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.
Sci Rep ; 12(1): 2505, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169179

ABSTRACT

Mpro, the main protease of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is essential for the viral life cycle. Accordingly, several groups have performed in silico screens to identify Mpro inhibitors that might be used to treat SARS-CoV-2 infections. We selected more than five hundred compounds from the top-ranking hits of two very large in silico screens for on-demand synthesis. We then examined whether these compounds could bind to Mpro and inhibit its protease activity. Two interesting chemotypes were identified, which were further evaluated by characterizing an additional five hundred synthesis on-demand analogues. The compounds of the first chemotype denatured Mpro and were considered not useful for further development. The compounds of the second chemotype bound to and enhanced the melting temperature of Mpro. The most active compound from this chemotype inhibited Mpro in vitro with an IC50 value of 1 µM and suppressed replication of the SARS-CoV-2 virus in tissue culture cells. Its mode of binding to Mpro was determined by X-ray crystallography, revealing that it is a non-covalent inhibitor. We propose that the inhibitors described here could form the basis for medicinal chemistry efforts that could lead to the development of clinically relevant inhibitors.


Subject(s)
Coronavirus 3C Proteases/antagonists & inhibitors , Protease Inhibitors/chemistry , SARS-CoV-2/enzymology , Binding Sites , COVID-19/pathology , COVID-19/virology , Coronavirus 3C Proteases/genetics , Coronavirus 3C Proteases/metabolism , Crystallography, X-Ray , Humans , Molecular Conformation , Molecular Docking Simulation , Nitriles/chemistry , Nitriles/metabolism , Nitriles/pharmacology , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Quinazolines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Virus Replication/drug effects
2.
Mol Cell ; 64(6): 1127-1134, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27984746

ABSTRACT

Human cancers are characterized by the presence of oncogene-induced DNA replication stress (DRS), making them dependent on repair pathways such as break-induced replication (BIR) for damaged DNA replication forks. To better understand BIR, we performed a targeted siRNA screen for genes whose depletion inhibited G1 to S phase progression when oncogenic cyclin E was overexpressed. RAD52, a gene dispensable for normal development in mice, was among the top hits. In cells in which fork collapse was induced by oncogenes or chemicals, the Rad52 protein localized to DRS foci. Depletion of Rad52 by siRNA or knockout of the gene by CRISPR/Cas9 compromised restart of collapsed forks and led to DNA damage in cells experiencing DRS. Furthermore, in cancer-prone, heterozygous APC mutant mice, homozygous deletion of the Rad52 gene suppressed tumor growth and prolonged lifespan. We therefore propose that mammalian RAD52 facilitates repair of collapsed DNA replication forks in cancer cells.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Cyclin E/genetics , DNA Breaks, Double-Stranded , DNA/genetics , Osteosarcoma/genetics , Rad52 DNA Repair and Recombination Protein/genetics , Recombinational DNA Repair , Adenomatous Polyposis Coli Protein/deficiency , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin E/metabolism , DNA/metabolism , G1 Phase , Gene Expression , Genomic Instability , Humans , Mice , Mice, Knockout , Nocodazole/pharmacology , Osteosarcoma/metabolism , Osteosarcoma/mortality , Osteosarcoma/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rad52 DNA Repair and Recombination Protein/antagonists & inhibitors , Rad52 DNA Repair and Recombination Protein/metabolism , S Phase , Stress, Physiological , Survival Analysis
3.
Diabetes ; 54(5): 1415-22, 2005 May.
Article in English | MEDLINE | ID: mdl-15855328

ABSTRACT

Diabetogenic T-cells can be detected in pre-diabetic nonobese diabetic (NOD) mice after transfer in NOD-SCID recipients. Here we demonstrate that 6-week-old pre-diabetic NOD mice, >2 months before disease onset, already harbor pathogenic T-cells in equal numbers to overtly diabetic animals. The delay in diabetes appearance is explained by the presence of regulatory CD4+ CD25+ T-cells that control diabetogenic effectors and that are, in our hands, transforming growth factor (TGF)-beta-dependent. Our present results suggest, however, that diabetes onset is only partly explained by a decline in this regulatory T-cell activity. Another major factor appears to be the progressive resistance of diabetogenic cells to TGF-beta-dependent mediated inhibition. We propose that progression to overt disease correlates with the pathogenic T-cell's escape from TGF-beta-dependent T-cell-mediated regulation.


Subject(s)
Aging/immunology , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Cytokines/blood , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Interleukin-2/blood , Spleen/immunology , T-Lymphocytes/pathology
4.
Proc Natl Acad Sci U S A ; 101 Suppl 2: 14580-5, 2004 Oct 05.
Article in English | MEDLINE | ID: mdl-15340148

ABSTRACT

Converging experimental evidence indicates that CD4(+) regulatory T cells control progression of autoimmune insulitis in nonobese diabetic (NOD) mice. Here, we studied the nature of these regulatory T cells and their mode of action in diabetes-prone NOD Rag(-/-) or severe combined immunodeficient (SCID) mice harboring a transgenic T cell receptor derived from the diabetogenic T cell clone BDC2.5. We first show that diabetes onset is prevented in such mice by infusion of polyclonal CD4(+) T cells expressing L-selectin (CD62L) but not prevented or only marginally prevented by CD4(+)CD25(+) T cells. Similarly, we found with a cotransfer model that CD4(+)CD62L(+) T cells but not CD4(+)CD25(+) T cells inhibited diabetes transfer into NOD SCID recipients by transgenic NOD BDC2.5 SCID cells. Unexpectedly, cotransfer of transgenic NOD BDC2.5 SCID cells and spleen cells from WT diabetic NOD mice did not induce diabetes, whereas each individual population did so. Data are presented arguing for the role of CD4(+)CD62L(+) T cells present within the polyclonal diabetogenic population in mediating this apparently paradoxical effect. Collectively, these data confirm the central role of CD4(+)CD62L(+) regulatory T cells in controlling disease onset in a well defined transgenic model of autoimmune diabetes and suggest the intervention of homeostatic mechanisms as part of their mode of action.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/administration & dosage , CD3 Complex , Diabetes Mellitus, Type 1/etiology , Diabetes Mellitus, Type 1/prevention & control , Genes, T-Cell Receptor , L-Selectin/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , T-Lymphocyte Subsets/immunology
5.
Nat Med ; 9(9): 1202-8, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937416

ABSTRACT

CD3-specific antibodies have the unique capacity to restore self-tolerance in established autoimmunity. They induce long-term remission of overt diabetes in nonobese diabetic (NOD) mice and in human type I diabetes. The underlying mechanisms had been unclear until now. Here we report that treatment with CD3epsilon-specific antibodies induces transferable T-cell-mediated tolerance involving CD4+CD25+ cells. However, these CD4+CD25+ T cells are distinct from naturally occurring regulatory T cells that control physiological autoreactivity. CD3-specific antibody treatment induced remission in NOD Cd28-/- mice that were devoid of such regulatory cells. Remission of diabetes was abrogated by coadministration of a neutralizing transforming growth factor (TGF)-beta-specific antibody. The central role of TGF-beta was further suggested by its increased, long-lasting production by CD4+ T cells from tolerant mice. These data explain the intriguing tolerogenic effect of CD3-specific antibodies and position them as the first clinically applicable pharmacological stimulant of TGF-beta-producing regulatory CD4+ T cells.


Subject(s)
CD3 Complex/immunology , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/immunology , T-Lymphocytes/immunology , Transforming Growth Factor beta/metabolism , Animals , Antibodies/pharmacology , Antigens, CD , Antigens, Differentiation/immunology , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Immune Tolerance/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, Interleukin-2/immunology , Receptors, Interleukin-2/metabolism , Spleen/cytology , Spleen/immunology , T-Lymphocytes/metabolism , Transforming Growth Factor beta/immunology
SELECTION OF CITATIONS
SEARCH DETAIL