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










Publication year range
1.
Blood ; 142(24): 2079-2091, 2023 12 14.
Article in English | MEDLINE | ID: mdl-37595362

ABSTRACT

PPM1D encodes a phosphatase that is recurrently activated across cancer, most notably in therapy-related myeloid neoplasms. However, the function of PPM1D in hematopoiesis and its contribution to tumor cell growth remain incompletely understood. Using conditional mouse models, we uncover a central role for Ppm1d in hematopoiesis and validate its potential as a therapeutic target. We find that Ppm1d regulates the competitive fitness and self-renewal of hematopoietic stem cells (HSCs) with and without exogenous genotoxic stresses. We also show that although Ppm1d activation confers cellular resistance to cytotoxic therapy, it does so to a lesser degree than p53 loss, informing the clonal competition phenotypes often observed in human studies. Notably, loss of Ppm1d sensitizes leukemias to cytotoxic therapies in vitro and in vivo, even in the absence of a Ppm1d mutation. Vulnerability to PPM1D inhibition is observed across many cancer types and dependent on p53 activity. Importantly, organism-wide loss of Ppm1d in adult mice is well tolerated, supporting the tolerability of pharmacologically targeting PPM1D. Our data link PPM1D gain-of-function mutations to the clonal expansion of HSCs, inform human genetic observations, and support the therapeutic targeting of PPM1D in cancer.


Subject(s)
DNA Damage , Tumor Suppressor Protein p53 , Adult , Humans , Animals , Mice , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Protein Phosphatase 2C , Mutation , Phosphoric Monoester Hydrolases/genetics , Cell Cycle
3.
Cell Metab ; 33(5): 1027-1041.e8, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33770508

ABSTRACT

Mitochondria have an independent genome (mtDNA) and protein synthesis machinery that coordinately activate for mitochondrial generation. Here, we report that the Krebs cycle intermediate fumarate links metabolism to mitobiogenesis through binding to malic enzyme 2 (ME2). Mechanistically, fumarate binds ME2 with two complementary consequences. First, promoting the formation of ME2 dimers, which activate deoxyuridine 5'-triphosphate nucleotidohydrolase (DUT). DUT fosters thymidine generation and an increase of mtDNA. Second, fumarate-induced ME2 dimers abrogate ME2 monomer binding to mitochondrial ribosome protein L45, freeing it for mitoribosome assembly and mtDNA-encoded protein production. Methylation of the ME2-fumarate binding site by protein arginine methyltransferase-1 inhibits fumarate signaling to constrain mitobiogenesis. Notably, acute myeloid leukemia is highly dependent on mitochondrial function and is sensitive to targeting of the fumarate-ME2 axis. Therefore, mitobiogenesis can be manipulated in normal and malignant cells through ME2, an unanticipated governor of mitochondrial biomass production that senses nutrient availability through fumarate.


Subject(s)
Fumarates/metabolism , Malate Dehydrogenase/metabolism , Mitochondria/metabolism , Animals , Cell Line , Citric Acid Cycle , DNA, Mitochondrial/metabolism , Dimerization , Humans , Leukemia/pathology , Leukemia/veterinary , Malate Dehydrogenase/antagonists & inhibitors , Malate Dehydrogenase/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mitochondria/genetics , Protein Binding , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Pyrophosphatases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Ribosomal Proteins/metabolism , Thymidine/metabolism
4.
Cell Metab ; 32(3): 391-403.e6, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32763164

ABSTRACT

Cancer relapse begins when malignant cells pass through the extreme metabolic bottleneck of stress from chemotherapy and the byproducts of the massive cell death in the surrounding region. In acute myeloid leukemia, complete remissions are common, but few are cured. We tracked leukemia cells in vivo, defined the moment of maximal response following chemotherapy, captured persisting cells, and conducted unbiased metabolomics, revealing a metabolite profile distinct from the pre-chemo growth or post-chemo relapse phase. Persisting cells used glutamine in a distinctive manner, preferentially fueling pyrimidine and glutathione generation, but not the mitochondrial tricarboxylic acid cycle. Notably, malignant cell pyrimidine synthesis also required aspartate provided by specific bone marrow stromal cells. Blunting glutamine metabolism or pyrimidine synthesis selected against residual leukemia-initiating cells and improved survival in leukemia mouse models and patient-derived xenografts. We propose that timed cell-intrinsic or niche-focused metabolic disruption can exploit a transient vulnerability and induce metabolic collapse in cancer cells to overcome chemoresistance.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Animals , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Inbred NOD
5.
Blood ; 136(11): 1303-1316, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32458004

ABSTRACT

Metabolic alterations in cancer represent convergent effects of oncogenic mutations. We hypothesized that a metabolism-restricted genetic screen, comparing normal primary mouse hematopoietic cells and their malignant counterparts in an ex vivo system mimicking the bone marrow microenvironment, would define distinctive vulnerabilities in acute myeloid leukemia (AML). Leukemic cells, but not their normal myeloid counterparts, depended on the aldehyde dehydrogenase 3a2 (Aldh3a2) enzyme that oxidizes long-chain aliphatic aldehydes to prevent cellular oxidative damage. Aldehydes are by-products of increased oxidative phosphorylation and nucleotide synthesis in cancer and are generated from lipid peroxides underlying the non-caspase-dependent form of cell death, ferroptosis. Leukemic cell dependence on Aldh3a2 was seen across multiple mouse and human myeloid leukemias. Aldh3a2 inhibition was synthetically lethal with glutathione peroxidase-4 (GPX4) inhibition; GPX4 inhibition is a known trigger of ferroptosis that by itself minimally affects AML cells. Inhibiting Aldh3a2 provides a therapeutic opportunity and a unique synthetic lethality to exploit the distinctive metabolic state of malignant cells.


Subject(s)
Aldehyde Oxidoreductases/physiology , Carbolines/pharmacology , Cyclohexylamines/pharmacology , Ferroptosis/drug effects , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/enzymology , Neoplasm Proteins/physiology , Phenylenediamines/pharmacology , Aldehyde Oxidoreductases/genetics , Aldehydes/pharmacology , Animals , Cell Line, Tumor , Cytarabine/administration & dosage , Doxorubicin/administration & dosage , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Lipid Peroxidation , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Lymphoid Leukemia Protein/physiology , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Oleic Acid/pharmacology , Oncogene Proteins, Fusion/physiology , Oxidation-Reduction , Oxidative Stress , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/physiology
6.
Nature ; 579(7797): 111-117, 2020 03.
Article in English | MEDLINE | ID: mdl-32103177

ABSTRACT

The avascular nature of cartilage makes it a unique tissue1-4, but whether and how the absence of nutrient supply regulates chondrogenesis remain unknown. Here we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate forkhead box O (FOXO) transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing oxidation of fatty acids, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FOXO transcription factors during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.


Subject(s)
Bone and Bones/cytology , Cellular Microenvironment , Chondrogenesis , Lipid Metabolism , SOX9 Transcription Factor/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Bone and Bones/blood supply , Chondrocytes/cytology , Chondrocytes/metabolism , Fatty Acids/metabolism , Female , Food Deprivation , Forkhead Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Osteogenesis , Oxidation-Reduction , SOX9 Transcription Factor/genetics , Signal Transduction , Wound Healing
7.
Nat Biomed Eng ; 4(1): 40-51, 2020 01.
Article in English | MEDLINE | ID: mdl-31937942

ABSTRACT

Acute myeloid leukaemia (AML) is a malignancy of haematopoietic origin that has limited therapeutic options. The standard-of-care cytoreductive chemotherapy depletes AML cells to induce remission, but is infrequently curative. An immunosuppressive AML microenvironment in the bone marrow and the paucity of suitable immunotherapy targets limit the induction of effective immune responses. Here, in mouse models of AML, we show that a macroporous-biomaterial vaccine that delivers the cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF), the Toll-like-receptor-9 agonist cytosine-guanosine oligodeoxynucleotide and one or multiple leukaemia antigens (in the form of a defined peptide antigen, cell lysates or antigens sourced from AML cells recruited in vivo) induces local immune-cell infiltration and activated dendritic cells, evoking a potent anti-AML response. The biomaterial-based vaccine prevented the engraftment of AML cells when administered as a prophylactic and when combined with chemotherapy, and eradicated established AML even in the absence of a defined vaccine antigen. Biomaterial-based AML vaccination can induce potent immune responses, deplete AML cells and prevent disease relapse.


Subject(s)
Cancer Vaccines/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Animals , Biocompatible Materials , Bone Marrow/drug effects , Bone Marrow/immunology , Disease Models, Animal , Drug Development , Female , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Leukemia, Myeloid, Acute/immunology , Mice, Inbred C57BL , Toll-Like Receptor 9/agonists
8.
Nat Biotechnol ; 37(3): 293-302, 2019 03.
Article in English | MEDLINE | ID: mdl-30742125

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for multiple disorders, but deficiency and dysregulation of T cells limit its utility. Here we report a biomaterial-based scaffold that mimics features of T cell lymphopoiesis in the bone marrow. The bone marrow cryogel (BMC) releases bone morphogenetic protein-2 to recruit stromal cells and presents the Notch ligand Delta-like ligand-4 to facilitate T cell lineage specification of mouse and human hematopoietic progenitor cells. BMCs subcutaneously injected in mice at the time of HSCT enhanced T cell progenitor seeding of the thymus, T cell neogenesis and diversification of the T cell receptor repertoire. Peripheral T cell reconstitution increased ~6-fold in mouse HSCT and ~2-fold in human xenogeneic HSCT. Furthermore, BMCs promoted donor CD4+ regulatory T cell generation and improved survival after allogeneic HSCT. In comparison to adoptive transfer of T cell progenitors, BMCs increased donor chimerism, T cell generation and antigen-specific T cell responses to vaccination. BMCs may provide an off-the-shelf approach for enhancing T cell regeneration and mitigating graft-versus-host disease in HSCT.


Subject(s)
Bone Marrow Transplantation , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , T-Lymphocytes, Regulatory/immunology , Tissue Scaffolds , Adoptive Transfer/methods , Animals , Bone Marrow , Chimerism , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Mice , T-Lymphocytes, Regulatory/cytology , Transplantation, Heterologous/methods , Transplantation, Homologous
9.
J Asthma Allergy ; 11: 159-171, 2018.
Article in English | MEDLINE | ID: mdl-30122959

ABSTRACT

BACKGROUND: Purinergic receptors control cell proliferation, apoptosis, migration, inflammation, and cytokine secretion. Increased expression of specific purinergic receptors is reported in asthma. The role of purinergic P2Y6 receptors (P2Y6R) in asthma is controversial. HYPOTHESIS: P2Y6R activation in asthma improves pulmonary function and reduces inflammation and smooth muscle amount. METHODS: Female mice (C57/BL6, age 30 days) were randomly assigned to receive intranasal house dust mite (HDM) antigen (40 or 80 µg) or saline, 5 days/week, for 6 weeks. Randomly selected subgroups received intraperitoneal P2Y6R agonist prodrug (GC021109; 10 or 100 µg/kg weight/dose) simultaneously with HDM. After 6 weeks, lung function was measured. Lung lavage fluid (LLF) was used to measure total cell count, total protein, and cytokines. Immunohistochemistry for alpha smooth muscle actin (α-SMA) was done. Airway wall thickness was measured on micro-computed tomography (micro-CT) images. RESULTS: Pulmonary function testing revealed a HDM dose-dependent airway hyperresponsiveness. Airway resistance was increased 2-fold while compliance was decreased by 50% at the higher HDM dose (P<0.05). GC021109 prevented these changes. HDM-exposed mice had elevated inflammatory cell and total protein levels in LLF which were prevented by GC021109 (P<0.05). HDM mice also had elevated LLF levels of interleukin (IL)-4, IL-5, IL-12, granulocyte colony stimulating factor, chemokine (C-X-C) motif ligand 1, and leukemia inhibitory factor that were reduced by GC021109 with a dose-dependent pattern. HDM mice had increased peribronchial and perivascular inflammatory cell infiltration and increased α-SMA; these changes were absent with GC021109. Airway wall thickness measured on micro-CT images was increased after HDM exposure and significantly reduced by GC021109 treatment. CONCLUSION: The P2Y6R prodrug GC021109 inhibited allergen-induced changes in pulmonary function, inflammatory responses, and airway and vascular smooth muscle mass. P2Y6R activation may be an effective therapeutic maintenance strategy in asthma.

10.
Front Biosci (Elite Ed) ; 8(3): 370-7, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27100345

ABSTRACT

The temporal origins of childhood asthma are incompletely understood. We hypothesize that allergen sensitization which begins in early infancy causes IgE-mediated airway and vascular remodeling, and airway hyper-responsiveness. Mice were sensitized with ovalbumin (OVA) without or with anti-IgE antibody from postnatal day (P) 10 through P42. We studied airway resistance in response to Methacholine (MCh) challenge, bronchoalveolar lavage fluid (BAL) inflammatory cell content, immunohistochemistry for inflammation, alpha-smooth muscle actin (alpha-SMA) and platelet/endothelial cell adhesion molecule (PECAM) proteins, and Western blotting for vascular endothelial growth factor (VEGF) protein. Compared to controls, mice treated with OVA had increased airway resistance (baseline: 192% of control; MCH 12 mg/mL 170% of control; P less than 0.0.5). OVA treatment also increased lung alpha-SMA, VEGF and PECAM compared to controls. Inflammatory cells in the BAL and perivascular and peribronchiolar inflammatory cell infiltrates increased over controls with OVA exposure. These changes were counteracted by anti-IgE treatment. We conclude that mice sensitized in early infancy develop an IgE-mediated hyper-reactive airway disease with airway and vascular remodeling. Preventive approaches in early infancy of at-risk individuals may reduce childhood asthma.


Subject(s)
Immunoglobulin E/physiology , Vascular Remodeling , Animals , Asthma/etiology , Asthma/immunology , Asthma/pathology , Blotting, Western , Bronchial Hyperreactivity/etiology , Bronchial Hyperreactivity/pathology , Immunization , Immunohistochemistry , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...