Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters











Database
Language
Publication year range
1.
Science ; 382(6676): 1270-1276, 2023 12 15.
Article in English | MEDLINE | ID: mdl-38096385

ABSTRACT

Current HIV vaccines designed to stimulate CD8+ T cells have failed to induce immunologic control upon infection. The functions of vaccine-induced HIV-specific CD8+ T cells were investigated here in detail. Cytotoxic capacity was significantly lower than in HIV controllers and was not a consequence of low frequency or unaccumulated functional cytotoxic proteins. Low cytotoxic capacity was attributable to impaired degranulation in response to the low antigen levels present on HIV-infected targets. The vaccine-induced T cell receptor (TCR) repertoire was polyclonal and transduction of these TCRs conferred the same reduced functions. These results define a mechanism accounting for poor antiviral activity induced by these vaccines and suggest that an effective CD8+ T cell response may require a vaccination strategy that drives further TCR clonal selection.


Subject(s)
AIDS Vaccines , Cell Degranulation , Cytotoxicity, Immunologic , HIV Infections , T-Lymphocytes, Cytotoxic , Humans , AIDS Vaccines/immunology , Clone Cells , HIV Infections/prevention & control , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology , Cell Degranulation/immunology
2.
Sci Rep ; 9(1): 15458, 2019 10 29.
Article in English | MEDLINE | ID: mdl-31664073

ABSTRACT

Cholesterol is a critical component of membranes and a precursor for hormones and other signaling molecules. Previously, we showed that unlike astrocytes, glioblastoma cells do not downregulate cholesterol synthesis when plated at high density. In this report, we show that high cell density induces ABCA1 expression in glioblastoma cells, enabling them to get rid of excess cholesterol generated by an activated cholesterol biosynthesis pathway. Because oxysterols are agonists for Liver X Receptors (LXRs), we investigated whether increased cholesterol activates LXRs to maintain cholesterol homeostasis in highly-dense glioblastoma cells. We observed that dense cells had increased oxysterols, which activated LXRß to upregulate ABCA1. Cells with CRISPR-mediated knockdown of LXRß, but not ABCA1, had decreased cell cycle progression and cell survival, and decreased feedback repression of the mevalonate pathway in densely-plated glioma cells. LXRß gene expression poorly correlates with ABCA1 in glioblastoma patients, and expression of each gene correlates with poor patient prognosis in different prognostic subtypes. Finally, gene expression and lipidomics analyses cells revealed that LXRß regulates the expression of immune response gene sets and lipids known to be involved in immune modulation. Thus, therapeutic targeting of LXRß in glioblastoma might be effective through diverse mechanisms.


Subject(s)
ATP Binding Cassette Transporter 1/physiology , Brain Neoplasms/pathology , Cell Proliferation/physiology , Glioblastoma/pathology , Lipid Metabolism , Liver X Receptors/physiology , ATP Binding Cassette Transporter 1/genetics , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cholesterol/metabolism , Glioblastoma/immunology , Glioblastoma/metabolism , Homeostasis , Humans , Liver X Receptors/metabolism , Mevalonic Acid/metabolism , Signal Transduction , Transcription, Genetic
3.
Mol Cell ; 67(6): 1013-1025.e9, 2017 Sep 21.
Article in English | MEDLINE | ID: mdl-28867293

ABSTRACT

In response to stresses, cells often halt normal cellular processes, yet stress-specific pathways must bypass such inhibition to generate effective responses. We investigated how cells redistribute global transcriptional activity in response to DNA damage. We show that an oscillatory increase of p53 levels in response to double-strand breaks drives a counter-oscillatory decrease of MYC levels. Using RNA sequencing (RNA-seq) of newly synthesized transcripts, we found that p53-mediated reduction of MYC suppressed general transcription, with the most highly expressed transcripts reduced to a greater extent. In contrast, upregulation of p53 targets was relatively unaffected by MYC suppression. Reducing MYC during the DNA damage response was important for cell-fate regulation, as counteracting MYC repression reduced cell-cycle arrest and elevated apoptosis. Our study shows that global inhibition with specific activation of transcriptional pathways is important for the proper response to DNA damage; this mechanism may be a general principle used in many stress responses.


Subject(s)
Breast Neoplasms/genetics , DNA Breaks, Double-Stranded , Proto-Oncogene Proteins c-myc/genetics , Transcription, Genetic , Transcriptome , Tumor Suppressor Protein p53/genetics , Apoptosis , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CRISPR-Cas Systems , Cell Cycle Checkpoints , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , MCF-7 Cells , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Signal Transduction , Time Factors , Transfection , Tumor Suppressor Protein p53/metabolism
4.
mBio ; 8(4)2017 07 11.
Article in English | MEDLINE | ID: mdl-28698273

ABSTRACT

From various screens, we found that Kaposi's sarcoma-associated herpesvirus (KSHV) viral microRNAs (miRNAs) target several enzymes in the mevalonate/cholesterol pathway. 3-Hydroxy-3-methylglutaryl-coenzyme A (CoA) synthase 1 (HMGCS1), 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR [a rate-limiting step in the mevalonate pathway]), and farnesyl-diphosphate farnesyltransferase 1 (FDFT1 [a committed step in the cholesterol branch]) are repressed by multiple KSHV miRNAs. Transfection of viral miRNA mimics in primary endothelial cells (human umbilical vein endothelial cells [HUVECs]) is sufficient to reduce intracellular cholesterol levels; however, small interfering RNAs (siRNAs) targeting only HMGCS1 did not reduce cholesterol levels. This suggests that multiple targets are needed to perturb this tightly regulated pathway. We also report here that cholesterol levels were decreased in de novo-infected HUVECs after 7 days. This reduction is at least partially due to viral miRNAs, since the mutant form of KSHV lacking 10 of the 12 miRNA genes had increased cholesterol compared to wild-type infections. We hypothesized that KSHV is downregulating cholesterol to suppress the antiviral response by a modified form of cholesterol, 25-hydroxycholesterol (25HC). We found that the cholesterol 25-hydroxylase (CH25H) gene, which is responsible for generating 25HC, had increased expression in de novo-infected HUVECs but was strongly suppressed in long-term latently infected cell lines. We found that 25HC inhibits KSHV infection when added exogenously prior to de novo infection. In conclusion, we found that multiple KSHV viral miRNAs target enzymes in the mevalonate pathway to modulate cholesterol in infected cells during latency. This repression of cholesterol levels could potentially be beneficial to viral infection by decreasing the levels of 25HC.IMPORTANCE A subset of viruses express unique microRNAs (miRNAs), which act like cellular miRNAs to generally repress host gene expression. A cancer virus, Kaposi's sarcoma-associated herpesvirus (KSHV, or human herpesvirus 8 [HHV-8]), encodes multiple miRNAs that repress gene expression of multiple enzymes that are important for cholesterol synthesis. In cells with these viral miRNAs or with natural infection, cholesterol levels are reduced, indicating these viral miRNAs decrease cholesterol levels. A modified form of cholesterol, 25-hydroxycholesterol, is generated directly from cholesterol. Addition of 25-hydroxycholesterol to primary cells inhibited KSHV infection of cells, suggesting that viral miRNAs may decrease cholesterol levels to decrease the concentration of 25-hydroxycholesterol and to promote infection. These results suggest a new virus-host relationship and indicate a previously unidentified viral strategy to lower cholesterol levels.


Subject(s)
Cholesterol/metabolism , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Hydroxycholesterols/pharmacology , MicroRNAs/metabolism , RNA, Viral/metabolism , Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Cell Line , Endothelial Cells/virology , Gene Expression Regulation, Viral , Herpesvirus 8, Human/metabolism , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Metabolic Networks and Pathways , MicroRNAs/genetics , RNA, Small Interfering/metabolism , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Steroid Hydroxylases/genetics , Virus Latency
5.
Oncotarget ; 8(9): 14860-14875, 2017 Feb 28.
Article in English | MEDLINE | ID: mdl-28118603

ABSTRACT

A hallmark of cellular transformation is the evasion of contact-dependent inhibition of growth. To find new therapeutic targets for glioblastoma, we looked for pathways that are inhibited by high cell density in astrocytes but not in glioma cells. Here we report that glioma cells have disabled the normal controls on cholesterol synthesis. At high cell density, astrocytes turn off cholesterol synthesis genes and have low cholesterol levels, but glioma cells keep this pathway on and maintain high cholesterol. Correspondingly, cholesterol pathway upregulation is associated with poor prognosis in glioblastoma patients. Densely-plated glioma cells increase oxygen consumption, aerobic glycolysis, and the pentose phosphate pathway to synthesize cholesterol, resulting in a decrease in reactive oxygen species, TCA cycle intermediates, and ATP. This constitutive cholesterol synthesis is controlled by the cell cycle, as it can be turned off by cyclin-dependent kinase inhibitors and it correlates with disabled cell cycle control though loss of p53 and RB. Finally, glioma cells, but not astrocytes, are sensitive to cholesterol synthesis inhibition downstream of the mevalonate pathway, suggesting that specifically targeting cholesterol synthesis might be an effective treatment for glioblastoma.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Transformation, Neoplastic/pathology , Cholesterol/metabolism , Glioblastoma/pathology , Astrocytes/cytology , Astrocytes/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Count , Cell Division , Cell Transformation, Neoplastic/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glycolysis/drug effects , Humans , Oxygen Consumption/drug effects , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL