspase-1 and increasing the anti-inflammatory factor IL-10. Transcriptome analysis showed that CME significantly altered inflammation-related cytokine pathways, and identified four major genes involved in regulation of inflammation, CCL7, CSF2RB, LIF, and IL-1ß. In addition, RT-qPCR was performed to verify these differential genes. Conclusion: CME significantly alleviated the inflammatory progression of GA and ameliorated the onset of GA. The underlying mechanism may be related to triggering the cytokine-cytokine receptor interaction signaling pathway to inhibit the activation of the inflammasome and regulate the immune system. And it regulates the inflammatory response induced by MSU crystals through the genes CCL7, CSF2RB, and IL-1ß.

4.
Aging (Albany NY) ; 14(13): 5376-5389, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35696640

ABSTRACT

The mushroom Ganoderma lucidum is a traditional Chinese medicine and G. lucidum spore oil (GLSO) is the lipid fraction isolated from Ganoderma spores. We examined the effect of GLSO on burn wound healing in mice. Following wounding, GLSO was applied on the wounds twice daily. Repair analysis was performed by Sirius-Red-staining at different time points. Cell proliferation and migration assays were performed to verify the effect of GLSO on growth. Network pharmacology analysis to identify possible targets was also carried out, followed by Western blotting, nuclear translocation, cell proliferation, and immunofluorescence assays for in-depth investigation of the mechanism. Our study showed that GLSO significantly promoted cell proliferation, and network pharmacology analysis suggested that GLSO might act through transient receptor potential vanilloid receptor 1 (TRPV1)/SMAD signaling. Furthermore, GLSO elevated SMAD2/3 expression in skin burn and promoted its nuclear translocation, and TRPV1 expression was also increased upon exposure to GLSO. Cell proliferation and immunofluorescence assays with TRPV1 inhibitor showed that GLSO accelerated skin burn wound healing through TRPV1 and SMADs signaling, which provides a foundation for clinical application of GLSO in the healing of deep skin burns.


Subject(s)
Burns , Reishi , Animals , Burns/drug therapy , Cell Proliferation , Mice , Oils/pharmacology , Smad Proteins , TRPV Cation Channels/pharmacology , Wound Healing
5.
Aging (Albany NY) ; 12(14): 14125-14140, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32692722

ABSTRACT

The mushroom Ganoderma lucidum (G. lucidum Leyss. ex Fr.) Karst has been a traditional Chinese medicine for millennia. In this study, we isolated the Ganoderma lucidum spore oil (GLSO) and evaluated the effect of GLSO on skin burn wound healing and the underlying mechanisms. Mice were used to perform skin wound healing assay. Wound analysis was performed by photography, hematoxylin/eosin staining, Masson's Trichrome staining and immunohistochemical analysis. Microbiota on the wounds were analyzed using the 16s rRNA sequence and quantitative statistics. The lipopolysaccharide (LPS) content was examined in skin wounds and serum using an enzyme-linked immunosorbent assay (ELISA). The expression of Toll-like receptor 4 (TLR4) and the relative levels of inflammatory cytokines were determined by qPCR and immunofluorescence assay. A pseudo-germfree mouse model treated with antibiotics was used to investigate whether GLSO accelerated skin burn wound healing through the skin microbiota. We found that GLSO significantly accelerated the process of skin wound healing and regulated the levels of gram-negative and gram-positive bacteria. Furthermore, GLSO reduced LPS and TLR4, and levels of some other related inflammatory cytokines. The assay with the pseudo-germfree mice model showed that GLSO had a significant acceleration on skin wound healing in comparison with antibiotic treatment. Thus, GLSO downregulated the inflammation by regulating skin microbiota to accelerate skin wound healing. These findings provide a scientific rationale for the potential therapeutic use of GLSO in skin burn injury.


Subject(s)
Dermatitis/drug therapy , Oils/pharmacology , Reishi/chemistry , Skin/microbiology , Spores, Fungal/chemistry , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , Burns/complications , Cytokines/biosynthesis , Germ-Free Life , Lipopolysaccharides/metabolism , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred ICR , Oils/chemistry , Toll-Like Receptor 4/biosynthesis
6.
Food Funct ; 11(1): 1110-1121, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31825431

ABSTRACT

Sarcodon imbricatus (S. imbricatus), a well-known edible mushroom, is one of the most commonly consumed wild mushrooms in China because of its nutritional value. Previous studies have demonstrated that S. imbricatus has immunoregulatory activity. We previously described the potential anti-tumor activity of several types of mushrooms, including S. imbricatus. In this study, the results demonstrate that an aqueous extract of S. imbricatus (SIE) effectively inhibits the growth, migration, and invasion properties of breast cancer cells in vitro and reduces tumor growth in vivo. In addition, the SIE increased serum concentrations of interleukin (IL)-2, IL-6 and tumor necrosis factor-α, natural killer cell activity and the viability of splenocytes and reduced the expression of programmed cell death-Ligand 1 (PD-L1) in 4T1 tumor-bearing mice. Collectively, these results are the first demonstration that the SIE has anti-tumor and immunomodulatory effects in the 4T1 mouse breast cancer model. These findings provide a scientific rationale for the potential therapeutic use of S. imbricatus in breast cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Basidiomycota/chemistry , Biological Products/pharmacology , Breast Neoplasms/drug therapy , Immunomodulation , Agaricales/chemistry , Animals , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/immunology , Cell Line, Tumor , Female , Humans , Interleukin-2/blood , Interleukin-6/blood , MCF-7 Cells , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/blood
7.
J Ethnopharmacol ; 247: 112256, 2020 Jan 30.
Article in English | MEDLINE | ID: mdl-31586690

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The mushroom Ganoderma lucidum (G. lucidum) is a traditional Chinese medicine reported to have a variety of pharmacological properties, including anti-cancer activity. G. lucidum spore oil (GLSO) is a lipid substance extracted from sporoderm-broken spore of G. lucidum. However, the effect of GLSO on breast cancer and the underlying molecular mechanism remain unclear. AIM OF THE STUDY: The aim of this study was to identify the effects of GLSO on breast cancer cells in vitro and in vivo as well as to investigate the mechanistic basis for the anticancer effect of GLSO. MATERIALS AND METHODS: First, in vitro MDA-MB-231 cells were treated with GLSO (0.2, 0.4, and 0.6 µL/mL). The protein levels of B-cell lymphoma-2 (Bcl-2), Bcl-2-associated X (Bax), X-linked inhibitor of apoptosis (XIAP), total poly (ADP-ribose) polymerase (PARP), caspase-3 and caspase-8 were examined using western blotting. The mRNA expression levels of Fas-associated protein with death domain (FADD), TNF receptor-associated factor 2 (TRAF2), caspases-3, -8, -9 and Bax were examined using qRT-PCR. Second, in vivo the anticancer properties of GLSO were assessed by H&E, TUNEL and immunohistochemistry in BALB/c mice injected with 4T1 cells. In addition, the levels of caspase-9/caspase-3 signaling pathway proteins in tumor tissue were evaluated by immunoblotting. Finally, MDA-MB-231 cells were treated with caspase inhibitors to measure cell viability, the protein levels were examined with western blotting. RESULTS: The results in vitro showed that GLSO up-regulated the expression of Bax and caspase-3 in MDA-MB-231 cells, but had no effect on the expression of caspase-8. Moreover, the growth of tumors in vivo was significantly suppressed in the GLSO-treated group. The results of Western blot were consistent with in vitro. In vitro, co-treatment of MDA-MB-231 cells with caspase inhibitors reduced the inhibitory effect of GLSO on cell growth. CONCLUSIONS: GLSO inhibits the growth of MDA-MB-231 cells and tumors in vivo by inducing apoptosis, which may be achieved through the mitochondrial apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Medicine, Chinese Traditional/methods , Oils/pharmacology , Reishi/chemistry , Animals , Breast Neoplasms/pathology , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Mice , Oils/therapeutic use , Spores, Fungal/chemistry
8.
Front Immunol ; 9: 1765, 2018.
Article in English | MEDLINE | ID: mdl-30108589

ABSTRACT

As breast cancer is the leading cause of cancer-related deaths in women population worldwide, ongoing endeavor has been made for alternative regimens with improved efficacy but fewer adverse effects. Recently, active components from the spores of Ganoderma lucidum have attracted much attention for their versatile biological activities owing to the advance in sporoderm-breaking technology. Here, anticancer potential of an extract derived from the sporoderm-breaking spores of G. lucidum (ESG) was explored in a 4T1-breast cancer xenograft mice model. Results showed that ESG was able to suppress 4T1 tumor growth in vivo rather than in vitro. Flowcytometry analysis revealed that ESG could significantly increase both cytotoxic T cell (Tc) population and the ratio of Tc to helper T cell (Th) in peripheral blood of the tumor-bearing mouse; similar promotion on Tc was also found in tumor-infiltrating lymphocyte. Moreover, ESG evidently downregulated the two immune checkpoints, programmed cell death protein-1 (PD-1, in the spleen) and cytotoxic T lymphocyte antigen-4 (CTLA-4, in the tumor), suggesting that ESG could effectively restore the T cell paradigm by recovering the exhaustion status via suppressing the co-inhibitory checkpoints. By 16S rRNA gene sequence analysis on the fecal microbiota, it was found that ESG would remodeling the overall structure of the samples from tumor-bearing mice toward that of the normal counterparts, including 18 genera in 5 phyla, together with regulations on several genes that are responsible for signaling pathways involved in metabolism, cellular processes, and environmental information processing. Collectively, this study demonstrated that ESG would serve as a natural anticancer adjuvant via a restoration on the exhausted Tc, highlighting important clinical implications for the treatment of breast cancer.


Subject(s)
Biological Products/pharmacology , Gastrointestinal Microbiome/drug effects , Mammary Neoplasms, Experimental/drug therapy , Reishi/chemistry , Spores, Fungal/chemistry , T-Lymphocytes, Cytotoxic/drug effects , Animals , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Biological Products/chemistry , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Female , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , RNA, Ribosomal, 16S/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...