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










Publication year range
1.
Cell Rep ; 43(5): 114139, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38652658

ABSTRACT

Glioblastoma (GBM) is a deadly brain tumor, and the kinesin motor KIF11 is an attractive therapeutic target with roles in proliferation and invasion. Resistance to KIF11 inhibitors, which has mainly been studied in animal models, presents significant challenges. We use lineage-tracing barcodes and single-cell RNA sequencing to analyze resistance in patient-derived GBM neurospheres treated with ispinesib, a potent KIF11 inhibitor. Similar to GBM progression in patients, untreated cells lose their neural lineage identity and become mesenchymal, which is associated with poor prognosis. Conversely, cells subjected to long-term ispinesib treatment exhibit a proneural phenotype. We generate patient-derived xenografts and show that ispinesib-resistant cells form less aggressive tumors in vivo, even in the absence of drug. Moreover, treatment of human ex vivo GBM slices with ispinesib demonstrates phenotypic alignment with in vitro responses, underscoring the clinical relevance of our findings. Finally, using retrospective lineage tracing, we identify drugs that are synergistic with ispinesib.

2.
bioRxiv ; 2023 Sep 12.
Article in English | MEDLINE | ID: mdl-37745469

ABSTRACT

Glioblastoma (GBM) is a deadly brain tumor, and the kinesin motor KIF11 is an attractive therapeutic target because of its dual roles in proliferation and invasion. The clinical utility of KIF11 inhibitors has been limited by drug resistance, which has mainly been studied in animal models. We used multiplexed lineage tracing barcodes and scRNA-seq to analyze drug resistance time courses for patient-derived GBM neurospheres treated with ispinesib, a potent KIF11 inhibitor. Similar to GBM progression in patients, untreated cells lost their neural lineage identity and transitioned to a mesenchymal phenotype, which is associated with poor prognosis. In contrast, cells subjected to long-term ispinesib treatment exhibited a proneural phenotype. We generated patient-derived xenografts to show that ispinesib-resistant cells form less aggressive tumors in vivo, even in the absence of drug. Finally, we used lineage barcodes to nominate drug combination targets by retrospective analysis of ispinesib-resistant clones in the drug-naïve setting and identified drugs that are synergistic with ispinesib.

3.
Genetics ; 219(2)2021 10 02.
Article in English | MEDLINE | ID: mdl-34173831

ABSTRACT

Filamins are highly conserved actin-crosslinking proteins that regulate organization of the actin cytoskeleton. As key components of versatile signaling scaffolds, filamins are implicated in developmental anomalies and cancer. Multiple isoforms of filamins exist, raising the possibility of distinct functions for each isoform during development and in disease. Here, we provide an initial characterization of jitterbug (jbug), which encodes one of the two filamin-type proteins in Drosophila. We generate Jbug antiserum that recognizes all of the spliced forms and reveals differential expression of different Jbug isoforms during development, and a significant maternal contribution of Jbug protein. To reveal the function of Jbug isoforms, we create new genetic tools, including a null allele that deletes all isoforms, hypomorphic alleles that affect only a subset, and UAS lines for Gal4-driven expression of the major isoforms. Using these tools, we demonstrate that Jbug is required for viability and that specific isoforms are required in the formation of actin-rich protrusions including thoracic bristles in adults and ventral denticles in the embryo. We also show that specific isoforms of Jbug show differential localization within epithelia and that maternal and zygotic loss of jbug disrupts Crumbs (Crb) localization in several epithelial cell types.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Gene Expression Regulation, Developmental , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Epithelial Cells/cytology , Epithelial Cells/metabolism , Morphogenesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
4.
Nat Commun ; 10(1): 4706, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624246

ABSTRACT

Human T cells coordinate adaptive immunity in diverse anatomic compartments through production of cytokines and effector molecules, but it is unclear how tissue site influences T cell persistence and function. Here, we use single cell RNA-sequencing (scRNA-seq) to define the heterogeneity of human T cells isolated from lungs, lymph nodes, bone marrow and blood, and their functional responses following stimulation. Through analysis of >50,000 resting and activated T cells, we reveal tissue T cell signatures in mucosal and lymphoid sites, and lineage-specific activation states across all sites including distinct effector states for CD8+ T cells and an interferon-response state for CD4+ T cells. Comparing scRNA-seq profiles of tumor-associated T cells to our dataset reveals predominant activated CD8+ compared to CD4+ T cell states within multiple tumor types. Our results therefore establish a high dimensional reference map of human T cell activation in health for analyzing T cells in disease.


Subject(s)
Lung/metabolism , Lymph Nodes/metabolism , Neoplasms/genetics , Single-Cell Analysis/methods , T-Lymphocytes/metabolism , Transcriptome/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Lymph Nodes/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mucous Membrane/immunology , Mucous Membrane/metabolism , Neoplasms/pathology , T-Lymphocytes/immunology
5.
Mol Syst Biol ; 15(2): e8557, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30796088

ABSTRACT

Common approaches to gene signature discovery in single-cell RNA-sequencing (scRNA-seq) depend upon predefined structures like clusters or pseudo-temporal order, require prior normalization, or do not account for the sparsity of single-cell data. We present single-cell hierarchical Poisson factorization (scHPF), a Bayesian factorization method that adapts hierarchical Poisson factorization (Gopalan et al, 2015, Proceedings of the 31st Conference on Uncertainty in Artificial Intelligence, 326) for de novo discovery of both continuous and discrete expression patterns from scRNA-seq. scHPF does not require prior normalization and captures statistical properties of single-cell data better than other methods in benchmark datasets. Applied to scRNA-seq of the core and margin of a high-grade glioma, scHPF uncovers marked differences in the abundance of glioma subpopulations across tumor regions and regionally associated expression biases within glioma subpopulations. scHFP revealed an expression signature that was spatially biased toward the glioma-infiltrated margins and associated with inferior survival in glioblastoma.


Subject(s)
Glioma/genetics , High-Throughput Nucleotide Sequencing/methods , Single-Cell Analysis , Transcriptome/genetics , Bayes Theorem , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Humans , Poisson Distribution
6.
Semin Nephrol ; 38(1): 40-51, 2018 01.
Article in English | MEDLINE | ID: mdl-29291761

ABSTRACT

Acute kidney injury (AKI) currently is diagnosed by a temporal trend of a single blood analyte: serum creatinine. This measurement is neither sensitive nor specific to kidney injury or its protean forms. Newer biomarkers, neutrophil gelatinase-associated lipocalin (NGAL, Lipocalin 2, Siderocalin), or kidney injury molecule-1 (KIM-1, Hepatitis A Virus Cellular Receptor 1), accelerate the diagnosis of AKI as well as prospectively distinguish rapidly reversible from prolonged causes of serum creatinine increase. Nonetheless, these biomarkers lack the capacity to subfractionate AKI further (eg, sepsis versus ischemia versus nephrotoxicity from medications, enzymes, or metals) or inform us about the primary and secondary sites of injury. It also is unknown whether all nephrons are injured in AKI, whether all cells in a nephron are affected, and whether injury responses can be stimulus-specific or cell type-specific or both. In this review, we summarize fully agnostic tissue interrogation approaches that may help to redefine AKI in cellular and molecular terms, including single-cell and single-nuclei RNA sequencing technology. These approaches will empower a shift in the current paradigm of AKI diagnosis, classification, and staging, and provide the renal community with a significant advance toward precision medicine in the analysis AKI.


Subject(s)
Acute Kidney Injury/diagnosis , Precision Medicine , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Creatinine/blood , Humans , Quantitative Trait Loci , Sequence Analysis, RNA
7.
Cell Rep ; 13(10): 2174-88, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26628373

ABSTRACT

Multiple inositol polyphosphate phosphatase (Mipp), a highly conserved but poorly understood histidine phosphatase, dephosphorylates higher-order IPs (IP4-IP6) to IP3. To gain insight into the biological roles of these enzymes, we have characterized Drosophila mipp1. mipp1 is dynamically expressed in the embryonic trachea, specifically in the leading cells of migrating branches at late stages, where Mipp1 localizes to the plasma membrane and filopodia. FGF signaling activates mipp1 expression in these cells, where extensive filopodia form to drive migration and elongation by cell intercalation. We show that Mipp1 facilitates formation and/or stabilization of filopodia in leading cells through its extracellular activity. mipp1 loss decreases filopodia number, whereas mipp1 overexpression increases filopodia number in a phosphatase-activity-dependent manner. Importantly, expression of Mipp1 gives cells a migratory advantage for the lead position in elongating tracheal branches. Altogether, these findings suggest that extracellular pools of inositol polyphosphates affect cell behavior during development.


Subject(s)
Cell Movement , Drosophila/embryology , Drosophila/metabolism , Epithelial Cells/metabolism , Phosphoric Monoester Hydrolases/metabolism , Trachea/embryology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Blotting, Western , Cell Movement/physiology , Embryo, Nonmammalian , Extracellular Space/metabolism , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Protein Conformation , RNA, Small Interfering , Transfection
8.
PLoS One ; 8(10): e77618, 2013.
Article in English | MEDLINE | ID: mdl-24204897

ABSTRACT

Core components of the secretory pathway have largely been identified and studied in single cell systems such as the budding yeast S. cerevisiae or in mammalian tissue culture. These studies provide details on the molecular functions of the secretory machinery; they fail, however, to provide insight into the role of these proteins in the context of specialized organs of higher eukaryotes. Here, we identify and characterize the first loss-of-function mutations in a KDEL receptor gene from higher eukaryotes. Transcripts from the Drosophila KDEL receptor gene KdelR - formerly known as dmErd2 - are provided maternally and, at later stages, are at elevated levels in several embryonic cell types, including the salivary gland secretory cells, the fat body and the epidermis. We show that, unlike Saccharomyces cerevisiae Erd2 mutants, which are viable, KdelR mutations are early larval lethal, with homozygous mutant animals dying as first instar larvae. KdelR mutants have larval cuticle defects similar to those observed with loss-of-function mutations in other core secretory pathway genes and with mutations in CrebA, which encodes a bZip transcription factor that coordinately upregulates secretory pathway component genes in specialized secretory cell types. Using the salivary gland, we demonstrate a requirement for KdelR in maintaining the ER pool of a subset of soluble resident ER proteins. These studies underscore the utility of the Drosophila salivary gland as a unique system for studying the molecular machinery of the secretory pathway in vivo in a complex eukaryote.


Subject(s)
Drosophila/genetics , Drosophila/metabolism , Epidermis/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Salivary Glands/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Cyclic AMP Response Element-Binding Protein A/genetics , Cyclic AMP Response Element-Binding Protein A/metabolism , Drosophila/embryology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Epidermis/embryology , Genes, Insect/genetics , Mutation/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Salivary Glands/embryology , Secretory Pathway/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Med Chem ; 56(10): 3996-4016, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23634668

ABSTRACT

Methionine aminopeptidases (MetAPs), which remove the initiator methionine from nascent peptides, are essential in all organisms. While MetAP2 has been demonstrated to be a therapeutic target for inhibiting angiogenesis in mammals, MetAP1 seems to be vital for cell proliferation. Our earlier efforts identified two structural classes of human MetAP1 (HsMetAP1)-selective inhibitors (1-4), but all of them failed to inhibit cellular HsMetAP1. Using Mn(II) or Zn(II) to activate HsMetAP1, we found that 1-4 could only effectively inhibit purified HsMetAP1 in the presence of physiologically unachievable concentrations of Co(II). In an effort to seek Co(II)-independent inhibitors, a novel structural class containing a 2-(pyridin-2-yl)quinazoline core has been discovered. Many compounds in this class potently and selectively inhibited HsMetAP1 without Co(II). Subsequently, we demonstrated that 11j, an auxiliary metal-dependent inhibitor, effectively inhibited HsMetAP1 in primary cells. This is the first report that an HsMetAP1-selective inhibitor is effective against its target in cells.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Protease Inhibitors/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Aminopeptidases/biosynthesis , Animals , Cell Proliferation/drug effects , Chelating Agents/pharmacology , Chromatography, Thin Layer , Cobalt/pharmacology , Crystallography, X-Ray , Down-Regulation/drug effects , Enzyme Activation/drug effects , HeLa Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Indicators and Reagents , Manganese/pharmacology , Metals/chemistry , Methionine/metabolism , Mice , Models, Molecular , Pyridines/chemistry , Quinazolines/chemistry , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Thymidine/metabolism , Transfection , Zinc/pharmacology
10.
Org Lett ; 11(9): 1971-4, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19331423

ABSTRACT

HDAC inhibitor Spiruchostatin A was synthesized via a route that differs significantly from previously reported routes. The key step involves a latent thioester that initiates a chemoselective transformation similar to native chemical ligation to form the macrocyclic alanine-cysteine amide bond. The easily prepared latent thioester--the first such moiety reported in enantiomerically pure form--is designed with a pendant carboxylic acid to serve as a solid-phase linker for the synthesis of cyclic, cysteine-containing, peptidic materials.


Subject(s)
Histone Deacetylase Inhibitors , Peptides, Cyclic/chemical synthesis , Cyclization , Molecular Structure , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Stereoisomerism
SELECTION OF CITATIONS
SEARCH DETAIL
...