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 Stem Cell ; 31(2): 244-259.e10, 2024 02 01.
Article in English | MEDLINE | ID: mdl-38183977

ABSTRACT

The paradigmatic hematopoietic tree model is increasingly recognized to be limited, as it is based on heterogeneous populations largely defined by non-homeostatic assays testing cell fate potentials. Here, we combine persistent labeling with time-series single-cell RNA sequencing to build a real-time, quantitative model of in vivo tissue dynamics for murine bone marrow hematopoiesis. We couple cascading single-cell expression patterns with dynamic changes in differentiation and growth speeds. The resulting explicit linkage between molecular states and cellular behavior reveals widely varying self-renewal and differentiation properties across distinct lineages. Transplanted stem cells show strong acceleration of differentiation at specific stages of erythroid and neutrophil production, illustrating how the model can quantify the impact of perturbations. Our reconstruction of dynamic behavior from snapshot measurements is akin to how a kinetoscope allows sequential images to merge into a movie. We posit that this approach is generally applicable to understanding tissue-scale dynamics at high resolution.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Animals , Mice , Hematopoietic Stem Cells/metabolism , Hematopoiesis/genetics , Cell Differentiation
2.
Clin Cancer Res ; 30(7): 1338-1351, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37967136

ABSTRACT

PURPOSE: We evaluated the properties and activity of AZD9574, a blood-brain barrier (BBB) penetrant selective inhibitor of PARP1, and assessed its efficacy and safety alone and in combination with temozolomide (TMZ) in preclinical models. EXPERIMENTAL DESIGN: AZD9574 was interrogated in vitro for selectivity, PARylation inhibition, PARP-DNA trapping, the ability to cross the BBB, and the potential to inhibit cancer cell proliferation. In vivo efficacy was determined using subcutaneous as well as intracranial mouse xenograft models. Mouse, rat, and monkey were used to assess AZD9574 BBB penetration and rat models were used to evaluate potential hematotoxicity for AZD9574 monotherapy and the TMZ combination. RESULTS: AZD9574 demonstrated PARP1-selectivity in fluorescence anisotropy, PARylation, and PARP-DNA trapping assays and in vivo experiments demonstrated BBB penetration. AZD9574 showed potent single agent efficacy in preclinical models with homologous recombination repair deficiency in vitro and in vivo. In an O6-methylguanine-DNA methyltransferase (MGMT)-methylated orthotopic glioma model, AZD9574 in combination with TMZ was superior in extending the survival of tumor-bearing mice compared with TMZ alone. CONCLUSIONS: The combination of three key features-PARP1 selectivity, PARP1 trapping profile, and high central nervous system penetration in a single molecule-supports the development of AZD9574 as the best-in-class PARP inhibitor for the treatment of primary and secondary brain tumors. As documented by in vitro and in vivo studies, AZD9574 shows robust anticancer efficacy as a single agent as well as in combination with TMZ. AZD9574 is currently in a phase I trial (NCT05417594). See related commentary by Lynce and Lin, p. 1217.


Subject(s)
Brain Neoplasms , Glioma , Animals , Humans , Mice , Rats , Antineoplastic Agents, Alkylating/pharmacology , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor , DNA , Glioma/drug therapy , Glioma/pathology , O(6)-Methylguanine-DNA Methyltransferase/genetics , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
3.
Cell Mol Gastroenterol Hepatol ; 10(4): 797-810, 2020.
Article in English | MEDLINE | ID: mdl-32502530

ABSTRACT

BACKGROUND & AIMS: Intestinal crypts have a remarkable capacity to regenerate after injury from loss of crypt base columnar (CBC) stem cells. After injury, facultative stem cells (FSCs) are activated to replenish the epithelium and replace lost CBCs. Our aim was to assess the role of insulin-like growth factor-1 (IGF-1) to activate FSCs for crypt repair. METHODS: The intestinal regenerative response was measured after whole body 12-Gy γ-irradiation of adult mice. IGF-1 signaling or its downstream effector mammalian target of rapamycin complex 1 (mTORC1) was inhibited by administering BMS-754807 or rapamycin, respectively. Mice with inducible Rptor gene deletion were studied to test the role of mTORC1 signaling in the intestinal epithelium. FSC activation post-irradiation was measured by lineage tracing. RESULTS: We observed a coordinate increase in growth factor expression, including IGF-1, at 2 days post-irradiation, followed by a surge in mTORC1 activity during the regenerative phase of crypt repair at day 4. IGF-1 was localized to pericryptal mesenchymal cells, and IGF-1 receptor was broadly expressed in crypt progenitor cells. Inhibition of IGF-1 signaling via BMS-754807 treatment impaired crypt regeneration after 12-Gy irradiation, with no effect on homeostasis. Similarly, rapamycin inhibition of mTORC1 during the growth factor surge blunted the regenerative response. Analysis of Villin-CreERT2;Rptorfl/fl mice showed that epithelial mTORC1 signaling was essential for crypt regeneration. Lineage tracing from Bmi1-marked cells showed that rapamycin blocked FSC activation post-irradiation. CONCLUSIONS: Our study shows that IGF-1 signaling through mTORC1 drives crypt regeneration. We propose that IGF-1 release from pericryptal cells stimulates mTORC1 in FSCs to regenerate lost CBCs.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Intestinal Mucosa/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Radiation Injuries, Experimental/metabolism , Signal Transduction , Animals , Gamma Rays/adverse effects , Intestinal Mucosa/physiology , Intestinal Mucosa/radiation effects , Male , Mice , Regeneration
4.
Stem Cell Reports ; 15(1): 156-170, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32531190

ABSTRACT

Intestinal crypts have great capacity for repair and regeneration after intestinal stem cell (ISC) injury. Here, we define the cellular remodeling process resulting from ISC niche interruption by transient Notch pathway inhibition in adult mice. Although ISCs were retained, lineage tracing demonstrated a marked reduction in ISC function after Notch disruption. Surprisingly, Notch ligand-expressing Paneth cells were rapidly lost by apoptotic cell death. The ISC-Paneth cell changes were followed by a regenerative response, characterized by expansion of cells expressing Notch ligands Dll1 and Dll4, enhanced Notch signaling, and a proliferative surge. Lineage tracing and organoid studies showed that Dll1-expressing cells were activated to function as multipotential progenitors, generating both absorptive and secretory cells and replenishing the vacant Paneth cell pool. Our analysis uncovered a dynamic, multicellular remodeling response to acute Notch inhibition to repair the niche and restore homeostasis. Notably, this crypt regenerative response did not require ISC loss.


Subject(s)
Intestines/cytology , Intestines/physiology , Receptors, Notch/metabolism , Regeneration , Stem Cell Niche , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cell Proliferation , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice, Transgenic , Models, Biological , Stem Cells/cytology , Stem Cells/metabolism
5.
Stem Cell Reports ; 11(6): 1337-1346, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30449703

ABSTRACT

With the tamoxifen-inducible CreERT2 system, genetic recombination can be temporally controlled in a cell-type-specific manner in intact animals, permitting dissection of the molecular underpinnings of mammalian physiology. Here we present a significant drawback to CreERT2 technology for analysis of intestinal stem cells. Using the intestine-specific Villin-CreERT2 mouse strain, we observed delayed intestinal regeneration post irradiation. Villin-CreERT2 activation was associated with DNA damage and cryptic loxP site cleavage. Analysis of stem cell-specific CreERT2 strains showed that the genome toxicity impairs function of crypt base columnar stem cells, resulting in loss of organoid initiating activity. Importantly, the stem cell impairment is short-lived, with return to normal by 7 days post tamoxifen treatment. Our findings demonstrate that mouse genetic experiments that utilize CreERT2 should consider the confounding effects of enhanced stem cell sensitivity to genome toxicity resulting from CreERT2 activation.


Subject(s)
Genome , Integrases/metabolism , Intestines/cytology , Receptors, Estrogen/metabolism , Stem Cells/metabolism , Animals , DNA Cleavage/drug effects , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Organoids/drug effects , Organoids/metabolism , Regeneration/drug effects , Stem Cells/cytology , Stem Cells/drug effects , Tamoxifen/pharmacology
6.
Blood ; 124(26): 3947-55, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25361812

ABSTRACT

Oncogenic NRAS mutations are highly prevalent in acute myeloid leukemia (AML). Genetic analysis supports the hypothesis that NRAS mutations cooperate with antecedent molecular lesions in leukemogenesis, but have limited independent prognostic significance. Using short hairpin RNA-mediated knockdown in human cell lines and primary mouse leukemias, we show that AML cells with NRAS/Nras mutations are dependent on continued oncogene expression in vitro and in vivo. Using the Mx1-Cre transgene to inactivate a conditional mutant Nras allele, we analyzed hematopoiesis and hematopoietic stem and progenitor cells (HSPCs) under normal and stressed conditions and found that HSPCs lacking Nras expression are functionally equivalent to normal HSPCs in the adult mouse. Treating recipient mice transplanted with primary Nras(G12D) AMLs with 2 potent allosteric mitogen-activated protein kinase kinase (MEK) inhibitors (PD0325901 or trametinib/GlaxoSmithKline 1120212) significantly prolonged survival and reduced proliferation but did not induce apoptosis, promote differentiation, or drive clonal evolution. The phosphatidylinositol 3-kinase inhibitor GDC-0941 was ineffective as a single agent and did not augment the activity of PD0325901. All mice ultimately succumbed to progressive leukemia. Together, these data validate oncogenic N-Ras signaling as a therapeutic target in AML and support testing combination regimens that include MEK inhibitors.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Kinase Kinases/metabolism , Monomeric GTP-Binding Proteins/metabolism , Mutation , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Cell Survival , Genes, ras , Hematopoietic Stem Cells/cytology , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Monomeric GTP-Binding Proteins/genetics , Protein Kinase Inhibitors/chemistry , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cells/cytology , Transgenes
7.
Mol Endocrinol ; 28(9): 1471-86, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25029241

ABSTRACT

Wnt/ß-catenin (ßcat) signaling is critical for adrenal homeostasis. To elucidate how Wnt/ßcat signaling elicits homeostatic maintenance of the adrenal cortex, we characterized the identity of the adrenocortical Wnt-responsive population. We find that Wnt-responsive cells consist of sonic hedgehog (Shh)-producing adrenocortical progenitors and differentiated, steroidogenic cells of the zona glomerulosa, but not the zona fasciculata and rarely cells that are actively proliferating. To determine potential direct inhibitory effects of ßcat signaling on zona fasciculata-associated steroidogenesis, we used the mouse ATCL7 adrenocortical cell line that serves as a model system of glucocorticoid-producing fasciculata cells. Stimulation of ßcat signaling caused decreased corticosterone release consistent with the observed reduced transcription of steroidogenic genes Cyp11a1, Cyp11b1, Star, and Mc2r. Decreased steroidogenic gene expression was correlated with diminished steroidogenic factor 1 (Sf1; Nr5a1) expression and occupancy on steroidogenic promoters. Additionally, ßcat signaling suppressed the ability of Sf1 to transactivate steroidogenic promoters independent of changes in Sf1 expression level. To investigate Sf1-independent effects of ßcat on steroidogenesis, we used Affymetrix gene expression profiling of Wnt-responsive cells in vivo and in vitro. One candidate gene identified, Ccdc80, encodes a secreted protein with unknown signaling mechanisms. We report that Ccdc80 is a novel ßcat-regulated gene in adrenocortical cells. Treatment of adrenocortical cells with media containing secreted Ccdc80 partially phenocopies ßcat-induced suppression of steroidogenesis, albeit through an Sf1-independent mechanism. This study reveals multiple mechanisms of ßcat-mediated suppression of steroidogenesis and suggests that Wnt/ßcat signaling may regulate adrenal homeostasis by inhibiting fasciculata differentiation and promoting the undifferentiated state of progenitor cells.


Subject(s)
Adrenal Cortex/metabolism , Steroids/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Adrenal Cortex Hormones/metabolism , Animals , Cell Differentiation , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Extracellular Matrix Proteins , Gene Expression Profiling , Gene Expression Regulation , Glucocorticoids/metabolism , Glycoproteins/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Homeostasis , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Mutagenesis , Oligonucleotide Array Sequence Analysis , RNA Splicing Factors , Subcellular Fractions , Transcription Factors/metabolism , Wnt Proteins/metabolism
8.
Nature ; 504(7478): 143-147, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24284627

ABSTRACT

'Pre-leukaemic' mutations are thought to promote clonal expansion of haematopoietic stem cells (HSCs) by increasing self-renewal and competitiveness; however, mutations that increase HSC proliferation tend to reduce competitiveness and self-renewal potential, raising the question of how a mutant HSC can sustainably outcompete wild-type HSCs. Activating mutations in NRAS are prevalent in human myeloproliferative neoplasms and leukaemia. Here we show that a single allele of oncogenic Nras(G12D) increases HSC proliferation but also increases reconstituting and self-renewal potential upon serial transplantation in irradiated mice, all prior to leukaemia initiation. Nras(G12D) also confers long-term self-renewal potential to multipotent progenitors. To explore the mechanism by which Nras(G12D) promotes HSC proliferation and self-renewal, we assessed cell-cycle kinetics using H2B-GFP label retention and 5-bromodeoxyuridine (BrdU) incorporation. Nras(G12D) had a bimodal effect on HSCs, increasing the frequency with which some HSCs divide and reducing the frequency with which others divide. This mirrored bimodal effects on reconstituting potential, as rarely dividing Nras(G12D) HSCs outcompeted wild-type HSCs, whereas frequently dividing Nras(G12D) HSCs did not. Nras(G12D) caused these effects by promoting STAT5 signalling, inducing different transcriptional responses in different subsets of HSCs. One signal can therefore increase HSC proliferation, competitiveness and self-renewal through bimodal effects on HSC gene expression, cycling and reconstituting potential.


Subject(s)
Genes, ras/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Bone Marrow Transplantation , Cell Proliferation , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Time Factors
9.
Cancer Biol Ther ; 12(8): 718-26, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21775822

ABSTRACT

Activation of the epidermal growth factor receptor (EGFR) regulates cellular proliferation, survival, and migration of breast cancer cells. In particular, EGFR recruits signaling proteins to the cell membrane leading to their phosphorylation and activation. However, EGFR also localizes to other cellular structures, including endosomes, mitochondrion, and nuclei. Recently, we demonstrated that lipid raft localization of EGFR in triple-negative breast cancer cell lines promotes EGFR protein-dependent, EGFR kinase-independent activation of Akt. Here, we further define the mechanism by which lipid rafts regulate EGFR signaling to Akt. Specifically, we show that the non-receptor tyrosine kinase c-Src co-localizes and co-associates with EGFR and lipid rafts. Breast cancer cells resistant to treatment with EGFR inhibitors, were also resistant to treatment with Src family kinase (SFK) inhibitors; however, the combination of EGFR and SFK inhibitors synergistically decreases cell viability. We found that this decrease in cell viability observed with EGFR and SFK inhibitor co-treatment correlates with loss of Akt phosphorylation. In addition, we found that in breast cancer cell lines with EGFR and c-Src co-localized to lipid rafts, phospho-inositide 3 kinase (PI3K) was also associated with lipid rafts. Together, the data herein suggest that lipid rafts provide a platform for the interaction of EGFR, c-Src, and PI3K, leading to activation of cellular survival signaling in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Membrane Microdomains/metabolism , src-Family Kinases/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Female , Humans , Oncogene Protein v-akt/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , src-Family Kinases/antagonists & inhibitors
10.
J Cell Physiol ; 226(9): 2316-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21660955

ABSTRACT

Epidermal growth factor receptor (EGFR) is overexpressed in many cancer types including ~30% of breast cancers. Several small molecule tyrosine kinase inhibitors (TKIs) targeting EGFR have shown clinical efficacy in lung and colon cancers, but no benefit has been noted in breast cancer. Thirteen EGFR expressing breast cancer cell lines were analyzed for response to EGFR TKIs. Seven were found to be EGFR TKI resistant; while shRNA knockdown of EGFR determined that four of these cell lines retained the requirement of EGFR protein expression for growth. Interestingly, EGFR localized to plasma membrane lipid rafts in all four of these EGFR TKI-resistant cell lines, as determined by biochemical raft isolation and immunofluorescence. When lipid rafts were depleted of cholesterol using lovastatin, all four cell lines were sensitized to EGFR TKIs. In fact, the effects of the cholesterol biosynthesis inhibitors and gefitinib were synergistic. While gefitinib effectively abrogated phosphorylation of Akt- and mitogen-activated protein kinase in an EGFR TKI-sensitive cell line, phosphorylation of Akt persisted in two EGFR TKI-resistant cell lines, however, this phosphorylation was abrogated by lovastatin treatment. Thus, we have shown that lipid raft localization of EGFR correlates with resistance to EGFR TKI-induced growth inhibition and pharmacological depletion of cholesterol from lipid rafts decreases this resistance in breast cancer cell lines. Furthermore, we have presented evidence to suggest that when EGFR localizes to lipid rafts, these rafts provide a platform to facilitate activation of Akt signaling in the absence of EGFR kinase activity.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , ErbB Receptors/antagonists & inhibitors , Membrane Microdomains/enzymology , Protein Kinase Inhibitors/pharmacology , Quinazolines/therapeutic use , Atorvastatin , Benzylamines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cholesterol/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , ErbB Receptors/metabolism , Female , Gefitinib , Heptanoic Acids/pharmacology , Humans , Lovastatin/pharmacology , Membrane Microdomains/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrroles/pharmacology , Quinazolines/pharmacology , Thiophenes/pharmacology , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...