Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 42(10): 113220, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37804509

ABSTRACT

A high-sugar diet induces lifestyle-associated metabolic diseases, such as obesity and diabetes, which may underlie the pro-tumor effects of a high-sugar diet. We supply GL261 syngeneic glioblastoma (GBM) mice with a short-term high-glucose drink (HGD) and find an increased survival rate with no evidence of metabolic disease. Modulation of the gut microbiota through HGD supplementation is critical for enhancing the anti-tumor immune response. Single-cell RNA sequencing shows that gut microbiota modulation by HGD supplementation increases the T cell-mediated anti-tumor immune response in GBM mice. We find that the cytotoxic CD4+ T cell population in GBM is increased due to synergy with anti-programmed cell death protein 1 (anti-PD-1) immune checkpoint inhibitors, but this effect depends upon HGD supplementation. Thus, we determine that HGD supplementation enhances anti-tumor immune responses in GBM mice through gut microbiota modulation and suggest that the role of HGD supplementation in GBM should be re-examined.


Subject(s)
Brain Neoplasms , Gastrointestinal Microbiome , Glioblastoma , Mice , Animals , Glioblastoma/metabolism , Brain Neoplasms/metabolism , Glucose , Immunity , Dietary Supplements , Sugars
2.
STAR Protoc ; 4(2): 102278, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37289592

ABSTRACT

Obesity affects susceptibility to sexually transmitted diseases like genital herpes, caused by herpes simplex virus (HSV) 2. The γδ T cells in the vagina play a major role in HSV-2 suppression. Here, we present a protocol for inducing HSV-2 infection intravaginally in high-fat diet-induced obese mice. We describe steps for isolating single cells from vaginal tissue and analyzing cells using single-cell RNA sequencing and flow cytometry. We then detail confirmation of the γδ T cell phenotype in vitro. For complete details on the use and execution of this protocol, please refer to Park et al.1.

3.
Front Immunol ; 13: 919728, 2022.
Article in English | MEDLINE | ID: mdl-35837395

ABSTRACT

The female reproductive tract harbors a unique microbiome, especially the vagina. The human vaginal microbiome exhibits a low diversity and is dominated by Lactobacillus species, compared to the microbiome of other organs. The host and vaginal microbiome mutually coexist in the vaginal microenvironment. Host cells provide Lactobacillus glycogen as an energy source, and Lactobacillus produce lactic acid, which lowers vaginal pH thereby preventing growth of other bacteria. Bacterial vaginosis can modulate host immune systems, and is frequently associated with various aspects of disease, including sexually transmitted infection, gynecologic cancer, and poor pregnancy outcomes. Because of this, numerous studies focused on the impact of the vaginal microbiome on women`s health and disease. Furthermore, numerous epidemiologic studies also have demonstrated various host factors regulate the vaginal microbiome. The female reproductive tract undergoes constant fluctuations due to hormonal cycle, pregnancy, and other extrinsic factors. Depending on these fluctuations, the vaginal microbiome composition can shift temporally and dynamically. In this review, we highlight the current knowledge of how host factors modulate vaginal microbiome composition and how the vaginal microbiome contributes to maintaining homeostasis or inducing pathogenesis. A better understanding of relationship between host and vaginal microbiome could identify novel targets for diagnosis, prognosis, or treatment of microbiome-related diseases.


Subject(s)
Microbiota , Sexually Transmitted Diseases , Vaginosis, Bacterial , Female , Humans , Lactobacillus , Microbiota/physiology , Pregnancy , Vagina
4.
Am J Reprod Immunol ; 82(4): e13167, 2019 10.
Article in English | MEDLINE | ID: mdl-31295378

ABSTRACT

PROBLEM: Dysregulation of extravillous trophoblast (EVT) invasion leads to pregnancy complications, such as pre-eclampsia, fetal growth restriction, and placenta accreta. The aim of this study was to explore the role of SIRT1 in EVT invasion and its underlying mechanism. METHOD OF STUDY: SIRT1-specific siRNA was transfected into Swan 71 cells, an immortalized first trimester trophoblast cell line. The Boyden chamber invasion assay, the scratch wound healing assay, and cell proliferation assay were performed. The expression levels of epithelial-to-mesenchymal transition (EMT) markers, matrix metalloproteinase-2 (MMP-2), MMP-9, p-Akt, Akt, p-p38MAPK, p38MAPK, p-ERK, ERK, p-JNK, JNK, Fas, and Fas ligand (FasL) were examined by western blot. Tube formation assay was conducted by using Matrigel. RESULTS: SIRT1 knockdown by siRNA significantly enhanced invasion and migration as well as the expression of MMP-2, MMP-9, and EMT markers in Swan 71 cells, but reduced proliferation. The effects of SIRT1 knockdown on invasion, migration, proliferation, and endothelial-like tube formation in Swan 71 cells were reversely regulated by blockade of Akt and p38MAPK signaling. In addition, SIRT1 knockdown markedly promoted colocalization of Swan 71 cells to human umbilical vein endothelial cell (HUVEC) networks and induced reduction in Fas and enhancement of FasL. Conditioned media of SIRT1 knockdown-Swan 71 cells caused reduction in cell proliferation and augmentation of cytotoxicity along with increased Fas expression in HUVECs. CONCLUSION: Our results suggest that SIRT1 may be associated with placental development by controlling EVT invasion and spiral artery remodeling via modulation of EMT, MMP-2, MMP-9, Akt/p38MAPK signaling, and Fas/FasL.


Subject(s)
Neovascularization, Physiologic , Sirtuin 1/physiology , Trophoblasts/physiology , Cell Line , Cell Movement , Cell Proliferation , Chorionic Villi , Epithelial-Mesenchymal Transition , Fas Ligand Protein/physiology , Female , Human Umbilical Vein Endothelial Cells/physiology , Humans , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/physiology , Pregnancy , Proto-Oncogene Proteins c-akt/physiology , RNA, Small Interfering , Sirtuin 1/genetics , fas Receptor/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL