Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2435, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499573

ABSTRACT

The potential of immune checkpoint inhibitors (ICI) may be limited in situations where immune cell fitness is impaired. Here, we show that the efficacy of cancer immunotherapies is compromised by the accumulation of senescent cells in mice and in the context of therapy-induced senescence (TIS). Resistance to immunotherapy is associated with a decrease in the accumulation and activation of CD8 T cells within tumors. Elimination of senescent cells restores immune homeostasis within the tumor micro-environment (TME) and increases mice survival in response to immunotherapy. Using single-cell transcriptomic analysis, we observe that the injection of ABT263 (Navitoclax) reverses the exacerbated immunosuppressive profile of myeloid cells in the TME. Elimination of these myeloid cells also restores CD8 T cell proliferation in vitro and abrogates immunotherapy resistance in vivo. Overall, our study suggests that the use of senolytic drugs before ICI may constitute a pharmacological approach to improve the effectiveness of cancer immunotherapies.


Subject(s)
Neoplasms , Tumor Microenvironment , Animals , Mice , Immunotherapy , Neoplasms/pathology , Cellular Senescence
4.
Cell Rep Methods ; 2(1): 100153, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35474871

ABSTRACT

Modeling the tumor-immune cell interactions in humanized mice is complex and limits drug development. Here, we generated easily accessible tumor models by transforming either primary skin fibroblasts or induced pluripotent stem cell-derived cell lines injected in immune-deficient mice reconstituted with human autologous immune cells. Our results showed that fibroblastic, hepatic, or neural tumors were all efficiently infiltrated and partially or totally rejected by autologous immune cells in humanized mice. Characterization of tumor-immune infiltrates revealed high expression levels of the dysfunction markers Tim3 and PD-1 in T cells and an enrichment in regulatory T cells, suggesting rapid establishment of immunomodulatory phenotypes. Inhibition of PD-1 by Nivolumab in humanized mice resulted in increased immune cell infiltration and a slight decrease in tumor growth. We expect that these versatile and accessible cancer models will facilitate preclinical studies and the evaluation of autologous cancer immunotherapies across a range of different tumor cell types.


Subject(s)
Induced Pluripotent Stem Cells , Neoplasms , Mice , Humans , Animals , Induced Pluripotent Stem Cells/metabolism , Programmed Cell Death 1 Receptor , Neoplasms/therapy , Nivolumab , Immunotherapy/methods
5.
Nat Commun ; 13(1): 2061, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35443751

ABSTRACT

The defining features of Alzheimer's disease (AD) include alterations in protein aggregation, immunity, lipid metabolism, synapses, and learning and memory. Of these, lipid abnormalities are the least understood. Here, we investigate the role of Stearoyl-CoA desaturase (SCD), a crucial regulator of fatty acid desaturation, in AD pathogenesis. We show that inhibiting brain SCD activity for 1-month in the 3xTg mouse model of AD alters core AD-related transcriptomic pathways in the hippocampus, and that it concomitantly restores essential components of hippocampal function, including dendritic spines and structure, immediate-early gene expression, and learning and memory itself. Moreover, SCD inhibition dampens activation of microglia, key mediators of spine loss during AD and the main immune cells of the brain. These data reveal that brain fatty acid metabolism links AD genes to downstream immune, synaptic, and functional impairments, identifying SCD as a potential target for AD treatment.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/metabolism , Animals , Cognitive Dysfunction/metabolism , Disease Models, Animal , Fatty Acids/metabolism , Hippocampus/metabolism , Mice , Mice, Transgenic , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
6.
Sci Adv ; 7(48): eabj5629, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34826237

ABSTRACT

Despite advances in COVID-19 management, identifying patients evolving toward death remains challenging. To identify early predictors of mortality within 60 days of symptom onset (DSO), we performed immunovirological assessments on plasma from 279 individuals. On samples collected at DSO11 in a discovery cohort, high severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral RNA (vRNA), low receptor binding domain­specific immunoglobulin G and antibody-dependent cellular cytotoxicity, and elevated cytokines and tissue injury markers were strongly associated with mortality, including in patients on mechanical ventilation. A three-variable model of vRNA, with predefined adjustment by age and sex, robustly identified patients with fatal outcome (adjusted hazard ratio for log-transformed vRNA = 3.5). This model remained robust in independent validation and confirmation cohorts. Since plasma vRNA's predictive accuracy was maintained at earlier time points, its quantitation can help us understand disease heterogeneity and identify patients who may benefit from new therapies.

7.
J Immunol ; 203(10): 2735-2745, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31578272

ABSTRACT

Therapeutic uses of mesenchymal stromal cells (MSCs) have emerged over the past decade. Yet, their effect on tumor growth remains highly debated, particularly in an immune competent environment. In this study, we wanted to investigate the impact of human umbilical cord-derived MSCs (hUC-MSCs) on tumor growth in humanized mice generated by the human adoptive transfer of PBMCs or the cotransplantation of hematopoietic stem cells and human thymic tissue (human BLT [Hu-BLT]). Our results showed that the growth and immune rejection of engineered human fibroblastic tumors was not altered by the injection of hUC-MSCs in immune-deficient or humanized mice, respectively. This was observed whether tumor cells were injected s.c. or i.v. and independently of the injection route of the hUC-MSCs. Moreover, only in Hu-BLT mice did hUC-MSCs have some effects on the tumor-immune infiltrate, yet without altering tumor growth. These results demonstrate that hUC-MSCs do not promote fibroblastic tumor growth and neither do they prevent tumor infiltration and rejection by immune cells in humanized mice.


Subject(s)
Lymphocytes, Tumor-Infiltrating/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Adoptive Transfer , Animals , Cell Line, Transformed/transplantation , Fibroblasts/transplantation , Genetic Vectors , Graft Rejection/immunology , Heterografts , Humans , Injections, Intravenous , Injections, Subcutaneous , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/immunology , Radiation Chimera , Specific Pathogen-Free Organisms , Telomerase/genetics , Telomerase/physiology , Thymus Gland/transplantation , Wharton Jelly/cytology
8.
J Exp Clin Cancer Res ; 38(1): 251, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31196146

ABSTRACT

BACKGROUND: Cardiac glycosides are approved for the treatment of heart failure as Na+/K+ pump inhibitors. Their repurposing in oncology is currently investigated in preclinical and clinical studies. However, the identification of a specific cancer type defined by a molecular signature to design targeted clinical trials with cardiac glycosides remains to be characterized. Here, we demonstrate that cardiac glycoside proscillaridin A specifically targets MYC overexpressing leukemia cells and leukemia stem cells by causing MYC degradation, epigenetic reprogramming and leukemia differentiation through loss of lysine acetylation. METHODS: Proscillaridin A anticancer activity was investigated against a panel of human leukemia and solid tumor cell lines with different MYC expression levels, overexpression in vitro systems and leukemia stem cells. RNA-sequencing and differentiation studies were used to characterize transcriptional and phenotypic changes. Drug-induced epigenetic changes were studied by chromatin post-translational modification analysis, expression of chromatin regulators, chromatin immunoprecipitation, and mass-spectrometry. RESULTS: At a clinically relevant dose, proscillaridin A rapidly altered MYC protein half-life causing MYC degradation and growth inhibition. Transcriptomic profile of leukemic cells after treatment showed a downregulation of genes involved in MYC pathways, cell replication and an upregulation of hematopoietic differentiation genes. Functional studies confirmed cell cycle inhibition and the onset of leukemia differentiation even after drug removal. Proscillaridin A induced a significant loss of lysine acetylation in histone H3 (at lysine 9, 14, 18 and 27) and in non-histone proteins such as MYC itself, MYC target proteins, and a series of histone acetylation regulators. Global loss of acetylation correlated with the rapid downregulation of histone acetyltransferases. Importantly, proscillaridin A demonstrated anticancer activity against lymphoid and myeloid stem cell populations characterized by MYC overexpression. CONCLUSION: Overall, these results strongly support the repurposing of proscillaridin A in MYC overexpressing leukemia.


Subject(s)
Antineoplastic Agents/adverse effects , Gene Expression/drug effects , Genes, myc , Heart Failure/etiology , Leukemia/genetics , Lysine/metabolism , Proscillaridin/adverse effects , Acetylation , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/genetics , Chromatin/metabolism , Dose-Response Relationship, Drug , Epigenesis, Genetic/drug effects , Gene Expression Profiling , Histones/metabolism , Humans , Leukemia/complications , Leukemia/drug therapy , Leukemia/metabolism , Models, Biological , Proscillaridin/therapeutic use , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
9.
Aging Cell ; 18(4): e12971, 2019 08.
Article in English | MEDLINE | ID: mdl-31148373

ABSTRACT

Some studies show eliminating senescent cells rejuvenate aged mice and attenuate deleterious effects of chemotherapy. Nevertheless, it remains unclear whether senescence affects immune cell function. We provide evidence that exposure of mice to ionizing radiation (IR) promotes the senescent-associated secretory phenotype (SASP) and expression of p16INK4a in splenic cell populations. We observe splenic T cells exhibit a reduced proliferative response when cultured with allogenic cells in vitro and following viral infection in vivo. Using p16-3MR mice that allow elimination of p16INK4a -positive cells with exposure to ganciclovir, we show that impaired T-cell proliferation is partially reversed, mechanistically dependent on p16INK4a expression and the SASP. Moreover, we found macrophages isolated from irradiated spleens to have a reduced phagocytosis activity in vitro, a defect also restored by the elimination of p16INK4a expression. Our results provide molecular insight on how senescence-inducing IR promotes loss of immune cell fitness, which suggest senolytic drugs may improve immune cell function in aged and patients undergoing cancer treatment.


Subject(s)
Cellular Senescence/radiation effects , Radiation, Ionizing , Spleen/metabolism , Spleen/radiation effects , T-Lymphocytes/immunology , T-Lymphocytes/radiation effects , Animals , Antiviral Agents/therapeutic use , Arenaviridae Infections/drug therapy , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Cell Proliferation/radiation effects , Cells, Cultured , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Ganciclovir/therapeutic use , Lymphocytic choriomeningitis virus/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Phenotype , Rejuvenation/physiology , Spleen/virology
10.
J Vis Exp ; (71): e50057, 2013 Jan 30.
Article in English | MEDLINE | ID: mdl-23407302

ABSTRACT

Assessing the expression pattern of a gene, as well as the subcellular localization properties of its transcribed RNA, are key features for understanding its biological function during development. RNA in situ hybridization (RNA-ISH) is a powerful method used for visualizing RNA distribution properties, be it at the organismal, cellular or subcellular levels. RNA-ISH is based on the hybridization of a labeled nucleic acid probe (e.g. antisense RNA, oligonucleotides) complementary to the sequence of an mRNA or a non-coding RNA target of interest. As the procedure requires primary sequence information alone to generate sequence-specific probes, it can be universally applied to a broad range of organisms and tissue specimens. Indeed, a number of large-scale ISH studies have been implemented to document gene expression and RNA localization dynamics in various model organisms, which has led to the establishment of important community resources. While a variety of probe labeling and detection strategies have been developed over the years, the combined usage of fluorescently-labeled detection reagents and enzymatic signal amplification steps offer significant enhancements in the sensitivity and resolution of the procedure. Here, we describe an optimized fluorescent in situ hybridization method (FISH) employing tyramide signal amplification (TSA) to visualize RNA expression and localization dynamics in staged Drosophila embryos. The procedure is carried out in 96-well PCR plate format, which greatly facilitates the simultaneous processing of large numbers of samples.


Subject(s)
Drosophila/genetics , In Situ Hybridization, Fluorescence/methods , RNA/analysis , Animals , Drosophila/chemistry , Drosophila/embryology , RNA/biosynthesis , RNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...