Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bio Protoc ; 13(2)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36789164

ABSTRACT

Genome-wide CRISPR-based screening is a powerful tool in forward genetics, enabling biologic discovery by linking a desired phenotype to a specific genetic perturbation. However, hits from a genome-wide screen require individual validation to reproduce and accurately quantify their effects outside of a pooled experiment. Here, we describe a step-by-step protocol to rapidly assess the effects of individual sgRNAs from CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems. All steps, including cloning, lentivirus generation, cell transduction, and phenotypic readout, can be performed entirely in 96-well plates. The system is highly flexible in both cell type and selection system, requiring only that the phenotype(s) of interest be read out via flow cytometry. We expect that this protocol will provide researchers with a rapid way to sift through potential screening hits, and prioritize them for deeper analysis in more complex in vitro or even in vivo systems. Graphical abstract.

2.
Sci Transl Med ; 14(662): eabj8670, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36103516

ABSTRACT

The low-density lipoprotein receptor (LDLR) controls cellular delivery of cholesterol and clears LDL from the bloodstream, protecting against atherosclerotic heart disease, the leading cause of death in the United States. We therefore sought to identify regulators of the LDLR beyond the targets of current therapies and known causes of familial hypercholesterolemia. We found that cold shock domain-containing protein E1 (CSDE1) enhanced hepatic LDLR messenger RNA (mRNA) decay via its 3' untranslated region and regulated atherogenic lipoproteins in vivo. Using parallel phenotypic genome-wide CRISPR interference screens in a tissue culture model, we identified 40 specific regulators of the LDLR that were not previously identified by observational human genetic studies. Among these, we demonstrated that, in HepG2 cells, CSDE1 regulated the LDLR at least as strongly as statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors. In addition, we showed that hepatic gene silencing of Csde1 treated diet-induced dyslipidemia in mice to a similar degree as Pcsk9 silencing. These results suggest the therapeutic potential of targeting CSDE1 to manipulate the posttranscriptional regulation of the LDLR mRNA for the prevention of cardiovascular disease. Our approach of modeling a clinically relevant phenotype in a forward genetic screen, followed by mechanistic pharmacologic dissection and in vivo validation, may serve as a generalizable template for the identification of therapeutic targets in other human disease states.


Subject(s)
Cold-Shock Response , DNA-Binding Proteins/metabolism , Proprotein Convertase 9 , RNA-Binding Proteins/metabolism , Animals , Humans , Mice , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , RNA, Messenger/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Transcription, Genetic
4.
J Pediatr Hematol Oncol ; 43(6): e791-e794, 2021 08 01.
Article in English | MEDLINE | ID: mdl-32852399

ABSTRACT

Subcutaneous panniculitis-like T-cell lymphoma is a cutaneous lymphoma characterized by CD8+ T-cell infiltrate in the subcutis that is rare in children. Acute lymphoblastic lymphoma is the most common pediatric malignancy and often presents with fevers and pancytopenia. Herein, we report 2 pediatric patients presenting with subcutaneous panniculitis-like T-cell lymphoma and B-cell acute lymphoblastic lymphoma, distinct hematologic malignancies arising from different lymphoid lineages, with no identifiable germline cancer predisposition.


Subject(s)
Lymphoma, T-Cell/complications , Panniculitis/complications , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/complications , B-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Child, Preschool , Female , Humans , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/pathology , Male , Panniculitis/diagnosis , Panniculitis/pathology , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology
5.
Haematologica ; 103(7): 1218-1228, 2018 07.
Article in English | MEDLINE | ID: mdl-29622655

ABSTRACT

The myeloma bone marrow microenvironment promotes proliferation of malignant plasma cells and resistance to therapy. Activation of JAK/STAT signaling is thought to be a central component of these microenvironment-induced phenotypes. In a prior drug repurposing screen, we identified tofacitinib, a pan-JAK inhibitor Food and Drug Administration (FDA) approved for rheumatoid arthritis, as an agent that may reverse the tumor-stimulating effects of bone marrow mesenchymal stromal cells. Herein, we validated in vitro, in stromal-responsive human myeloma cell lines, and in vivo, in orthotopic disseminated xenograft models of myeloma, that tofacitinib showed efficacy in myeloma models. Furthermore, tofacitinib strongly synergized with venetoclax in coculture with bone marrow stromal cells but not in monoculture. Surprisingly, we found that ruxolitinib, an FDA approved agent targeting JAK1 and JAK2, did not lead to the same anti-myeloma effects. Combination with a novel irreversible JAK3-selective inhibitor also did not enhance ruxolitinib effects. Transcriptome analysis and unbiased phosphoproteomics revealed that bone marrow stromal cells stimulate a JAK/STAT-mediated proliferative program in myeloma cells, and tofacitinib reversed the large majority of these pro-growth signals. Taken together, our results suggest that tofacitinib reverses the growth-promoting effects of the tumor microenvironment. As tofacitinib is already FDA approved, these results can be rapidly translated into potential clinical benefits for myeloma patients.


Subject(s)
Bone Marrow/drug effects , Bone Marrow/pathology , Drug Repositioning , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Tumor Microenvironment/drug effects , Animals , Cell Communication , Disease Models, Animal , Humans , Janus Kinases/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Multiple Myeloma/metabolism , Phosphoproteins/metabolism , Piperidines/administration & dosage , Plasma Cells/metabolism , Plasma Cells/pathology , Protein Kinase Inhibitors/administration & dosage , Proteome , Proteomics/methods , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
6.
Sci Signal ; 10(510)2017 Dec 19.
Article in English | MEDLINE | ID: mdl-29259099

ABSTRACT

Interleukin-2 (IL-2) stimulates both activated CD4+ and CD8+ T cells to proliferate. IL-2 signals through an identical receptor complex and promotes the same dose-dependent phosphorylation of the canonical transcription factor STAT5 in both cell types. Despite this, CD8+ T cells enter the S phase earlier and proliferate to a greater extent than do CD4+ T cells in response to IL-2. We identified distinct IL-2 signaling dynamics in CD4+ and CD8+ T cells. In IL-2-stimulated CD8+ T cells, STAT5 phosphorylation increased rapidly and was sustained for 6 hours. In contrast, CD4+ T cells had a biphasic response, with maxima at 15 min and 2 to 4 hours after stimulation. Both cell types required vesicular trafficking, but only CD4+ T cells required new protein synthesis to maintain high phosphorylation of STAT5. Two subunits of the IL-2 receptor, IL-2Rß and IL-2Rγ, were twice as abundant in CD8+ T cells than in CD4+ T cells. Reduction of IL-2Rß abundance by 50% was sufficient to convert CD8+ T cells to a CD4+ T cell-like signaling pattern and delay S phase entry. These results suggest that the larger pool of IL-2Rß chains in CD8+ T cells is required to sustain IL-2 signaling and contributes to the quantitatively greater proliferative response to IL-2 relative to that of CD4+ T cells. This cell type-specific difference in IL-2Rß abundance appears to tune responses, potentially preventing extensive, autoimmune proliferation of CD4+ T cells, while still enabling sufficient proliferation of CD8+ T cells to control viral infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-2 Receptor beta Subunit/immunology , Interleukin-2/immunology , Animals , Autoimmunity/immunology , Cell Proliferation , Female , Immunity, Cellular/immunology , Interleukin Receptor Common gamma Subunit/immunology , Interleukin Receptor Common gamma Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Primary Cell Culture , Recombinant Proteins/metabolism , S Phase , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism
7.
Elife ; 52016 12 09.
Article in English | MEDLINE | ID: mdl-27935477

ABSTRACT

IgE can trigger potent allergic responses, yet the mechanisms regulating IgE production are poorly understood. Here we reveal that IgE+ B cells are constrained by chronic activity of the IgE B cell receptor (BCR). In the absence of cognate antigen, the IgE BCR promoted terminal differentiation of B cells into plasma cells (PCs) under cell culture conditions mimicking T cell help. This antigen-independent PC differentiation involved multiple IgE domains and Syk, CD19, BLNK, Btk, and IRF4. Disruption of BCR signaling in mice led to consistently exaggerated IgE+ germinal center (GC) B cell but variably increased PC responses. We were unable to confirm reports that the IgE BCR directly promoted intrinsic apoptosis. Instead, IgE+ GC B cells exhibited poor antigen presentation and prolonged cell cycles, suggesting reduced competition for T cell help. We propose that chronic BCR activity and access to T cell help play critical roles in regulating IgE responses.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin E/metabolism , Lymphocyte Activation , Receptors, IgE/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Mice, Inbred C57BL
8.
Nat Chem Biol ; 12(5): 373-9, 2016 May.
Article in English | MEDLINE | ID: mdl-27018889

ABSTRACT

To drive lymphocyte proliferation and differentiation, common γ-chain (γc) cytokine receptors require hours to days of sustained stimulation. JAK1 and JAK3 kinases are found together in all γc-receptor complexes, but how their respective catalytic activities contribute to signaling over time is not known. Here we dissect the temporal requirements for JAK3 kinase activity with a selective covalent inhibitor (JAK3i). By monitoring phosphorylation of the transcription factor STAT5 over 20 h in CD4(+) T cells stimulated with interleukin 2 (IL-2), we document a second wave of signaling that is much more sensitive to JAK3i than the first wave. Selective inhibition of this second wave is sufficient to block cyclin expression and entry to S phase. An inhibitor-resistant JAK3 mutant (C905S) rescued all effects of JAK3i in isolated T cells and in mice. Our chemical genetic toolkit elucidates a biphasic requirement for JAK3 kinase activity in IL-2-driven T cell proliferation and will find broad utility in studies of γc-receptor signaling.


Subject(s)
Janus Kinase 3/metabolism , Receptors, Interleukin-2/metabolism , Signal Transduction , Animals , Apoptosis , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/physiology , Cells, Cultured , Female , Gene Expression Regulation/physiology , Interferon-gamma , Interleukin-2/genetics , Interleukin-2/metabolism , Janus Kinase 3/genetics , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Receptors, Interleukin-2/genetics
9.
Elife ; 42015 Dec 10.
Article in English | MEDLINE | ID: mdl-26651998

ABSTRACT

Cyclic peptide natural products have evolved to exploit diverse protein targets, many of which control essential cellular processes. Inspired by a series of cyclic peptides with partially elucidated structures, we designed synthetic variants of ternatin, a cytotoxic and anti-adipogenic natural product whose molecular mode of action was unknown. The new ternatin variants are cytotoxic toward cancer cells, with up to 500-fold greater potency than ternatin itself. Using a ternatin photo-affinity probe, we identify the translation elongation factor-1A ternary complex (eEF1A·GTP·aminoacyl-tRNA) as a specific target and demonstrate competitive binding by the unrelated natural products, didemnin and cytotrienin. Mutations in domain III of eEF1A prevent ternatin binding and confer resistance to its cytotoxic effects, implicating the adjacent hydrophobic surface as a functional hot spot for eEF1A modulation. We conclude that the eukaryotic elongation factor-1A and its ternary complex with GTP and aminoacyl-tRNA are common targets for the evolution of cytotoxic natural products.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death , Peptide Elongation Factor 1/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Drug Resistance , Guanosine Triphosphate/metabolism , Humans , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutation , Peptide Elongation Factor 1/genetics , Peptides, Cyclic/chemical synthesis , Protein Binding , RNA, Transfer/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...