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.
Biomaterials ; 312: 122750, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39126779

ABSTRACT

Infiltration of immunosuppressive cells into the breast tumor microenvironment (TME) is associated with suppressed effector T cell (Teff) responses, accelerated tumor growth, and poor clinical outcomes. Previous studies from our group and others identified infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) as critical contributors to immune dysfunction in the orthotopic claudin-low tumor model, limiting the efficacy of adoptive cellular therapy. However, approaches to target these cells in the TME are currently lacking. To overcome this barrier, polymeric micellular nanoparticles (PMNPs) were used for the co-delivery of small molecule drugs activating Toll-like receptors 7 and 8 (TLR7/8) and inhibiting PI3K delta (PI3Kδ). The immunomodulation of the TME by TLR7/8 agonist and PI3K inhibitor led to type 1 macrophage polarization, decreased MDSC accumulation and selectively decreased tissue-resident Tregs in the TME, while enhancing the T and B cell adaptive immune responses. PMNPs significantly enhanced the anti-tumor activity of local radiation therapy (RT) in mice bearing orthotopic claudin-low tumors compared to RT alone. Taken together, these data demonstrate that RT combined with a nanoformulated immunostimulant diminished the immunosuppressive TME resulting in tumor regression. These findings set the stage for clinical studies of this approach.


Subject(s)
Nanoparticles , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Tumor Microenvironment , Animals , Tumor Microenvironment/drug effects , Toll-Like Receptor 7/agonists , Female , Nanoparticles/chemistry , Mice , Toll-Like Receptor 8/agonists , Immunomodulation/drug effects , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Mice, Inbred BALB C , Micelles , Humans
3.
Nat Commun ; 15(1): 6000, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39019846

ABSTRACT

Type II innate lymphoid cells (ILC2s) maintain homeostasis and barrier integrity in mucosal tissues. In both mice and humans, ILC2s poorly reconstitute after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Determining the mechanisms involved in their impaired reconstitution could improve transplant outcomes. By integrating single-cell chromatin and transcriptomic analyses of transplanted ILC2s, we identify a previously unreported population of converted ILC1-like cells in the mouse small intestine post-transplant. Exposure of ILC2s to proinflammatory cytokines resulted in a mixed ILC1-ILC2 phenotype but was able to convert only a small population of ILC2s to ILC1s, which were found post-transplant. Whereas ILC2s protected against acute graft-versus-host disease (aGVHD) mediated mortality, infusion of proinflammatory cytokine-exposed ILC2s accelerated aGvHD. Interestingly, murine ILC2 reconstitution post-HSCT is decreased in the presence of alloreactive T cells. Finally, peripheral blood cells from human patients with aGvHD have an altered ILC2-associated chromatin landscape compared to transplanted controls. These data demonstrate that following transplantation ILC2s convert to a pro-pathogenic population with an ILC1-like chromatin state and provide insights into the contribution of ILC plasticity to the impaired reconstitution of ILC2 cells, which is one of several potential mechanisms for the poor reconstitution of these important cells after allo-HSCT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Immunity, Innate , Lymphocytes , Mice, Inbred C57BL , Transplantation, Homologous , Hematopoietic Stem Cell Transplantation/methods , Animals , Humans , Graft vs Host Disease/immunology , Mice , Lymphocytes/immunology , Cytokines/metabolism , Cell Plasticity , Female , Intestine, Small/immunology , Male , Mice, Inbred BALB C , Chromatin/metabolism
4.
bioRxiv ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38559220

ABSTRACT

Infiltration of immunosuppressive cells into the breast tumor microenvironment (TME) is associated with suppressed effector T cell (Teff) responses, accelerated tumor growth, and poor clinical outcomes. Previous studies from our group and others identified infiltration of immunosuppressive myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) as critical contributors to immune dysfunction in the orthotopic triple-negative breast cancer (TNBC) tumor model limiting the efficacy of adoptive cellular therapy. However, approaches to target these cells specifically in the TME are currently lacking. To overcome this barrier, polymeric micelles nanoparticles (PMNPs) were used for co-delivery of small molecule drugs activating Toll-like receptors 7 and 8 (TLR7/8) and inhibiting PI3K delta. The immunomodulation of the TME by TLR7/8 agonist and PI3K inhibitor altered macrophage polarization, reduced MDSC accumulation and selectively decreased tissue-resident Tregs in the TME, while enhancing the T and B cell adaptive immune response. PMNPs significantly enhanced the anti-tumor activity of local radiation therapy (RT) in mice bearing orthotopic TNBC tumors compared to RT alone. Taken together, these data demonstrate that RT combined with a nanoformulated immunostimulant restructured the TME and has promising potential for future translation combined with RT for patients with TNBC.

5.
J Immunol ; 207(10): 2598-2607, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34607937

ABSTRACT

We have previously shown in a model of claudin-low breast cancer that regulatory T cells (Tregs) are increased in the tumor microenvironment (TME) and express high levels of PD-1. In mouse models and patients with triple-negative breast cancer, it is postulated that one cause for the lack of activity of anti-PD-1 therapy is the activation of PD-1-expressing Tregs in the TME. We hypothesized that the expression of PD-1 on Tregs would lead to enhanced suppressive function of Tregs and worsen antitumor immunity during PD-1 blockade. To evaluate this, we isolated Tregs from claudin-low tumors and functionally evaluated them ex vivo. We compared transcriptional profiles of Tregs isolated from tumor-bearing mice with or without anti-PD-1 therapy using RNA sequencing. We found several genes associated with survival and proliferation pathways; for example, Jun, Fos, and Bcl2 were significantly upregulated in Tregs exposed to anti-PD-1 treatment. Based on these data, we hypothesized that anti-PD-1 treatment on Tregs results in a prosurvival phenotype. Indeed, Tregs exposed to PD-1 blockade had significantly higher levels of Bcl-2 expression, and this led to increased protection from glucocorticoid-induced apoptosis. In addition, we found in vitro and in vivo that Tregs in the presence of anti-PD-1 proliferated more than control Tregs PD-1 blockade significantly increased the suppressive activity of Tregs at biologically relevant Treg/Tnaive cell ratios. Altogether, we show that this immunotherapy blockade increases proliferation, protection from apoptosis, and suppressive capabilities of Tregs, thus leading to enhanced immunosuppression in the TME.


Subject(s)
Immune Checkpoint Inhibitors/pharmacology , T-Lymphocytes, Regulatory/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes, Regulatory/drug effects , Tumor Escape/drug effects , Tumor Escape/immunology , Tumor Microenvironment/genetics
6.
Blood Adv ; 2(18): 2307-2319, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30228128

ABSTRACT

Chronic graft-versus-host disease (cGVHD) causes significant morbidity and mortality in patients after allogeneic bone marrow (BM) or stem cell transplantation (allo-SCT). Recent work has indicated that both T and B lymphocytes play an important role in the pathophysiology of cGVHD. Previously, our group showed a critical role for the germinal center response in the function of B cells using a bronchiolitis obliterans (BO) model of cGVHD. Here, we demonstrated for the first time that cGVHD is associated with severe defects in the generation of BM B lymphoid and uncommitted common lymphoid progenitor cells. We found an increase in the number of donor CD4+ T cells in the BM of mice with cGVHD that was negatively correlated with B-cell development and the frequency of osteoblasts and Prrx-1-expressing perivascular stromal cells, which are present in the B-cell niche. Use of anti-DR3 monoclonal antibodies to enhance the number of donor regulatory T cells (Tregs) in the donor T-cell inoculum ameliorated the pathology associated with BO in this model. This correlated with an increased number of endosteal osteoblastic cells and significantly improved the generation of B-cell precursors in the BM after allo-SCT. Our work indicates that donor Tregs play a critical role in preserving the generation of B-cell precursors in the BM after allo-SCT. Approaches to enhance the number and/or function of donor Tregs that do not enhance conventional T-cell activity may be important to decrease the incidence and severity of cGVHD in part through normal B-cell lymphopoiesis.


Subject(s)
B-Lymphocytes/immunology , Bone Marrow Cells/immunology , Bronchiolitis Obliterans/etiology , Cell Differentiation , Graft vs Host Disease/etiology , Animals , B-Lymphocytes/metabolism , Biomarkers , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bronchiolitis Obliterans/pathology , Bronchiolitis Obliterans/physiopathology , Cell Differentiation/immunology , Chronic Disease , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Graft vs Host Disease/pathology , Immunophenotyping , Lymphocyte Depletion , Mice , Mice, Transgenic , Osteoblasts/immunology , Osteoblasts/metabolism , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Precursor Cells, B-Lymphoid/metabolism , Severity of Illness Index , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
7.
Blood ; 130(3): 348-359, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28550042

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that affects the function and development of immune cells. Here, we show that recipient mice receiving AhR-/- T cells have improved survival and decreased acute graft-versus-host disease (aGVHD) in 2 different murine allogeneic bone marrow transplant (BMT) models. We also show that CD4+ T cells lacking AhR demonstrate reduced accumulation in secondary lymphoid tissue because of low levels of proliferation 4 days after BMT. Additionally, we found a significant increase in the quantity of peripherally induced regulatory donor T (pTreg) cells in the colon of recipients transplanted with AhR-/- T cells 14 days after transplant. Blockade of AhR using a clinically available AhR antagonist greatly enhanced the in vitro generation of inducible Treg (iTreg) cells from naïve CD4+ human T cells. We have identified AhR as a novel target on donor T cells that is critical to the pathogenesis of aGVHD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Bone Marrow Transplantation , Colon/immunology , Graft vs Host Disease/prevention & control , Receptors, Aryl Hydrocarbon/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Colon/pathology , Gene Expression Regulation , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Purines/pharmacology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Sirolimus/pharmacology , Survival Analysis , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/transplantation , Transplantation, Heterologous
8.
Biochim Biophys Acta ; 1790(10): 1266-73, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19520149

ABSTRACT

BACKGROUND: In chordates, retinoid metabolism is an important target of short-chain dehydrogenases/reductases (SDRs). It is not known whether SDRs play a role in retinoid metabolism of protostomes, such as Drosophila melanogaster. METHODS: Drosophila genome was searched for genes encoding proteins with approximately 50% identity to human retinol dehydrogenase 12 (RDH12). The corresponding proteins were expressed in Sf9 cells and biochemically characterized. Their phylogenetic relationships were analyzed using PHYLIP software. RESULTS: A total of six Drosophila SDR genes were identified. Five of these genes are clustered on chromosome 2 and one is located on chromosome X. The deduced proteins are 300 to 406 amino acids long and are associated with microsomal membranes. They recognize all-trans-retinaldehyde and all-trans-3-hydroxyretinaldehyde as substrates and prefer NADPH as a cofactor. Phylogenetically, Drosophila SDRs belong to the same branch of the SDR superfamily as human RDH12, indicating a common ancestry early in bilaterian evolution, before a protostome-deuterostome split. CONCLUSIONS: Similarities in the substrate and cofactor specificities of Drosophila versus human SDRs suggest conservation of their function in retinoid metabolism throughout protostome and deuterostome phyla. GENERAL SIGNIFICANCE: The discovery of Drosophila retinaldehyde reductases sheds new light on the conversion of beta-carotene and zeaxantine to visual pigment and provides a better understanding of the evolutionary roots of retinoid-active SDRs.


Subject(s)
Drosophila Proteins/genetics , Fatty Acid Synthases/genetics , NADH, NADPH Oxidoreductases/genetics , Retinoids/metabolism , Alcohol Oxidoreductases/classification , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amino Acid Sequence , Animals , Cell Line , Drosophila Proteins/metabolism , Electrophoresis, Polyacrylamide Gel , Fatty Acid Synthases/classification , Fatty Acid Synthases/metabolism , Humans , Isoenzymes/classification , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Molecular Sequence Data , NADH, NADPH Oxidoreductases/classification , NADH, NADPH Oxidoreductases/metabolism , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retinaldehyde/chemistry , Retinaldehyde/metabolism , Sequence Homology, Amino Acid , Spodoptera , Stereoisomerism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL