Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Leukemia ; 38(3): 521-529, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38245602

ABSTRACT

Constitutional trisomy 21 (T21) is a state of aneuploidy associated with high incidence of childhood acute myeloid leukemia (AML). T21-associated AML is preceded by transient abnormal myelopoiesis (TAM), which is triggered by truncating mutations in GATA1 generating a short GATA1 isoform (GATA1s). T21-associated AML emerges due to secondary mutations in hematopoietic clones bearing GATA1s. Since aneuploidy generally impairs cellular fitness, the paradoxically elevated risk of myeloid malignancy in T21 is not fully understood. We hypothesized that individuals with T21 bear inherent genome instability in hematopoietic lineages that promotes leukemogenic mutations driving the genesis of TAM and AML. We found that individuals with T21 show increased chromosomal copy number variations (CNVs) compared to euploid individuals, suggesting that genome instability could be underlying predisposition to TAM and AML. Acquisition of GATA1s enforces myeloid skewing and maintenance of the hematopoietic progenitor state independently of T21; however, GATA1s in T21 hematopoietic progenitor cells (HPCs) further augments genome instability. Increased dosage of the chromosome 21 (chr21) gene DYRK1A impairs homology-directed DNA repair as a mechanism of elevated mutagenesis. These results posit a model wherein inherent genome instability in T21 drives myeloid malignancy in concert with GATA1s mutations.


Subject(s)
Down Syndrome , Leukemia, Myeloid, Acute , Leukemoid Reaction , Myeloproliferative Disorders , Humans , Child , Down Syndrome/complications , DNA Copy Number Variations , Myeloproliferative Disorders/genetics , Genomic Instability , Leukemia, Myeloid, Acute/pathology , Aneuploidy , Trisomy , GATA1 Transcription Factor/genetics
2.
Res Sq ; 2023 Oct 05.
Article in English | MEDLINE | ID: mdl-37886538

ABSTRACT

Abundant macrophage infiltration and altered tumor metabolism are two key hallmarks of glioblastoma. By screening a cluster of metabolic small-molecule compounds, we show that inhibiting glioblastoma cell glycolysis impairs macrophage migration and lactate dehydrogenase (LDH) inhibitor stiripentol (an FDA-approved anti-seizure drug for Dravet Syndrome) emerges as the top hit. Combined profiling and functional studies demonstrate that LDHA-directed ERK pathway activates YAP1/STAT3 transcriptional co-activators in glioblastoma cells to upregulate CCL2 and CCL7, which recruit macrophages into the tumor microenvironment. Reciprocally, infiltrating macrophages produce LDHA-containing extracellular vesicles to promote glioblastoma cell glycolysis, proliferation, and survival. Genetic and pharmacological inhibition of LDHA-mediated tumor-macrophage symbiosis markedly suppresses tumor progression and macrophage infiltration in glioblastoma mouse models. Analysis of tumor and plasma samples of glioblastoma patients confirms that LDHA and its downstream signals are potential biomarkers correlating positively with macrophage density. Thus, LDHA-mediated tumor-macrophage symbiosis provides therapeutic targets for glioblastoma.

3.
Leukemia ; 37(8): 1698-1708, 2023 08.
Article in English | MEDLINE | ID: mdl-37391485

ABSTRACT

Many inherited bone marrow failure syndromes (IBMFSs) present a high risk of transformation to myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). During transformation of IBMFSs, hematopoietic stem and progenitor cells (HSPCs) with poor fitness gain ectopic, dysregulated self-renewal secondary to somatic mutations via undefined mechanisms. Here, in the context of the prototypical IBMFS Fanconi anemia (FA), we performed multiplexed gene editing of mutational hotspots in MDS-associated genes in human induced pluripotent stem cells (iPSCs) followed by hematopoietic differentiation. We observed aberrant self-renewal and impaired differentiation of HSPCs with enrichment of RUNX1 insertions and deletions (indels), generating a model of IBMFS-associated MDS. We observed that compared to the failure state, FA MDS cells show mutant RUNX1-mediated blunting of the G1/S cell cycle checkpoint that is normally activated in FA in response to DNA damage. RUNX1 indels also lead to activation of innate immune signaling, which stabilizes the homologous recombination (HR) effector BRCA1, and this pathway can be targeted to abrogate viability and restore sensitivity to genotoxins in FA MDS. Together, these studies develop a paradigm for modeling clonal evolution in IBMFSs, provide basic understanding of the pathogenesis of MDS, and uncover a therapeutic target in FA-associated MDS.


Subject(s)
Fanconi Anemia , Induced Pluripotent Stem Cells , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Fanconi Anemia/genetics , Fanconi Anemia/pathology , Fanconi Anemia/therapy , Congenital Bone Marrow Failure Syndromes/complications , Core Binding Factor Alpha 2 Subunit/genetics , Induced Pluripotent Stem Cells/pathology , Myelodysplastic Syndromes/pathology , Mutation , Leukemia, Myeloid, Acute/pathology
4.
Cell Rep ; 39(1): 110587, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35385744

ABSTRACT

Hematopoiesis changes over life to meet the demands of maturation and aging. Here, we find that the definitive hematopoietic stem and progenitor cell (HSPC) compartment is remodeled from gestation into adulthood, a process regulated by the heterochronic Lin28b/let-7 axis. Native fetal and neonatal HSPCs distribute with a pro-lymphoid/erythroid bias with a shift toward myeloid output in adulthood. By mining transcriptomic data comparing juvenile and adult HSPCs and reconstructing coordinately activated gene regulatory networks, we uncover the Polycomb repressor complex 1 (PRC1) component Cbx2 as an effector of Lin28b/let-7's control of hematopoietic maturation. We find that juvenile Cbx2-/- hematopoietic tissues show impairment of B-lymphopoiesis, a precocious adult-like myeloid bias, and that Cbx2/PRC1 regulates developmental timing of expression of key hematopoietic transcription factors. These findings define a mechanism of regulation of HSPC output via chromatin modification as a function of age with potential impact on age-biased pediatric and adult blood disorders.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells , MicroRNAs , Polycomb Repressive Complex 1 , RNA-Binding Proteins , Adult , Animals , Child , Gene Regulatory Networks , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Infant, Newborn , Lymphopoiesis , Mice , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
5.
Cell Rep ; 39(4): 110752, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35476984

ABSTRACT

High-risk forms of B-acute lymphoblastic leukemia (B-ALL) remain a therapeutic challenge. Leukemia-initiating cells (LICs) self-renew and spark relapse and therefore have been the subject of intensive investigation; however, the properties of LICs in high-risk B-ALL are not well understood. Here, we use single-cell transcriptomics and quantitative xenotransplantation to understand LICs in MLL-rearranged (MLL-r) B-ALL. Compared with reported LIC frequencies in acute myeloid leukemia (AML), engraftable LICs in MLL-r B-ALL are abundant. Although we find that multipotent, self-renewing LICs are enriched among phenotypically undifferentiated B-ALL cells, LICs with the capacity to replenish the leukemic cellular diversity can emerge from more mature fractions. While inhibiting oxidative phosphorylation blunts blast proliferation, this intervention promotes LIC emergence. Conversely, inhibiting hypoxia and glycolysis impairs MLL-r B-ALL LICs, providing a therapeutic benefit in xenotransplantation systems. These findings provide insight into the aggressive nature of MLL-r B-ALL and provide a rationale for therapeutic targeting of hypoxia and glycolysis.


Subject(s)
Leukemia, Myeloid, Acute , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Glycolysis , Humans , Hypoxia , Leukemia, Myeloid, Acute/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
6.
Blood Adv ; 4(19): 4679-4692, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33002135

ABSTRACT

Fanconi anemia (FA) is a disorder of DNA repair that manifests as bone marrow (BM) failure. The lack of accurate murine models of FA has refocused efforts toward differentiation of patient-derived induced pluripotent stem cells (IPSCs) to hematopoietic progenitor cells (HPCs). However, an intact FA DNA repair pathway is required for efficient IPSC derivation, hindering these efforts. To overcome this barrier, we used inducible complementation of FANCA-deficient IPSCs, which permitted robust maintenance of IPSCs. Modulation of FANCA during directed differentiation to HPCs enabled the production of FANCA-deficient human HPCs that recapitulated FA genotoxicity and hematopoietic phenotypes relative to isogenic FANCA-expressing HPCs. FANCA-deficient human HPCs underwent accelerated terminal differentiation driven by activation of p53/p21. We identified growth arrest specific 6 (GAS6) as a novel target of activated p53 in FANCA-deficient HPCs and modulate GAS6 signaling to rescue hematopoiesis in FANCA-deficient cells. This study validates our strategy to derive a sustainable, highly faithful human model of FA, uncovers a mechanism of HPC exhaustion in FA, and advances toward future cell therapy in FA.


Subject(s)
Fanconi Anemia , Induced Pluripotent Stem Cells , Animals , Cell Differentiation , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group A Protein/genetics , Humans , Mice , Tumor Suppressor Protein p53/genetics
7.
Proc Natl Acad Sci U S A ; 117(9): 4653-4663, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32060122

ABSTRACT

The LIN28:pre-let-7:TUTase ternary complex regulates pluripotency and oncogenesis by controlling processing of the let-7 family of microRNAs. The complex oligouridylates the 3' ends of pre-let-7 molecules, leading to their degradation via the DIS3L2 exonuclease. Previous studies suggest that components of this complex are potential therapeutic targets in malignancies that aberrantly express LIN28. In this study we developed a functional epitope selection approach to identify nanobody inhibitors of the LIN28:pre-let-7:TUT4 complex. We demonstrate that one of the identified nanobodies, Nb-S2A4, targets the 106-residue LIN28:let-7 interaction (LLI) fragment of TUT4. Nb-S2A4 can effectively inhibit oligouridylation and monouridylation of pre-let-7g in vitro. Expressing Nb-S2A4 allows maturation of the let-7 species in cells expressing LIN28, highlighting the therapeutic potential of targeting the LLI fragment.


Subject(s)
DNA-Binding Proteins/immunology , MicroRNAs/metabolism , RNA 3' End Processing , Single-Domain Antibodies/immunology , Animals , Binding Sites , DNA-Binding Proteins/chemistry , HEK293 Cells , HeLa Cells , Humans , Mice , MicroRNAs/genetics , Protein Binding , RNA Stability , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sf9 Cells , Spodoptera
8.
J Exp Med ; 216(3): 527-538, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30728174

ABSTRACT

Leukemia phenotypes vary with age of onset. Delineating mechanisms of age specificity in leukemia could improve disease models and uncover new therapeutic approaches. Here, we used heterochronic transplantation of leukemia driven by MLL/KMT2A translocations to investigate the contribution of the age of the hematopoietic microenvironment to age-specific leukemia phenotypes. When driven by MLL-AF9, leukemia cells in the adult microenvironment sustained a myeloid phenotype, whereas the neonatal microenvironment supported genesis of mixed early B cell/myeloid leukemia. In MLL-ENL leukemia, the neonatal microenvironment potentiated B-lymphoid differentiation compared with the adult. Ccl5 elaborated from adult marrow stroma inhibited B-lymphoid differentiation of leukemia cells, illuminating a mechanism of age-specific lineage commitment. Our study illustrates the contribution of the developmental stage of the hematopoietic microenvironment in defining the age specificity of leukemia.


Subject(s)
Hematopoiesis/physiology , Leukemia/pathology , Oncogene Proteins, Fusion/genetics , Aging , Animals , Animals, Newborn , B-Lymphocytes/pathology , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Female , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Histone-Lysine N-Methyltransferase/genetics , Leukemia/genetics , Leukocyte Common Antigens/metabolism , Male , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/genetics , Stromal Cells/pathology , Tumor Microenvironment
9.
Nat Rev Genet ; 20(7): 377-388, 2019 07.
Article in English | MEDLINE | ID: mdl-30737492

ABSTRACT

The derivation of induced pluripotent stem cells (iPSCs) over a decade ago sparked widespread enthusiasm for the development of new models of human disease, enhanced platforms for drug discovery and more widespread use of autologous cell-based therapy. Early studies using directed differentiation of iPSCs frequently uncovered cell-level phenotypes in monogenic diseases, but translation to tissue-level and organ-level diseases has required development of more complex, 3D, multicellular systems. Organoids and human-rodent chimaeras more accurately mirror the diverse cellular ecosystems of complex tissues and are being applied to iPSC disease models to recapitulate the pathobiology of a broad spectrum of human maladies, including infectious diseases, genetic disorders and cancer.


Subject(s)
Communicable Diseases/therapy , Genetic Diseases, Inborn/therapy , Induced Pluripotent Stem Cells/cytology , Models, Biological , Neoplasms/therapy , Tissue Engineering/methods , Animals , Cell Differentiation , Cell Lineage/genetics , Cell Lineage/immunology , Chimera/genetics , Chimera/immunology , Communicable Diseases/genetics , Communicable Diseases/immunology , Communicable Diseases/pathology , Drug Discovery/methods , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/immunology , Genetic Diseases, Inborn/pathology , Genetic Therapy/methods , Humans , Induced Pluripotent Stem Cells/immunology , Induced Pluripotent Stem Cells/transplantation , Models, Animal , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Organoids/cytology , Organoids/drug effects , Organoids/immunology , Tissue Transplantation/methods , Transplantation, Heterologous
10.
Blood Adv ; 3(1): 72-82, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622145

ABSTRACT

Platelets are essential for hemostasis; however, several studies have identified age-dependent differences in platelet function. To better understand the origins of fetal platelet function, we have evaluated the contribution of the fetal-specific RNA binding protein Lin28b in the megakaryocyte/platelet lineage. Because activated fetal platelets have very low levels of P-selectin, we hypothesized that the expression of platelet P-selectin is part of a fetal-specific hematopoietic program conferred by Lin28b. Using the mouse as a model, we find that activated fetal platelets have low levels of P-selectin and do not readily associate with granulocytes in vitro and in vivo, relative to adult controls. Transcriptional analysis revealed high levels of Lin28b and Hmga2 in fetal, but not adult, megakaryocytes. Overexpression of LIN28B in adult mice significantly reduces the expression of P-selectin in platelets, and therefore identifies Lin28b as a negative regulator of P-selectin expression. Transplantation of fetal hematopoietic progenitors resulted in the production of platelets with low levels of P-selectin, suggesting that the developmental regulation of P-selectin is intrinsic and independent of differences between fetal and adult microenvironments. Last, we observe that the upregulation of P-selectin expression occurs postnatally, and the temporal kinetics of this upregulation are recapitulated by transplantation of fetal hematopoietic stem and progenitor cells into adult recipients. Taken together, these studies identify Lin28b as a new intrinsic regulator of fetal platelet function.


Subject(s)
Blood Platelets/metabolism , Gene Expression Regulation , RNA-Binding Proteins/genetics , Age Factors , Animals , Biomarkers , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Mice , P-Selectin/genetics , P-Selectin/metabolism , Platelet Activation , Platelet Aggregation/genetics , Platelet Function Tests , RNA-Binding Proteins/metabolism
11.
Cell Rep ; 23(10): 3091-3101, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29874593

ABSTRACT

LIN28 is an RNA-binding protein that regulates the maturation of the let-7 family of microRNAs by bipartite interactions with let-7 precursors through its two distinct cold shock and zinc-knuckle domains. Through inhibition of let-7 biogenesis, LIN28 functions as a pluripotency factor, as well as a driver of tumorigenesis. Here, we report a fluorescence polarization assay to identify small-molecule inhibitors for both domains of LIN28 involved in let-7 interactions. Of 101,017 compounds screened, six inhibit LIN28:let-7 binding and impair LIN28-mediated let-7 oligouridylation. Upon further characterization, we demonstrate that the LIN28 inhibitor TPEN destabilizes the zinc-knuckle domain of LIN28, while LI71 binds the cold shock domain to suppress LIN28's activity against let-7 in leukemia cells and embryonic stem cells. Our results demonstrate selective pharmacologic inhibition of individual domains of LIN28 and provide a foundation for therapeutic inhibition of the let-7 biogenesis pathway in LIN28-driven diseases.


Subject(s)
MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , Small Molecule Libraries/pharmacology , Uridine/metabolism , Binding Sites , Cell Line, Tumor , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fluorescence Polarization , High-Throughput Screening Assays , Humans , MicroRNAs/genetics , Models, Molecular , Niacin/chemistry , Small Molecule Libraries/chemistry
12.
Nat Commun ; 9(1): 892, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29497036

ABSTRACT

A better understanding of the cell-fate transitions that occur in complex cellular ecosystems in normal development and disease could inform cell engineering efforts and lead to improved therapies. However, a major challenge is to simultaneously identify new cell states, and their transitions, to elucidate the gene expression dynamics governing cell-type diversification. Here, we present CellRouter, a multifaceted single-cell analysis platform that identifies complex cell-state transition trajectories by using flow networks to explore the subpopulation structure of multi-dimensional, single-cell omics data. We demonstrate its versatility by applying CellRouter to single-cell RNA sequencing data sets to reconstruct cell-state transition trajectories during hematopoietic stem and progenitor cell (HSPC) differentiation to the erythroid, myeloid and lymphoid lineages, as well as during re-specification of cell identity by cellular reprogramming of monocytes and B-cells to HSPCs. CellRouter opens previously undescribed paths for in-depth characterization of complex cellular ecosystems and establishment of enhanced cell engineering approaches.


Subject(s)
Hematopoietic Stem Cells/cytology , Single-Cell Analysis/methods , Cell Differentiation , Cell Lineage , Gene Expression , Humans , Sequence Analysis, RNA , Single-Cell Analysis/instrumentation
13.
Pediatr Blood Cancer ; 65(4)2018 04.
Article in English | MEDLINE | ID: mdl-29251406

ABSTRACT

BACKGROUND: In osteosarcoma, patient survival has not changed in over 30 years. Multiple phase II trials have been conducted in osteosarcoma using the Response Evaluation Criteria in Solid Tumors (RECIST) as a primary endpoint; however, none of these have revealed new treatment strategies. We investigated RECIST in newly diagnosed patients who received neoadjuvant chemotherapy proven to be beneficial. METHODS: Patients treated from 1986 to 2011 for newly diagnosed osteosarcoma with paired tumor imaging before and after adequate neoadjuvant chemotherapy were included in this retrospective study. Two radiologists performed independent, blinded (to image timing) RECIST measurements of primary tumor and lung metastases at diagnosis and post-neoadjuvant chemotherapy. Association between RECIST and histological necrosis and outcome were assessed. RESULTS: Seventy-four patients met inclusion criteria. Five-year overall survival and progression-free survival (PFS) were 77 ± 7% and 61 ± 8%, respectively. No patients had RECIST partial or complete response in the primary tumor. Sixty-four patients (86%) had stable disease, and 10 (14%) had progressive disease (PD). PD in the primary tumor was associated with significantly worse PFS in localized disease patients (P = 0.02). There was no association between RECIST in the primary tumor and necrosis. There were an insufficient number of patients with lung nodules ≥1 cm at diagnosis to evaluate RECIST in pulmonary metastases. CONCLUSIONS: PD by RECIST predicts poor outcome in localized disease patients. In bone lesions, chemotherapy proven to improve overall survival does not result in radiographic responses as measured by RECIST. Further investigation of RECIST in pulmonary metastatic disease in osteosarcoma is needed.


Subject(s)
Bone Neoplasms , Lung Neoplasms , Neoadjuvant Therapy , Osteosarcoma , Adolescent , Adult , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Child , Disease-Free Survival , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Neoplasm Metastasis , Osteosarcoma/drug therapy , Osteosarcoma/mortality , Osteosarcoma/pathology , Retrospective Studies , Survival Rate
14.
Nature ; 545(7655): 432-438, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28514439

ABSTRACT

A variety of tissue lineages can be differentiated from pluripotent stem cells by mimicking embryonic development through stepwise exposure to morphogens, or by conversion of one differentiated cell type into another by enforced expression of master transcription factors. Here, to yield functional human haematopoietic stem cells, we perform morphogen-directed differentiation of human pluripotent stem cells into haemogenic endothelium followed by screening of 26 candidate haematopoietic stem-cell-specifying transcription factors for their capacity to promote multi-lineage haematopoietic engraftment in mouse hosts. We recover seven transcription factors (ERG, HOXA5, HOXA9, HOXA10, LCOR, RUNX1 and SPI1) that are sufficient to convert haemogenic endothelium into haematopoietic stem and progenitor cells that engraft myeloid, B and T cells in primary and secondary mouse recipients. Our combined approach of morphogen-driven differentiation and transcription-factor-mediated cell fate conversion produces haematopoietic stem and progenitor cells from pluripotent stem cells and holds promise for modelling haematopoietic disease in humanized mice and for therapeutic strategies in genetic blood disorders.


Subject(s)
Cell Differentiation , Cell Lineage , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Animals , Cellular Reprogramming , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelium/cytology , Female , Hematopoietic Stem Cell Transplantation , Homeobox A10 Proteins , Homeodomain Proteins/metabolism , Humans , Mice , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcriptional Regulator ERG/metabolism
15.
Nature ; 541(7636): 166-167, 2017 01 12.
Article in English | MEDLINE | ID: mdl-27974805
16.
Development ; 143(21): 3956-3968, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27633994

ABSTRACT

During late embryogenesis, mammary epithelial cells initiate migration programs that drive ductal invasion into the surrounding adipose-rich mesenchyme. Currently, branching morphogenesis is thought to depend on the mobilization of the membrane-anchored matrix metalloproteinases MMP14 (MT1-MMP) and MMP15 (MT2-MMP), which drive epithelial cell invasion by remodeling the extracellular matrix and triggering associated signaling cascades. However, the roles that these proteinases play during mammary gland development in vivo remain undefined. Here, we characterize the impact of global Mmp14 and Mmp15 targeting on early postnatal mammary gland development in mice. Unexpectedly, both Mmp14-/- and Mmp15-/- mammary glands retain the ability to generate intact ductal networks. Although neither proteinase is required for branching morphogenesis, transcriptome profiling reveals a key role for MMP14 and MMP15 in regulating mammary gland adipocyte differentiation. Whereas MMP14 promotes the generation of white fat depots crucial for energy storage, MMP15 differentially controls the formation of thermogenic brown fat. Taken together, these data not only indicate that current paradigms relevant to proteinase-dependent morphogenesis need be revisited, but also identify new roles for the enzymes in regulating adipocyte fate determination in the developing mammary gland.


Subject(s)
Mammary Glands, Animal/growth & development , Matrix Metalloproteinase 14/physiology , Matrix Metalloproteinase 15/physiology , Morphogenesis/genetics , Adipocytes/physiology , Adipogenesis/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Energy Metabolism/genetics , Female , Matrix Metalloproteinase 14/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Thermogenesis/genetics
17.
J Exp Med ; 213(8): 1497-512, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27401346

ABSTRACT

For appropriate development, tissue and organ system morphogenesis and maturation must occur in synchrony with the overall developmental requirements of the host. Mistiming of such developmental events often results in disease. The hematopoietic system matures from the fetal state, characterized by robust erythrocytic output that supports prenatal growth in the hypoxic intrauterine environment, to the postnatal state wherein granulocytes predominate to provide innate immunity. Regulation of the developmental timing of these myeloerythroid states is not well understood. In this study, we find that expression of the heterochronic factor Lin28b decreases in common myeloid progenitors during hematopoietic maturation to adulthood in mice. This decrease in Lin28b coincides with accumulation of mature let-7 microRNAs, whose biogenesis is regulated by Lin28 proteins. We find that inhibition of let-7 in the adult hematopoietic system recapitulates fetal erythroid-dominant hematopoiesis. Conversely, deletion of Lin28b or ectopic activation of let-7 microRNAs in the fetal state induces a shift toward adult-like myeloid-dominant output. Furthermore, we identify Hmga2 as an effector of this genetic switch. These studies provide the first detailed analysis of the roles of endogenous Lin28b and let-7 in the timing of hematopoietic states during development.


Subject(s)
DNA-Binding Proteins/metabolism , Erythropoiesis/physiology , Gene Expression Regulation, Developmental/physiology , HMGA2 Protein/biosynthesis , MicroRNAs/metabolism , Myeloid Progenitor Cells/metabolism , Animals , DNA-Binding Proteins/genetics , HMGA2 Protein/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Myeloid Progenitor Cells/cytology , RNA-Binding Proteins
18.
Cell Stem Cell ; 18(6): 707-720, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27257760

ABSTRACT

Cell engineering has brought us tantalizingly close to the goal of deriving patient-specific hematopoietic stem cells (HSCs). While directed differentiation and transcription factor-mediated conversion strategies have generated progenitor cells with multilineage potential, to date, therapy-grade engineered HSCs remain elusive due to insufficient long-term self-renewal and inadequate differentiated progeny functionality. A cross-species approach involving zebrafish and mammalian systems offers complementary methodologies to improve understanding of native HSCs. Here, we discuss the role of conserved developmental timing processes in vertebrate hematopoiesis, highlighting how identification and manipulation of stage-specific factors that specify HSC developmental state must be harnessed to engineer HSCs for therapy.


Subject(s)
Cell Engineering/methods , Embryonic Development , Hematopoietic Stem Cells/cytology , Animals , Hematopoiesis , Humans , Models, Biological , Time Factors
19.
Biol Blood Marrow Transplant ; 22(7): 1275-1283, 2016 07.
Article in English | MEDLINE | ID: mdl-27090959

ABSTRACT

Cytomegalovirus (CMV) infection is a significant source of morbidity and mortality in allogeneic stem cell transplantation (SCT). We identified a cohort of 91 pediatric SCT patients at risk (defined as either donor and/or recipient seropositivity) for CMV infection at our institution. We retrospectively categorized at-risk SCT recipients as those who (1) were at risk of CMV infection in the post-SCT period, (2) had documented CMV infection before SCT, (3) experienced recurrence of post-SCT CMV viremia, or (4) experienced late post-SCT CMV viremia; categories were not mutually exclusive. We analyzed the impact of SCT-related factors on incidence of CMV infection and outcome, and we described the outcome of each of these cohorts. In univariate analysis, recipient CMV seropositivity, use of umbilical cord blood graft, and acute graft-versus-host disease (GVHD) predicted post-SCT CMV viremia, and the effects of acute GVHD (odds ratio, 4.018; 95% confidence interval, 1.032 to 15.643) and CMV seropositivity (odds ratio, 16.525; 95% confidence interval, 2.041 to 133.803) were confirmed in multivariate analysis. Patients with recurrence of post-SCT CMV viremia had a 50% all-cause mortality rate, compared with 12% in all 91 patients. Patients with pre-SCT CMV infection had a high incidence of post-SCT CMV infection but could successfully undergo SCT with antiviral prophylaxis and pre-emptive CMV treatment. All patients with late CMV infection had prior GVHD. Theses findings identify risk factors for post-SCT CMV infection and provide novel descriptions of childhood SCT recipients with pre-SCT, recurrent, and late CMV infection, which may contribute to risk stratification strategies for CMV at-risk patients in pediatric allogeneic SCT.


Subject(s)
Cytomegalovirus Infections/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Adolescent , Adult , Antiviral Agents/therapeutic use , Child , Child, Preschool , Cord Blood Stem Cell Transplantation/adverse effects , Cytomegalovirus Infections/prevention & control , Female , Graft vs Host Disease/prevention & control , Humans , Infant , Male , Premedication/methods , Recurrence , Retrospective Studies , Risk Factors , Time Factors , Viremia/etiology , Viremia/prevention & control , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...