Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
N Engl J Med ; 389(9): 820-832, 2023 Aug 31.
Article in English | MEDLINE | ID: mdl-37646679

ABSTRACT

BACKGROUND: Sickle cell disease is caused by a defect in the ß-globin subunit of adult hemoglobin. Sickle hemoglobin polymerizes under hypoxic conditions, producing deformed red cells that hemolyze and cause vaso-occlusion that results in progressive organ damage and early death. Elevated fetal hemoglobin levels in red cells protect against complications of sickle cell disease. OTQ923, a clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-edited CD34+ hematopoietic stem- and progenitor-cell (HSPC) product, has a targeted disruption of the HBG1 and HBG2 (γ-globin) gene promoters that increases fetal hemoglobin expression in red-cell progeny. METHODS: We performed a tiling CRISPR-Cas9 screen of the HBG1 and HBG2 promoters by electroporating CD34+ cells obtained from healthy donors with Cas9 complexed with one of 72 guide RNAs, and we assessed the fraction of fetal hemoglobin-immunostaining erythroblasts (F cells) in erythroid-differentiated progeny. The gRNA resulting in the highest level of F cells (gRNA-68) was selected for clinical development. We enrolled participants with severe sickle cell disease in a multicenter, phase 1-2 clinical study to assess the safety and adverse-effect profile of OTQ923. RESULTS: In preclinical experiments, CD34+ HSPCs (obtained from healthy donors and persons with sickle cell disease) edited with CRISPR-Cas9 and gRNA-68 had sustained on-target editing with no off-target mutations and produced high levels of fetal hemoglobin after in vitro differentiation or xenotransplantation into immunodeficient mice. In the study, three participants received autologous OTQ923 after myeloablative conditioning and were followed for 6 to 18 months. At the end of the follow-up period, all the participants had engraftment and stable induction of fetal hemoglobin (fetal hemoglobin as a percentage of total hemoglobin, 19.0 to 26.8%), with fetal hemoglobin broadly distributed in red cells (F cells as a percentage of red cells, 69.7 to 87.8%). Manifestations of sickle cell disease decreased during the follow-up period. CONCLUSIONS: CRISPR-Cas9 disruption of the HBG1 and HBG2 gene promoters was an effective strategy for induction of fetal hemoglobin. Infusion of autologous OTQ923 into three participants with severe sickle cell disease resulted in sustained induction of red-cell fetal hemoglobin and clinical improvement in disease severity. (Funded by Novartis Pharmaceuticals; ClinicalTrials.gov number, NCT04443907.).


Subject(s)
Anemia, Sickle Cell , CRISPR-Cas Systems , Erythrocytes , Fetal Hemoglobin , Hematopoietic Stem Cell Transplantation , Animals , Mice , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Antigens, CD34 , Fetal Hemoglobin/biosynthesis , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Hemoglobin, Sickle , Promoter Regions, Genetic
2.
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
3.
Sci Rep ; 8(1): 16304, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389991

ABSTRACT

CRISPR/Cas9 mediated gene editing of patient-derived hematopoietic stem and progenitor cells (HSPCs) ex vivo followed by autologous transplantation of the edited HSPCs back to the patient can provide a potential cure for monogenic blood disorders such as ß-hemoglobinopathies. One challenge for this strategy is efficient delivery of the ribonucleoprotein (RNP) complex, consisting of purified Cas9 protein and guide RNA, into HSPCs. Because ß-hemoglobinopathies are most prevalent in developing countries, it is desirable to have a reliable, efficient, easy-to-use and cost effective delivery method. With this goal in mind, we developed TRansmembrane Internalization Assisted by Membrane Filtration (TRIAMF), a new method to quickly and effectively deliver RNPs into HSPCs by passing a RNP and cell mixture through a filter membrane. We achieved robust gene editing in HSPCs using TRIAMF and demonstrated that the multilineage colony forming capacities and the competence for engraftment in immunocompromised mice of HSPCs were preserved post TRIAMF treatment. TRIAMF is a custom designed system using inexpensive components and has the capacity to process HSPCs at clinical scale.


Subject(s)
Fetal Hemoglobin/genetics , Filtration/methods , Gene Editing/methods , Hematopoietic Stem Cell Transplantation , Ribonucleoproteins/genetics , Animals , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , Cells, Cultured , Electroporation/methods , Female , Fetal Hemoglobin/metabolism , Filtration/economics , Filtration/instrumentation , Genetic Therapy/economics , Genetic Therapy/instrumentation , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Humans , Membranes, Artificial , Mice , Models, Animal , RNA, Guide, Kinetoplastida/genetics , Transplantation, Autologous
5.
Endocrinology ; 158(2): 264-276, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27906582

ABSTRACT

Factor inhibiting activating transcription factor 4 (ATF4)-mediated transcription (FIAT) interacts with ATF4 to repress its transcriptional activity. We performed a phenotypic analysis of Fiat-deficient male mice (Fiat-/Y) at 8 and 16 weeks of age. Microcomputed tomography analysis of the distal femur demonstrated 46% and 13% age-dependent increases in trabecular bone volume and thickness, respectively, in Fiat-/Y mice. Cortical bone measurements at the femoral midshaft revealed a substantial increase in cortical thickness in older Fiat-/Y mice. Bone gain was related to increased mineral apposition rate and increased osteoblast function. Femoral stiffness and strength were substantially increased in Fiat-/Y compared with wild-type (WT) mice. We also investigated whether FIAT contributes to metabolic function. When fed standard mouse chow, Fiat-/Y animals were glucose-tolerant. However, when fed a high-fat diet (HFD) for 8 weeks, Fiat-/Y mice gained more weight than control mice, with a specific increase in white adipose tissue fat mass. The increase in fat mass was due to reduced energy expenditure, which correlated with reduced fatty acid oxidation and lipolysis in the adipose tissue of mutant mice. The expression of the Scd1 gene, involved in lipogenesis, was upregulated in the subcutaneous adipose tissue of Fiat-/Y mice. Moreover, HFD-fed Fiat-/Y mice exhibited increased circulating leptin and insulin levels relative to WT mice, demonstrating that endocrine abnormalities are associated with the disturbance in energy balance. We conclude that Fiat-/Y mice exhibited an anabolic bone phenotype but displayed increased susceptibility to developing metabolic-related disorders when consuming an HFD.


Subject(s)
Bone Density , Co-Repressor Proteins/metabolism , Diet, High-Fat/adverse effects , Energy Metabolism , Nuclear Proteins/metabolism , Adipose Tissue, White/metabolism , Adiposity , Age Factors , Animals , Body Weight , Co-Repressor Proteins/genetics , Lipogenesis , Lipolysis , Mice, Knockout , Nuclear Proteins/genetics , Osteoblasts/metabolism , Oxygen Consumption , Phenotype , Weight-Bearing
6.
Cell ; 167(5): 1310-1322.e17, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863245

ABSTRACT

Stem cells determine homeostasis and repair of many tissues and are increasingly recognized as functionally heterogeneous. To define the extent of-and molecular basis for-heterogeneity, we overlaid functional, transcriptional, and epigenetic attributes of hematopoietic stem cells (HSCs) at a clonal level using endogenous fluorescent tagging. Endogenous HSC had clone-specific functional attributes over time in vivo. The intra-clonal behaviors were highly stereotypic, conserved under the stress of transplantation, inflammation, and genotoxic injury, and associated with distinctive transcriptional, DNA methylation, and chromatin accessibility patterns. Further, HSC function corresponded to epigenetic configuration but not always to transcriptional state. Therefore, hematopoiesis under homeostatic and stress conditions represents the integrated action of highly heterogeneous clones of HSC with epigenetically scripted behaviors. This high degree of epigenetically driven cell autonomy among HSCs implies that refinement of the concepts of stem cell plasticity and of the stem cell niche is warranted.


Subject(s)
Epigenomics , Hematopoietic Stem Cells/cytology , Animals , Cell Lineage , Clone Cells/cytology , Fluorescence , Hematopoiesis , Inflammation/pathology , Mice , Transcription, Genetic
7.
Stem Cell Reports ; 7(2): 220-35, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27453006

ABSTRACT

Bone marrow niches for hematopoietic progenitor cells are not well defined despite their critical role in blood homeostasis. We previously found that cells expressing osteocalcin, a marker of mature osteolineage cells, regulate the production of thymic-seeding T lymphoid progenitors. Here, using a selective cell deletion strategy, we demonstrate that a subset of mesenchymal cells expressing osterix, a marker of bone precursors in the adult, serve to regulate the maturation of early B lymphoid precursors by promoting pro-B to pre-B cell transition through insulin-like growth factor 1 (IGF-1) production. Loss of Osx(+) cells or Osx-specific deletion of IGF-1 led to a failure of B cell maturation and the impaired adaptive immune response. These data highlight the notion that bone marrow is a composite of specialized niches formed by pairings of specific mesenchymal cells with parenchymal stem or lineage committed progenitor cells, thereby providing distinctive functional units to regulate hematopoiesis.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Mesoderm/cytology , Animals , B-Lymphocytes/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Insulin-Like Growth Factor I/metabolism , Mice , Sp7 Transcription Factor/metabolism
8.
Curr Opin Hematol ; 23(4): 331-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27177311

ABSTRACT

PURPOSE OF REVIEW: The bone marrow niche is increasingly recognized as heterogeneous with specific subtypes of mesenchymal niche cells governing the development or homeostasis of selective parenchymal hematopoietic subsets. The present review outlines recent efforts in dissecting these microniches regulated by unique cell pairings within the bone marrow and provides an overview of how the bone marrow orchestrates multiple facets of hematopoiesis. RECENT FINDINGS: Recent advancement in technologies has significantly improved our understanding of the cellular and molecular constituents that contribute to regulation of hematopoiesis and to maintenance of the hematopoietic stem cells (HSCs). Transgenic mouse models that enable endogenous cell deletion or lineage tracing, coupled with advanced intravital microscopy has identified several mesenchymal cell types, including the osteolineage cells, megakaryocytes, macrophages, perivascular cells, and Schwann cells, to be indispensible regulators of hematopoiesis. These niche cells, when perturbed, each caused very specific hematopoietic consequences including impairment in B-cell maturation, T lineage development, erythropoiesis, and impact different aspects of HSC behavior such as quiescence, mobilization, and response to acute stress signals. SUMMARY: The emerging concept is that the bone marrow environment is composed of multiple microniches, each consisting of unique pairing of distinct supportive stromal cells with distinct hematopoietic subtypes to regulate a particular branch of hematopoietic cell process. The bone marrow can be viewed as a carrier with subcompartments tailored to support different hematopoietic activities.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Stem Cell Niche , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Communication , Cell Cycle , Cell Differentiation , Cell Movement , Erythropoiesis , Hematopoietic Stem Cells/cytology , Humans , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphopoiesis , Macrophages/metabolism , Megakaryocytes/metabolism , Mesenchymal Stem Cells/cytology , Osteogenesis , Stress, Physiological , Sympathetic Nervous System/physiology
9.
Genom Data ; 5: 100-2, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26484235

ABSTRACT

T cells are known to develop in the thymus. However, molecular events that control the transition from hematopoietic progenitor cells in the bone marrow to T precursor cells seeded in the thymus remained poorly defined. Our recent report showed that osteocalcin (Ocn)-expressing bone cells in the bone marrow have major impact on T cell immunity by regulating T progenitor development in the bone marrow (Yu et al., 2015) [1]. Selective endogenous depletion of Ocn(+) cells by inducible diphtheria toxin receptor expression (OcnCre;iDTR) led to reduction of T-competent common lymphoid progenitors (Ly6D(-) CLPs) in the bone marrow and loss of T cells in the thymus. Expression of the Notch ligand DLL4 by Ocn(+) cells in the bone marrow ensures the production of Ly6D(-) CLPs, and expression of chemotactic molecules CCR7 and PSGL1 to enable subsequent thymic seeding. These data indicate that specific mesenchymal cells in bone marrow provide key molecular drivers enforcing thymus-seeding progenitor generation and thereby directly link skeletal biology to the production of T cell based adaptive immunity. Here we present the transcriptome profiles of Ly6D(-) CLPs derived from Ocn(+) cells deleted mice (OcnCre(+);iDTR) compared to those derived from control littermates (OcnCre(-);iDTR). These data are publically available from NCBI Gene Expression Omnibus (GEO) with the accession number GSE66102.

10.
Genom Data ; 5: 318-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26484277

ABSTRACT

The bone marrow niche is recognized as a central player in maintaining and regulating the behavior of hematopoietic stem and progenitor cells. Specific gain-of and loss-of function experiments perturbing a range of osteolineage cells or their secreted proteins had been shown to affect stem cell maintenance (Calvi et al, 2003 [1]; Stier et al., 2005 [2]; Zhang et al., 2003 [3]; Nilsson et al., 2005 [4]; Greenbaum et al., 2013 [5]) and engraftment (Adam et al., 2006, 2009 [6,7]). We used specific in vivo cell deletion approaches to dissect the niche cell-parenchymal cell dependency in a complex bone marrow microenvironment. Endogenous deletion of osteocalcin-expressing (Ocn(+)) cells led to a loss of T immune cells (Yu et al., 2015 [8]. Ocn(+) cells express the Notch ligand DLL4 to communicate with T-competent progenitors, and thereby ensuring T precursor production and expression of chemotactic molecules on their cell surface for subsequent thymic seeding. In contrast, depletion of osterix-expressing (Osx(+)) osteoprogenitors led to reduced B immune cells. These distinct hematopoietic phenotypes suggest specific pairing of mesenchymal niche cells and parenchymal hematopoietic cells in the bone marrow to create unique functional units to support hematopoiesis. Here, we present the global gene expression profiles of these osteolineage subtypes utilizing a triple fluorescent transgenic mouse model (OsxCre(+);Rosa-mCh(+);Ocn:Topaz(+)) that labels Osx(+) cells red, Ocn(+) cells green, and Osx(+) Ocn(+) cells yellow. This system allows isolation of distinct osteolineage subsets within the same animal by flow cytometry. Array data that have been described in our study [8] are also publically available from NCBI Gene Expression Omnibus (GEO) with the accession number GSE66042. Differences in gene expression may correlate with functional difference in supporting hematopoiesis.

11.
Stem Cells ; 33(7): 2280-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25851125

ABSTRACT

Notch is long recognized as a signaling molecule important for stem cell self-renewal and fate determination. Here, we reveal a novel adhesive role of Notch-ligand engagement in hematopoietic stem and progenitor cells (HSPCs). Using mice with conditional loss of O-fucosylglycans on Notch EGF-like repeats important for the binding of Notch ligands, we report that HSPCs with faulty ligand binding ability display enhanced cycling accompanied by increased egress from the marrow, a phenotype mainly attributed to their reduced adhesion to Notch ligand-expressing stromal cells and osteoblastic cells and their altered occupation in osteoblastic niches. Adhesion to Notch ligand-bearing osteoblastic or stromal cells inhibits wild type but not O-fucosylglycan-deficient HSPC cycling, independent of RBP-JK -mediated canonical Notch signaling. Furthermore, Notch-ligand neutralizing antibodies induce RBP-JK -independent HSPC egress and enhanced HSPC mobilization. We, therefore, conclude that Notch receptor-ligand engagement controls HSPC quiescence and retention in the marrow niche that is dependent on O-fucosylglycans on Notch.


Subject(s)
Hematopoietic Stem Cells/metabolism , Receptors, Notch/metabolism , Stem Cell Niche/genetics , Stromal Cells/metabolism , Animals , Humans , Mice , Signal Transduction
12.
J Exp Med ; 212(5): 759-74, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25918341

ABSTRACT

Production of the cells that ultimately populate the thymus to generate α/ß T cells has been controversial, and their molecular drivers remain undefined. Here, we report that specific deletion of bone-producing osteocalcin (Ocn)-expressing cells in vivo markedly reduces T-competent progenitors and thymus-homing receptor expression among bone marrow hematopoietic cells. Decreased intrathymic T cell precursors and decreased generation of mature T cells occurred despite normal thymic function. The Notch ligand DLL4 is abundantly expressed on bone marrow Ocn(+) cells, and selective depletion of DLL4 from these cells recapitulated the thymopoietic abnormality. These data indicate that specific mesenchymal cells in bone marrow provide key molecular drivers enforcing thymus-seeding progenitor generation and thereby directly link skeletal biology to the production of T cell-based adaptive immunity.


Subject(s)
Bone Marrow Cells/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Mesenchymal Stem Cells/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Adaptor Proteins, Signal Transducing , Animals , Bone Marrow Cells/cytology , Calcium-Binding Proteins , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mesenchymal Stem Cells/cytology , Mice , Mice, Transgenic , Osteocalcin/genetics , Osteocalcin/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Thymus Gland/cytology
13.
J Exp Med ; 211(12): 2341-9, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25332287

ABSTRACT

Paradoxical to its importance for generating a diverse T cell repertoire, thymic function progressively declines throughout life. This process has been at least partially attributed to the effects of sex steroids, and their removal promotes enhanced thymopoiesis and recovery from immune injury. We show that one mechanism by which sex steroids influence thymopoiesis is through direct inhibition in cortical thymic epithelial cells (cTECs) of Delta-like 4 (Dll4), a Notch ligand crucial for the commitment and differentiation of T cell progenitors in a dose-dependent manner. Consistent with this, sex steroid ablation (SSA) led to increased expression of Dll4 and its downstream targets. Importantly, SSA induced by luteinizing hormone-releasing hormone (LHRH) receptor antagonism bypassed the surge in sex steroids caused by LHRH agonists, the gold standard for clinical ablation of sex steroids, thereby facilitating increased Dll4 expression and more rapid promotion of thymopoiesis. Collectively, these findings not only reveal a novel mechanism underlying improved thymic regeneration upon SSA but also offer an improved clinical strategy for successfully boosting immune function.


Subject(s)
Gonadal Steroid Hormones/immunology , Intracellular Signaling Peptides and Proteins/immunology , Membrane Proteins/immunology , Receptors, Notch/immunology , Signal Transduction/immunology , Thymocytes/immunology , Adaptor Proteins, Signal Transducing , Animals , Benzamides , Calcium-Binding Proteins , Cell Line , Dihydrotestosterone/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Flow Cytometry , Gonadal Steroid Hormones/antagonists & inhibitors , HEK293 Cells , Hormone Antagonists/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lymphopoiesis/drug effects , Lymphopoiesis/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Receptors, Androgen/immunology , Receptors, LHRH/agonists , Receptors, LHRH/antagonists & inhibitors , Receptors, LHRH/immunology , Receptors, Notch/metabolism , Signal Transduction/drug effects , Testosterone/blood , Testosterone/immunology , Thymocytes/cytology , Thymus Gland/cytology , Thymus Gland/immunology
14.
Blood ; 124(19): 2937-47, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25202142

ABSTRACT

The glycosyltransferase gene, Ext1, is essential for heparan sulfate production. Induced deletion of Ext1 selectively in Mx1-expressing bone marrow (BM) stromal cells, a known population of skeletal stem/progenitor cells, in adult mice resulted in marked changes in hematopoietic stem and progenitor cell (HSPC) localization. HSPC egressed from BM to spleen after Ext1 deletion. This was associated with altered signaling in the stromal cells and with reduced vascular cell adhesion molecule 1 production by them. Further, pharmacologic inhibition of heparan sulfate mobilized qualitatively more potent and quantitatively more HSPC from the BM than granulocyte colony-stimulating factor alone, including in a setting of granulocyte colony-stimulating factor resistance. The reduced presence of endogenous HSPC after Ext1 deletion was associated with engraftment of transfused HSPC without any toxic conditioning of the host. Therefore, inhibiting heparan sulfate production may provide a means for avoiding the toxicities of radiation or chemotherapy in HSPC transplantation for nonmalignant conditions.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Heparitin Sulfate/biosynthesis , N-Acetylglucosaminyltransferases/metabolism , Stromal Cells/metabolism , Transplantation Conditioning , Animals , Anticoagulants/pharmacology , Binding, Competitive/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Green Fluorescent Proteins/genetics , Heparin/pharmacology , Heparitin Sulfate/immunology , Male , Mice, Inbred C57BL , Mice, Transgenic , N-Acetylglucosaminyltransferases/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Stromal Cells/immunology , Vascular Cell Adhesion Molecule-1/immunology , Vascular Cell Adhesion Molecule-1/metabolism
15.
Cell ; 158(6): 1309-1323, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25215489

ABSTRACT

The balance between oxidative and nonoxidative glucose metabolism is essential for a number of pathophysiological processes. By deleting enzymes that affect aerobic glycolysis with different potencies, we examine how modulating glucose metabolism specifically affects hematopoietic and leukemic cell populations. We find that a deficiency in the M2 pyruvate kinase isoform (PKM2) reduces the levels of metabolic intermediates important for biosynthesis and impairs progenitor function without perturbing hematopoietic stem cells (HSCs), whereas lactate dehydrogenase A (LDHA) deletion significantly inhibits the function of both HSCs and progenitors during hematopoiesis. In contrast, leukemia initiation by transforming alleles putatively affecting either HSCs or progenitors is inhibited in the absence of either PKM2 or LDHA, indicating that the cell-state-specific responses to metabolic manipulation in hematopoiesis do not apply to the setting of leukemia. This finding suggests that fine-tuning the level of glycolysis may be explored therapeutically for treating leukemia while preserving HSC function.


Subject(s)
Glycolysis , Hematopoiesis , Leukemia/metabolism , Animals , Gene Deletion , Hematopoietic Stem Cells/metabolism , Humans , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactate Dehydrogenase 5 , Mice , Mice, Congenic , Mice, Inbred C57BL , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
16.
Gene ; 538(2): 328-33, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24440290

ABSTRACT

Factor inhibiting ATF4-mediated transcription (FIAT) interacts with Nascent polypeptide associated complex and coregulator alpha (αNAC). In cultured osteoblastic cells, this interaction contributes to maximal FIAT-mediated inhibition of Osteocalcin (Ocn) gene transcription. We set out to demonstrate the physiological relevance of this interaction by altering gene dosage in compound Fiat and Naca (encoding αNAC) heterozygous mice. Compound Naca(+/-); Fiat(+/-) heterozygous animals were viable, developed normally, and exhibited no significant difference in body weight compared with control littermate genotypes. Animals with a single Fiat allele had reduced Fiat mRNA expression without changes in the expression of related family members. Expression of the osteocyte differentiation marker Dmp1 was elevated in compound heterozygotes. Static histomorphometry parameters were assessed at 8weeks of age using microcomputed tomography (µCT). Trabecular measurements were not different between genotypes. Cortical thickness and area were not affected by gene dosage, but we measured a significant increase in cortical porosity in compound heterozygous mice, without changes in biomechanical parameters. The bone phenotype of compound Naca(+/-); Fiat(+/-) heterozygotes confirms that FIAT and αNAC are part of a common genetic pathway and support a role for the FIAT/αNAC interaction in normal bone physiology.


Subject(s)
Co-Repressor Proteins/genetics , Gene Dosage , Molecular Chaperones/genetics , Nuclear Proteins/genetics , Animals , Biomechanical Phenomena , Bone Development/genetics , Bone and Bones/physiology , Co-Repressor Proteins/metabolism , Epistasis, Genetic , Female , Gene Expression , Heterozygote , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Molecular Chaperones/metabolism , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Phenotype
17.
Gene Expr Patterns ; 9(5): 335-40, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19232401

ABSTRACT

FIAT represses osteocalcin gene transcription by heterodimerizing with ATF4 and preventing it from binding to DNA. We report here the expression profiles of FIAT and ATF4 during osteoblastogenesis. Messenger RNA levels for the osteoblast transcriptional regulators Satb2, Runx2, Fiat, and Atf4 were quantified using real-time reverse-transcription PCR (RT-qPCR) and respective protein levels monitored by immunodetection in differentiating primary osteoblast cultures. Satb2, Fiat, and Atf4 mRNA levels remained constant throughout the differentiation sequence, whereas Runx2 transcript levels were significantly increased by 12 days post-confluency. Using immunofluorescence, the SATB2, RUNX2, and ATF4 signals appeared to increase as a function of time in culture. FIAT protein expression was readily detected in early cultures, but signal intensity decreased thereafter. When immunoblotting was used to quantify the relative amounts of FIAT and ATF4 proteins, the expression levels of the two proteins were found to be inversely correlated. The decrease in FIAT protein levels coincided with increased binding of ATF4 to the osteocalcin gene promoter, and with increased osteocalcin expression measured by RT-qPCR or immunoblotting. Immunohistochemistry of long bones from mice at E16.5 and 2 days post-natal revealed that both proteins are initially expressed in osteoblasts. In adult bone, FIAT was detected in osteocytes, while ATF4 expression was observed in active osteoblasts and lining cells, but not in osteocytes. Taken together, these data support the idea that a stoichiometric excess of ATF4 over FIAT in mature osteoblasts releases ATF4 from sequestration by FIAT, thereby allowing ATF4 homodimerization and subsequent transactivation of the osteocalcin gene.


Subject(s)
Activating Transcription Factor 4/genetics , Carrier Proteins/genetics , Gene Expression Profiling , Nuclear Proteins/genetics , Osteoblasts/metabolism , Activating Transcription Factor 4/chemistry , Activating Transcription Factor 4/metabolism , Animals , Blotting, Western , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Co-Repressor Proteins , Femur/growth & development , Femur/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice , Mice, Inbred C57BL , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Osteoblasts/cytology , Osteocalcin/genetics , Osteocytes/cytology , Osteocytes/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Multimerization , Reverse Transcriptase Polymerase Chain Reaction , Tibia/growth & development , Tibia/metabolism , Time Factors
18.
J Cell Biochem ; 106(1): 186-92, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19016261

ABSTRACT

The ATF4 transcription factor is a key regulator of osteoblast differentiation that controls osteocalcin gene transcription and type I collagen protein synthesis. We have characterized factor-inhibiting ATF4-mediated transcription (FIAT), a leucine zipper protein that dimerizes with ATF4 to form inactive dimers that cannot bind DNA. Overexpression of FIAT in osteoblasts of transgenic mice inhibited osteocalcin gene transcription and reduced osteoblastic activity, leading to osteopenia (Yu et al. [2005] J Cell Biol 169:591-601). We therefore hypothesized that inhibition of FIAT would enhance ATF4 activity, leading to increased osteocalcin transcription, type I collagen synthesis, and mineralization. We used small interfering RNAs (siRNA) to knockdown FIAT in pools of MC3T3-E1 cells stably transfected with 1.3 kb of the mouse osteocalcin gene promoter driving expression of luciferase. Stable expression of the FIAT siRNA sequence inhibited FIAT expression without significantly affecting the level of total or Ribosomal S6 Kinase-2-phosphorylated ATF4 protein. Occupancy of the osteocalcin proximal promoter by ATF4 was increased and transcription of the osteocalcin-promoter-dependent luciferase reporter showed earlier onset and increased levels. Similarly, endogenous osteocalcin gene expression was enhanced in primary osteoblasts transfected with the FIAT siRNA. FIAT knockdown cells also displayed higher expression of bone sialoprotein, increased type I collagen protein synthesis, and enhanced mineralization. These data suggest that inhibition of FIAT expression increases ATF4 activity and confirm the important role of FIAT in osteoblast function.


Subject(s)
Activating Transcription Factor 4/metabolism , Carrier Proteins/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Osteoblasts/metabolism , Animals , Co-Repressor Proteins , Mice , Osteocalcin/genetics , Osteocalcin/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
19.
J Cell Biochem ; 105(3): 859-65, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18680144

ABSTRACT

We have characterized FIAT, a 66 kDa leucine zipper (LZ) protein that dimerizes with activating transcription factor 4 (ATF4) to form inactive dimers that cannot bind DNA. Computer analysis identifies three putative LZ motifs within the FIAT amino acid sequence. We have used deletion- and/or site-specific mutagenesis to individually inactivate these motifs in order to identify the functional LZ that mediates the FIAT-ATF4 interaction. Amino acids 194-222 that encode the FIAT LZ2 were deleted (mutant FIAT ZIP2 DEL). We inactivated each zipper individually by replacing two or three leucine residues within each zipper by alanine residues. The engineered mutations were L142A/L149A (mutant M1, first zipper), L208A/L215A/L222A (mutant M2, second zipper), and L441A/L448A (mutant M3, third zipper). MC3T3-E1 osteoblastic cells with an integrated 1.3 kb mouse osteocalcin gene promoter fragment driving expression of luciferase were transfected with expression vectors for ATF4 and the various FIAT deletion- or site-specific mutants. Inhibition of ATF4-mediated transcription was compared between wild-type (WT) and LZ FIAT mutants. The deletion mutant FIAT ZIP2 DEL and the sequence-specific M2 mutant did not interact with ATF4 and were unable to inhibit ATF4-mediated transcription. The M1 or M3 mutations did not affect the ability of FIAT to contact ATF4 or to inhibit its transcriptional activity. Stable expression of WT FIAT in osteoblastic cells inhibited mineralization, but not expression of the FIAT ZIP2 DEL and M2 mutants. This structure-function analysis reveals that FIAT interacts with ATF4 and modulates its activity through its second leucine zipper motif.


Subject(s)
Activating Transcription Factor 4/metabolism , Bone Matrix/metabolism , Calcification, Physiologic , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Leucine Zippers , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/genetics , Co-Repressor Proteins , Mice , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Osteocalcin/genetics , Osteocalcin/metabolism , Transcription, Genetic , Transfection
20.
Ann N Y Acad Sci ; 1068: 131-42, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16831913

ABSTRACT

The basic domain-leucine zipper protein, activating transcription factor 4 (ATF4), was recently shown to control key aspects of osteoblast biology. ATF4 regulates the timely onset of osteoblast differentiation, the synthesis of type I collagen, and the transcription of the osteocalcin and RANKL (receptor activator of NFkappa-B ligand) genes. Accordingly, the levels and activity of ATF4 are under tight control through mechanisms that include protein stability and phosphorylation. We have uncovered yet another mode of inhibition of ATF4 through its interaction with the leucine zipper protein FIAT (Factor Inhibiting ATF4-mediated Transcription, also described as gamma-taxilin). FIAT/gamma-taxilin localizes to the nucleus in osteoblasts and dimerizes with ATF4 to form inactive dimers, because it does not contain a DNA-binding basic domain moiety. The interaction of FIAT/gamma-taxilin with ATF4 thus inhibits ATF4-mediated transcription. Transgenic mice overexpressing FIAT/gamma-taxilin show osteopenia and reduced expression of the ATF4 target gene, osteocalcin. Interestingly, FIAT/gamma-taxilin also interacts with the transcriptional co-activator alphaNAC (Nascent polypeptide associated complex And Coactivator alpha), suggesting alternative, non-mutually exclusive mechanisms contributing to the inhibition of ATF4-dependent osteocalcin gene transcription by FIAT/gamma-taxilin.


Subject(s)
Activating Transcription Factor 4/antagonists & inhibitors , Bone Density/physiology , Carrier Proteins/physiology , Nuclear Proteins/physiology , Vesicular Transport Proteins/physiology , Animals , Bone Resorption/genetics , Cell Division , Co-Repressor Proteins , Humans , Membrane Glycoproteins/physiology , Mice , Mice, Transgenic , Models, Biological , Osteoblasts/cytology , Osteoblasts/physiology , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/physiology , Vesicular Transport Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...