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.
Oncoimmunology ; 11(1): 2026020, 2022.
Article in English | MEDLINE | ID: mdl-35111385

ABSTRACT

Galectin-3 (Gal3) can be expressed by many cells in the tumor microenvironment (TME), including cancer cells, cancer-associated fibroblasts, tumor-associated macrophages, and regulatory T cells (Tregs). In addition to immunosuppression, Gal3 expression has been connected to malignant cell transformation, tumor progression, and metastasis. In the present study, we found spontaneous T-cell responses against Gal3-derived peptides in PBMCs from both healthy donors and cancer patients. We isolated and expanded these Gal3-specific T cells in vitro and showed that they could directly recognize target cells that expressed Gal3. Finally, therapeutic vaccination with a long Gal3-derived peptide epitope, which induced the expansion of Gal3-specific CD8+ T cells in vivo, showed a significant tumor-growth delay in mice inoculated with EO771.LMB metastatic mammary tumor cells. This was associated with a significantly lower percentage of both Tregs and tumor-infiltrating Gal3+ cells in the non-myeloid CD45+CD11b- compartment and with an alteration of the T-cell memory populations in the spleens of Gal3-vaccinated mice. These results suggest that by activating Gal3-specific T cells by an immune-modulatory vaccination, we can target Gal3-producing cells in the TME, and thereby induce a more immune permissive TME. This indicates that Gal3 could be a novel target for therapeutic cancer vaccines.


Subject(s)
Cancer Vaccines , Neoplasms , Animals , CD8-Positive T-Lymphocytes/metabolism , Galectin 3/metabolism , Humans , Mice , Tumor Microenvironment , Vaccination , Vaccines, Subunit
2.
Cell Stress ; 3(10): 319-327, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31656949

ABSTRACT

PD-L1-specific T cells are a natural part of the T-cell repertoire in humans. Hence, we have previously described spontaneous CD8+ and CD4+ T-cell reactivity against PD-L1 in the peripheral blood of patients with various cancers as well as in healthy donors. It is well described that the expression of the PD-L1 protein is introduced in cells by pro-inflammatory cytokines, e.g. IFN-γ. In the current study, we were able to directly link inflammation with PD-L1-specific T cells by showing that inflammatory mediators such as IFN-γ generate measurable numbers of PD-L1-specific T cells in human PBMCs as well as in in vivo models. These PD-L1-specific T cells can vigorously modulate the cell compartments of the local environment. PD-L1-specific T cells may be important for immune homeostasis by sustaining the ongoing inflammatory response by the suppression of regulatory cell function both directly and indirectly.

3.
Mol Nutr Food Res ; 62(22): e1800510, 2018 11.
Article in English | MEDLINE | ID: mdl-30211987

ABSTRACT

SCOPE: This study addresses whether administration of Lactobacillus rhamnosus HN001 could mitigate the effects of a compromised gut microbiota on the composition of mature leukocytes and granulocyte-macrophage progenitor cells (GMPs) in newborn mice. METHODS AND RESULTS: Pregnant dams receive oral broad-spectrum antibiotics, which dramatically decrease the gut microbial composition analyzed by 16S rRNA sequencing. Perinatal antibiotic treatment decreases the proportions of bone marrow (BM) GMPs (postnatal day (PND2): 0.5% vs 0.8%, PND4: 0.2% to 0.6%) and mature granulocytes (33% vs 24% at PND2), and spleen granulocytes (7% vs 17% at PND2) and B cells (PND2:18% vs 28%, PND4:11% vs 22%). At PND35, T helper (Th) cells (20% vs 14%) and cytotoxic T (Tc) cells (10% vs 8%) decrease in the spleen. Oral administration of L. rhamnosus HN001 to neonatal pups (PND1-7) restores the antibiotic-induced changes of GMPs and granulocytes in BM and spleen, and further increases splenic granulocytes in control pups. At PND35, splenic proportions of B and Th but not Tc cells are restored. CONCLUSION: Postnatal administration of a single bacterial strain efficiently restores granulopoiesis and most T cell activation in neonatal mice that suffer from a perinatal antibiotic-induced compromised gut microbiota at birth.


Subject(s)
Anti-Bacterial Agents/adverse effects , Lacticaseibacillus rhamnosus , Myelopoiesis/drug effects , Probiotics/pharmacology , T-Lymphocytes/physiology , Administration, Oral , Animals , Animals, Newborn , Anti-Bacterial Agents/administration & dosage , Female , Gastrointestinal Microbiome , Litter Size/drug effects , Lymphocyte Activation/drug effects , Male , Mice, Inbred C57BL , Pregnancy , Spleen/cytology , T-Lymphocytes/drug effects
4.
J Diabetes Res ; 2016: 6321980, 2016.
Article in English | MEDLINE | ID: mdl-26783537

ABSTRACT

Neonatal studies in different mouse strains reveal that early life colonization affects the development of adaptive immunity in mice. The nonobese diabetic (NOD) mouse spontaneously develops autoimmune diabetes, but neonatal studies of NOD mice are lacking. We hypothesized that NOD mice deviate from another much used mouse strain, C57BL/6, with respect to postnatal microbiota and/or hematopoiesis and compared this in newborn mice of dams housed under the same conditions. A distinct bacteria profile rich in staphylococci was found at postnatal days (PND) 1-4 in NOD mice. Furthermore, a distinct splenic cell profile high in a granulocytic phenotype was evident in the neonatal NOD mice whereas neonatal C57BL/6 mice showed a profile rich in monocytes. Neonatal expression of Reg3g and Muc2 in the gut was deviating in NOD mice and coincided with fewer bacteria attaching to the Mucosal surface in NOD compared to C57BL/6 mice.


Subject(s)
Diabetes Mellitus/blood , Diabetes Mellitus/microbiology , Gastrointestinal Microbiome , Hematopoiesis , Intestines/microbiology , Animals , Animals, Newborn , Bacterial Adhesion , Biomarkers/metabolism , Diabetes Mellitus/genetics , Disease Models, Animal , Gene Expression Regulation, Developmental , Granulocytes/metabolism , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Monocytes/metabolism , Mucin-2/genetics , Mucin-2/metabolism , Pancreatitis-Associated Proteins , Phenotype , Proteins/genetics , Proteins/metabolism , Species Specificity , Spleen/cytology , Spleen/metabolism , Staphylococcus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...