Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Blood ; 144(4): 378-391, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38598841

ABSTRACT

ABSTRACT: Intrinsic molecular programs and extrinsic factors including proinflammatory molecules are understood to regulate hematopoietic aging. This is based on foundational studies using genetic perturbation to evaluate causality. However, individual organisms exhibit natural variation in the hematopoietic aging phenotypes and the molecular basis of this heterogeneity is poorly understood. Here, we generated individual single-cell transcriptomic profiles of hematopoietic and nonhematopoietic cell types in 5 young adult and 9 middle-aged C57BL/6J female mice, providing a web-accessible transcriptomic resource for the field. Among all assessed cell types, hematopoietic stem cells (HSCs) exhibited the greatest phenotypic variation in expansion among individual middle-aged mice. We computationally pooled samples to define modules representing the molecular signatures of middle-aged HSCs and interrogated, which extrinsic regulatory cell types and factors would predict the variance in these signatures between individual middle-aged mice. Decline in signaling mediated by adiponectin, kit ligand (KITL) and insulin-like growth factor 1 (IGF1) from mesenchymal stromal cells (MSCs) was predicted to have the greatest transcriptional impact on middle-aged HSCs, as opposed to signaling mediated by endothelial cells or mature hematopoietic cell types. In individual middle-aged mice, lower expression of Kitl and Igf1 in MSCs was highly correlated with reduced lymphoid lineage commitment of HSCs and increased signatures of differentiation-inactive HSCs. These signatures were independent of expression of aging-associated proinflammatory cytokines including interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor α and RANTES. In sum, we find that Kitl and Igf1 expression are coregulated and variable between individual mice at the middle age and expression of these factors is predictive of HSC activation and lymphoid commitment independently of inflammation.


Subject(s)
Cellular Senescence , Hematopoietic Stem Cells , Insulin-Like Growth Factor I , Stem Cell Factor , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice , Female , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/genetics , Stem Cell Factor/metabolism , Stem Cell Factor/genetics , Aging/metabolism , Aging/genetics , Mice, Inbred C57BL , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Transcriptome
2.
Blood ; 120(20): 4263-73, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23018639

ABSTRACT

BMP9 signaling has been implicated in hereditary hemorrhagic telangiectasia (HHT) and vascular remodeling, acting via the HHT target genes, endoglin and ALK1. This study sought to identify endothelial BMP9-regulated proteins that could affect the HHT phenotype. Gene ontology analysis of cDNA microarray data obtained after BMP9 treatment of primary human endothelial cells indicated regulation of chemokine, adhesion, and inflammation pathways. These responses included the up-regulation of the chemokine CXCL12/SDF1 and down-regulation of its receptor CXCR4. Quantitative mass spectrometry identified additional secreted proteins, including the chemokine CXCL10/IP10. RNA knockdown of endoglin and ALK1 impaired SDF1/CXCR4 regulation by BMP9. Because of the association of SDF1 with ischemia, we analyzed its expression under hypoxia in response to BMP9 in vitro, and during the response to hindlimb ischemia, in endoglin-deficient mice. BMP9 and hypoxia were additive inducers of SDF1 expression. Moreover, the data suggest that endoglin deficiency impaired SDF1 expression in endothelial cells in vivo. Our data implicate BMP9 in regulation of the SDF1/CXCR4 chemokine axis in endothelial cells and point to a role for BMP9 signaling via endoglin in a switch from an SDF1-responsive autocrine phenotype to an SDF1 nonresponsive paracrine state that represses endothelial cell migration and may promote vessel maturation.


Subject(s)
Endothelial Cells/cytology , Growth Differentiation Factors/physiology , Neovascularization, Physiologic/physiology , Activin Receptors, Type I/physiology , Activin Receptors, Type II/physiology , Animals , Antigens, CD/physiology , Aorta/cytology , Autocrine Communication , Cell Hypoxia , Cell Movement , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/metabolism , Culture Media, Conditioned , Endoglin , Endothelial Cells/drug effects , Growth Differentiation Factor 2/pharmacology , Growth Differentiation Factor 2/physiology , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Ischemia/physiopathology , Mice , Paracrine Communication , RNA, Messenger/biosynthesis , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/physiology , Transforming Growth Factor beta1/pharmacology
3.
Exp Hematol ; 130: 104131, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38000729

ABSTRACT

Age-associated clonal hematopoiesis (CH) occurs due to somatic mutations accrued in hematopoietic stem cells (HSCs) that confer a selective growth advantage in the context of aging. The mechanisms by which CH-mutant HSCs gain this advantage with aging are not comprehensively understood. Using unbiased transcriptomic approaches, we identified Oncostatin M (OSM) signaling as a candidate contributor to age-related Dnmt3a-mutant CH. We found that Dnmt3a-mutant HSCs from young adult mice (3-6 months old) subjected to acute OSM stimulation do not demonstrate altered proliferation, apoptosis, hematopoietic engraftment, or myeloid differentiation. Dnmt3a-mutant HSCs from young mice do transcriptionally upregulate an inflammatory cytokine network in response to acute in vitro OSM stimulation as evidenced by significant upregulation of the genes encoding IL-6, IL-1ß, and TNFα. OSM-stimulated Dnmt3a-mutant HSCs also demonstrate upregulation of the anti-inflammatory genes Socs3, Atf3, and Nr4a1. In the context of an aged bone marrow (BM) microenvironment, Dnmt3a-mutant HSCs upregulate proinflammatory genes but not the anti-inflammatory genes Socs3, Atf3, and Nr4a1. The results from our studies suggest that aging may exhaust the regulatory mechanisms that HSCs employ to resolve inflammatory states in response to factors such as OSM.


Subject(s)
Bone Marrow , Hematopoietic Stem Cells , Animals , Mice , Anti-Inflammatory Agents , Hematopoiesis/genetics , Oncostatin M/genetics
4.
bioRxiv ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38585779

ABSTRACT

Clonal hematopoiesis (CH) can predispose to blood cancers due to enhanced fitness of mutant hematopoietic stem and progenitor cells (HSPCs), but the mechanisms driving this progression are not understood. We hypothesized that malignant progression is related to microenvironment-remodelling properties of CH-mutant HSPCs. Single-cell transcriptomic profiling of the bone marrow microenvironment in Dnmt3a R878H/+ mice revealed signatures of cellular senescence in mesenchymal stromal cells (MSCs). Dnmt3a R878H/+ HSPCs caused MSCs to upregulate the senescence markers SA-ß-gal, BCL-2, BCL-xL, Cdkn1a (p21) and Cdkn2a (p16), ex vivo and in vivo . This effect was cell contact-independent and can be replicated by IL-6 or TNFα, which are produced by Dnmt3a R878H/+ HSPCs. Depletion of senescent MSCs in vivo reduced the fitness of Dnmt3a R878H/+ hematopoietic cells and the progression of CH to myeloid neoplasms using a sequentially inducible Dnmt3a ; Npm1 -mutant model. Thus, Dnmt3a -mutant HSPCs reprogram their microenvironment via senescence induction, creating a self-reinforcing niche favoring fitness and malignant progression. Statement of Significance: Mesenchymal stromal cell senescence induced by Dnmt3a -mutant hematopoietic stem and progenitor cells drives clonal hematopoiesis and initiation of hematologic malignancy.

5.
Res Sq ; 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38405837

ABSTRACT

Clonal hematopoiesis (CH) arises when a hematopoietic stem cell (HSC) acquires a mutation that confers a competitive advantage over wild-type (WT) HSCs, resulting in its clonal expansion. Individuals with CH are at an increased risk of developing hematologic neoplasms and a range of age-related inflammatory illnesses1-3. Therapeutic interventions that suppress the expansion of mutant HSCs have the potential to prevent these CH-related illnesses; however, such interventions have not yet been identified. The most common CH driver mutations are in the DNA methyltransferase 3 alpha (DNMT3A) gene with arginine 882 (R882) being a mutation hotspot. Here we show that murine hematopoietic stem and progenitor cells (HSPCs) carrying the Dnmt3aR878H/+ mutation, which is equivalent to human DNMT3AR882H/+, have increased mitochondrial respiration compared with WT cells and are dependent on this metabolic reprogramming for their competitive advantage. Importantly, treatment with metformin, an oral anti-diabetic drug with inhibitory activity against complex I in the electron transport chain (ETC), reduced the fitness of Dnmt3aR878H/+ HSCs. Through a multi-omics approach, we discovered that metformin acts by enhancing the methylation potential in Dnmt3aR878H/+ HSPCs and reversing their aberrant DNA CpG methylation and histone H3K27 trimethylation (H3K27me3) profiles. Metformin also reduced the fitness of human DNMT3AR882H HSPCs generated by prime editing. Our findings provide preclinical rationale for investigating metformin as a preventive intervention against illnesses associated with DNMT3AR882 mutation-driven CH in humans.

6.
Microorganisms ; 11(7)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37512873

ABSTRACT

Two formulations of Alltech Crop Science products (ACS), a proprietary blend of fermentation products and plant extracts with micronutrients (ACS5075), and a microbial based product (ACS3048), were tested to understand (1) their impact on the tomato plant immune response and (2) whether they are priming a resistance response in plants against root knot nematodes (RKN). Research findings reported previously indicate that tomato plants pre-treated with ACS5075 and ACS3048 were found less sensitive to Meloidogyne javanica infection. In the current study, the expression of six defence-related genes (PR-1, PR-3, PR-5T, ACO, CAT and JERF 3), relative to a housekeeping gene, were monitored via RT-PCR. Results suggest that the treatment with ACS5075 enhanced ACO and PR-1 gene expression levels, both post- treatment and post-infection with M. javanica. Reduced M. javanica infestation that was reported in the previous study could be attributed to the increased expression of these genes in the ACS5075-treated plants. Tomato plants treated with ACS3048, but without RKN infection, also demonstrated higher levels of ACO and PR-1 gene expression. Subsequently, 2D-gel electrophoresis was performed to study the differential protein expression in leaf tissues of treated tomato plants in an effort to elucidate a possible mechanism of action for these products. Protein spot 1 was identified as 'disease resistance protein RPP13-like', protein spot 2 as 'phosphatidylinositol 4-phosphate 5-kinase 2', spot 3 as 'protein SABRE like' and protein spot 4 as 'uncharacterized protein'. Overall research findings indicate that the ACS products could be used as plant immunity-boosting agents, as they play a significant role in the expression of certain genes and proteins associated with plant defence.

7.
bioRxiv ; 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37502912

ABSTRACT

Age-associated clonal hematopoiesis (CH) occurs due to somatic mutations accrued in hematopoietic stem cells (HSCs) that confer a selective advantage in the context of aging. The mechanisms by which CH-mutant HSCs gain this advantage with aging are not comprehensively understood. Using unbiased transcriptomic approaches, we identify Oncostatin M (OSM) signaling as a candidate contributor to aging-driven Dnmt3a -mutant CH. We find that Dnmt3a -mutant HSCs from young mice do not functionally respond to acute OSM stimulation with respect to proliferation, apoptosis, hematopoietic engraftment, or myeloid differentiation. However, young Dnmt3a -mutant HSCs transcriptionally upregulate an inflammatory cytokine network in response to acute OSM stimulation including genes encoding IL-6, IL-1ß and TNFα. In addition, OSM-stimulated Dnmt3a -mutant HSCs upregulate the anti-inflammatory genes Socs3, Atf3 and Nr4a1 , creating a negative feedback loop limiting sustained activation of the inflammatory network. In the context of an aged bone marrow (BM) microenvironment with chronically elevated levels of OSM, Dnmt3a -mutant HSCs upregulate pro-inflammatory genes but do not upregulate Socs3, Atf3 and Nr4a1 . Together, our work suggests that chronic inflammation with aging exhausts the regulatory mechanisms in young CH-mutant HSCs that resolve inflammatory states, and that OSM is a master regulator of an inflammatory network that contributes to age-associated CH.

8.
Cancer Discov ; 12(12): 2763-2773, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36169447

ABSTRACT

Clonal hematopoiesis resulting from the enhanced fitness of mutant hematopoietic stem cells (HSC) associates with both favorable and unfavorable health outcomes related to the types of mature mutant blood cells produced, but how this lineage output is regulated is unclear. Using a mouse model of a clonal hematopoiesis-associated mutation, DNMT3AR882/+ (Dnmt3aR878H/+), we found that aging-induced TNFα signaling promoted the selective advantage of mutant HSCs and stimulated the production of mutant B lymphoid cells. The genetic loss of the TNFα receptor TNFR1 ablated the selective advantage of mutant HSCs without altering their lineage output, whereas the loss of TNFR2 resulted in the overproduction of mutant myeloid cells without altering HSC fitness. These results nominate TNFR1 as a target to reduce clonal hematopoiesis and the risk of associated diseases and support a model in which clone size and mature blood lineage production can be independently controlled to modulate favorable and unfavorable clonal hematopoiesis outcomes. SIGNIFICANCE: Through the identification and dissection of TNFα signaling as a key driver of murine Dnmt3a-mutant hematopoiesis, we report the discovery that clone size and production of specific mature blood cell types can be independently regulated. See related commentary by Niño and Pietras, p. 2724. This article is highlighted in the In This Issue feature, p. 2711.


Subject(s)
Clonal Hematopoiesis , Receptors, Tumor Necrosis Factor, Type I , Animals , Mice , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Cell Lineage/genetics
9.
Cell Stem Cell ; 28(8): 1473-1482.e7, 2021 08 05.
Article in English | MEDLINE | ID: mdl-33848471

ABSTRACT

Decline in hematopoietic stem cell (HSC) function with age underlies limited health span of our blood and immune systems. In order to preserve health into older age, it is necessary to understand the nature and timing of initiating events that cause HSC aging. By performing a cross-sectional study in mice, we discover that hallmarks of aging in HSCs and hematopoiesis begin to accumulate by middle age and that the bone marrow (BM) microenvironment at middle age induces and is indispensable for hematopoietic aging. Using unbiased approaches, we find that decreased levels of the longevity-associated molecule IGF1 in the local middle-aged BM microenvironment are a factor causing HSC aging. Direct stimulation of middle-aged HSCs with IGF1 rescues molecular and functional hallmarks of aging, including restored mitochondrial activity. Thus, although decline in IGF1 supports longevity, our work indicates that this also compromises HSC function and limits hematopoietic health span.


Subject(s)
Bone Marrow , Stem Cell Niche , Aging , Animals , Cross-Sectional Studies , Hematopoiesis , Hematopoietic Stem Cells , Mice
10.
Biophys J ; 99(3): 897-904, 2010 Aug 04.
Article in English | MEDLINE | ID: mdl-20682268

ABSTRACT

Bone morphogenetic proteins (BMPs) play a crucial role during embryonic development and regulate processes as diverse as neurogenesis, skeletal formation, and hematopoesis. They signal through a hetero-oligomer complex of BMP receptors. Binding of the ligand to the receptors activates several pathways, including Smad and p38. BMP signaling is controlled in the extracellular space, the plasma membrane, and the intracellular space; however, the mechanism of receptor signaling at the plasma membrane and proteins that regulate this process still need to be identified. The experiments presented here identify the protein kinase casein kinase II (CK2) as a BMP receptor type Ia (BRIa) interacting protein. Fluorescence resonance energy transfer revealed that this interaction occurs at the plasma membrane. BMP2 stimulation of C2C12 cells leads to the release of CK2 from BRIa. Blocking this interaction with specific peptides that inhibit the binding sites for CK2 on BRIa demonstrated a redistribution of BRIa on the plasma membrane. Signaling was initiated once CK2 was released from BRIa, leading to the mineralization of C2C12 cells. These data suggest that CK2 is a negative regulator of BMP signaling and osteoblast differentiation.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Casein Kinase II/metabolism , Signal Transduction , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Calcification, Physiologic/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Genes, Dominant/genetics , Mice , Models, Biological , Peptides/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects , Smad Proteins/metabolism
11.
Exp Hematol ; 91: 1-9, 2020 11.
Article in English | MEDLINE | ID: mdl-32991978

ABSTRACT

Over the past 25 years, the importance of hematopoietic stem cell (HSC) aging in overall hematopoietic and immune system health span has been appreciated. Much work has been done in model organisms to understand the intrinsic dysregulation that occurs in HSCs during aging, with the goal of identifying modifiable mechanisms that represent the proverbial "fountain of youth." Much more recently, the discovery of somatic mutations that are found to provide a selective advantage to HSCs and accumulate in the hematopoietic system during aging, termed clonal hematopoiesis (CH), inspires revisiting many of these previously defined drivers of HSC aging in the context of these somatic mutations. To truly understand these processes and develop a holistic picture of HSC aging, ongoing and future studies must include investigation of the critical changes that occur in the HSC niche or bone marrow microenvironment with aging, as increasing evidence supports that these HSC-extrinsic alterations provide necessary inflammation, signaling pathway activation or repression, and other selective pressures to favor HSC aging-associated phenotypes and CH. Here, we provide our perspectives based on the past 8 years of our own laboratory's investigations into these mechanisms and chart a path for integrative studies that, in our opinion, will provide an ideal opportunity to discover HSC and hematopoietic health span-extending interventions. This path includes examining when and how aging-associated HSC-intrinsic and HSC-extrinsic changes accumulate over time in different individuals and developing new models to track and test relevant HSC-extrinsic changes, complementary to innovative HSC lineage tracing systems that have recently been developed.


Subject(s)
Aging/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Aging/genetics , Animals , Antineoplastic Agents/pharmacology , Bone Marrow/growth & development , Cellular Senescence/genetics , Cellular Senescence/physiology , Chromatin/genetics , Chromatin/ultrastructure , Clone Cells , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Damage , DNA Methylation , DNA Methyltransferase 3A , Feedback, Physiological , Female , Forecasting , Hematopoietic Stem Cells/classification , Humans , Inflammation/genetics , Male , Mice , Mutation , Myeloid Cells/cytology , Selection, Genetic , Stem Cell Niche
12.
Exp Hematol ; 82: 43-52.e4, 2020 02.
Article in English | MEDLINE | ID: mdl-32014431

ABSTRACT

Aged hematopoietic stem cells (HSCs) undergo biased lineage priming and differentiation toward production of myeloid cells. A comprehensive understanding of gene regulatory mechanisms causing HSC aging is needed to devise new strategies to sustainably improve immune function in aged individuals. Here, a focused short hairpin RNA screen of epigenetic factors reveals that the histone acetyltransferase Kat6b regulates myeloid cell production from hematopoietic progenitor cells. Within the stem and progenitor cell compartment, Kat6b is highly expressed in long-term (LT)-HSCs and is significantly decreased with aging at the transcript and protein levels. Knockdown of Kat6b in young LT-HSCs causes skewed production of myeloid cells at the expense of erythroid cells both in vitro and in vivo. Transcriptome analysis identifies enrichment of aging and macrophage-associated gene signatures alongside reduced expression of self-renewal and multilineage priming signatures. Together, our work identifies KAT6B as a novel epigenetic regulator of hematopoietic differentiation and a target to improve aged immune function.


Subject(s)
Aging/metabolism , Cell Differentiation , Erythroid Cells/enzymology , Gene Expression Regulation, Enzymologic , Histone Acetyltransferases/biosynthesis , Myeloid Progenitor Cells/enzymology , Aging/genetics , Aging/pathology , Animals , Epigenesis, Genetic , Erythroid Cells/pathology , Gene Expression Profiling , Gene Knockout Techniques , Histone Acetyltransferases/genetics , Male , Mice , Mice, Transgenic , Myeloid Progenitor Cells/pathology , Transcriptome
13.
Exp Hematol ; 89: 61-67.e3, 2020 09.
Article in English | MEDLINE | ID: mdl-32768438

ABSTRACT

The MLL-AF9 fusion protein occurring as a result of t(9;11) translocation gives rise to pediatric and adult acute leukemias of distinct lineages, including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and mixed-phenotype acute leukemia (MPAL). The mechanisms underlying how this same fusion protein results in diverse leukemia phenotypes among different individuals are not well understood. Given emerging evidence from genome-wide association studies that genetic risk factors contribute to MLL-rearranged leukemogenesis, here we tested the impact of genetic background on survival and phenotype of a well-characterized Mll-AF9 knockin mouse model. We crossed this model with five distinct inbred strains (129, A/J, C57BL/6, NOD, CAST) and tested their F1 hybrid progeny for dominant genetic effects on Mll-AF9 phenotypes. We discovered that genetic background altered peripheral blood composition, with Mll-AF9 CAST F1 having a significantly increased B-lymphocyte frequency, while the remainder of the strains exhibited myeloid-biased hematopoiesis, similar to the parental line. Genetic background also had an impact on overall survival, with Mll-AF9 A/J F1 and Mll-AF9 129 F1 having significantly shorter survival and Mll-AF9 CAST F1 having longer survival, compared with the parental line. Furthermore, we observed a range of hematologic malignancies, with Mll-AF9 A/J F1, Mll-AF9 129 F1, and Mll-AF9 B6 F1 developing exclusively myeloid cell malignancies (myeloproliferative disorder [MPD] and AML), whereas a subset of Mll-AF9 NOD F1 developed MPAL and Mll-AF9 CAST F1 developed ALL. This study provides a novel in vivo experimental model in which to evaluate the underlying mechanisms by which MLL-AF9 results in diverse leukemia phenotypes and provides definitive experimental evidence that genetic risk factors contribute to survival and phenotype of MLL-rearranged leukemogenesis.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Leukemic , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Myeloid, Acute/genetics , Myeloproliferative Disorders/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Lineage/genetics , Disease Progression , Female , Gene Knock-In Techniques , Genetic Predisposition to Disease , Humans , Leukemia, Biphenotypic, Acute/metabolism , Leukemia, Biphenotypic, Acute/mortality , Leukemia, Biphenotypic, Acute/pathology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Lymphocyte Count , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/pathology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/mortality , Myeloproliferative Disorders/pathology , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Survival Analysis
14.
Leukemia ; 33(7): 1635-1649, 2019 07.
Article in English | MEDLINE | ID: mdl-30692594

ABSTRACT

Clonal hematopoiesis (CH) is a common aging-associated condition with increased risk of hematologic malignancy. Knowledge of the mechanisms driving evolution from CH to overt malignancy has been hampered by a lack of in vivo models that orthogonally activate mutant alleles. Here, we develop independently regulatable mutations in DNA methyltransferase 3A (Dnmt3a) and nucleophosmin 1 (Npm1), observed in human CH and AML, respectively. We find Dnmt3a mutation expands hematopoietic stem and multipotent progenitor cells (HSC/MPPs), modeling CH. Induction of mutant Npm1 after development of Dnmt3a-mutant CH causes progression to myeloproliferative disorder (MPD), and more aggressive MPD is observed with longer latency between mutations. MPDs uniformly progress to acute myeloid leukemia (AML) following transplant, accompanied by a decrease in HSC/MPPs and an increase in myeloid-restricted progenitors, the latter of which propagate AML in tertiary recipient mice. At a molecular level, progression of CH to MPD is accompanied by selection for mutations activating Ras/Raf/MAPK signaling. Progression to AML is characterized by additional oncogenic signaling mutations (Ptpn11, Pik3r1, Flt3) and/or mutations in epigenetic regulators (Hdac1, Idh1, Arid1a). Together, our study demonstrates that Npm1 mutation drives evolution of Dnmt3a-mutant CH to AML and rate of disease progression is accelerated with longer latency of CH.


Subject(s)
Cell Transformation, Neoplastic/pathology , Clonal Evolution , DNA (Cytosine-5-)-Methyltransferases/genetics , Disease Models, Animal , Leukemia, Myeloid, Acute/etiology , Mutation , Myeloproliferative Disorders/pathology , Nuclear Proteins/genetics , Animals , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methyltransferase 3A , Disease Progression , Female , Hematopoiesis , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Progenitor Cells/pathology , Myeloid Progenitor Cells/transplantation , Myeloproliferative Disorders/genetics , Nuclear Proteins/physiology , Nucleophosmin
15.
Mol Cell Oncol ; 3(6): e1236770, 2016.
Article in English | MEDLINE | ID: mdl-28090579

ABSTRACT

Tumor cell of origin is an important prognostic measure but is challenging to assess. We recently demonstrated in acute myeloid leukemia (AML) that the chromatin landscape serves as a biomarker of transformed cell of origin. Thus, open chromatin loci offer important prognostic information as well as targets for development of novel therapies in cancer treatment.

16.
J Exp Med ; 213(11): 2259-2267, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27811054

ABSTRACT

Declining immune function with age is associated with reduced lymphoid output of hematopoietic stem cells (HSCs). Currently, there is poor understanding of changes with age in the heterogeneous multipotent progenitor (MPP) cell compartment, which is long lived and responsible for dynamically regulating output of mature hematopoietic cells. In this study, we observe an early and progressive loss of lymphoid-primed MPP cells (LMPP/MPP4) with aging, concomitant with expansion of HSCs. Transcriptome and in vitro functional analyses at the single-cell level reveal a concurrent increase in cycling of aging LMPP/MPP4 with loss of lymphoid priming and differentiation potential. Impaired lymphoid differentiation potential of aged LMPP/MPP4 is not rescued by transplantation into a young bone marrow microenvironment, demonstrating cell-autonomous changes in the MPP compartment with aging. These results pinpoint an age and cellular compartment to focus further interrogation of the drivers of lymphoid cell loss with aging.


Subject(s)
Aging/physiology , Hematopoietic Stem Cells/cytology , Lymphocytes/cytology , Multipotent Stem Cells/cytology , Animals , Cell Cycle/genetics , Cell Differentiation , Cell Lineage/genetics , Cells, Cultured , Cellular Senescence/genetics , Female , Hematopoietic Stem Cells/metabolism , Lymphocytes/metabolism , Mice, Inbred C57BL , Multipotent Stem Cells/metabolism , Myeloid Cells/cytology , Single-Cell Analysis , Transcriptome/genetics
17.
Nat Commun ; 7: 12166, 2016 07 11.
Article in English | MEDLINE | ID: mdl-27397025

ABSTRACT

The precise identity of a tumour's cell of origin can influence disease prognosis and outcome. Methods to reliably define tumour cell of origin from primary, bulk tumour cell samples has been a challenge. Here we use a well-defined model of MLL-rearranged acute myeloid leukaemia (AML) to demonstrate that transforming haematopoietic stem cells (HSCs) and multipotent progenitors results in more aggressive AML than transforming committed progenitor cells. Transcriptome profiling reveals a gene expression signature broadly distinguishing stem cell-derived versus progenitor cell-derived AML, including genes involved in immune escape, extravasation and small GTPase signal transduction. However, whole-genome profiling of open chromatin reveals precise and robust biomarkers reflecting each cell of origin tested, from bulk AML tumour cell sampling. We find that bulk AML tumour cells exhibit distinct open chromatin loci that reflect the transformed cell of origin and suggest that open chromatin patterns may be leveraged as prognostic signatures in human AML.


Subject(s)
Chromatin Assembly and Disassembly , Leukemia, Myeloid, Acute/etiology , Animals , Cell Transformation, Neoplastic , Epigenesis, Genetic , Female , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Mice, Inbred C57BL , Multipotent Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Transcriptome
18.
PLoS One ; 10(4): e0122892, 2015.
Article in English | MEDLINE | ID: mdl-25909848

ABSTRACT

Endoglin is a type III TGFß auxiliary receptor that is upregulated in endothelial cells during angiogenesis and, when mutated in humans, results in the vascular disease hereditary hemorrhagic telangiectasia (HHT). Though endoglin has been implicated in cell adhesion, the underlying molecular mechanisms are still poorly understood. Here we show endoglin expression in endothelial cells regulates subcellular localization of zyxin in focal adhesions in response to BMP9. RNA knockdown of endoglin resulted in mislocalization of zyxin and altered formation of focal adhesions. The mechanotransduction role of focal adhesions and their ability to transmit regulatory signals through binding of the extracellular matrix are altered by endoglin deficiency. BMP/TGFß transcription factors, SMADs, and zyxin have recently been implicated in a newly emerging signaling cascade, the Hippo pathway. The Hippo transcription coactivator, YAP1 (yes-associated protein 1), has been suggested to play a crucial role in mechanotransduction and cell-cell contact. Identification of BMP9-dependent nuclear localization of YAP1 in response to endoglin expression suggests a mechanism of crosstalk between the two pathways. Suppression of endoglin and YAP1 alters BMP9-dependent expression of YAP1 target genes CCN1 (cysteine-rich 61, CYR61) and CCN2 (connective tissue growth factor, CTGF) as well as the chemokine CCL2 (monocyte chemotactic protein 1, MCP-1). These results suggest a coordinate effect of endoglin deficiency on cell matrix remodeling and local inflammatory responses. Identification of a direct link between the Hippo pathway and endoglin may reveal novel mechanisms in the etiology of HHT.


Subject(s)
Chemokines/metabolism , Endothelial Cells/metabolism , Extracellular Matrix/metabolism , Growth Differentiation Factors/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD/metabolism , Chemokine CCL2/metabolism , Connective Tissue Growth Factor/metabolism , Cysteine-Rich Protein 61/metabolism , Endoglin , Focal Adhesions/metabolism , Growth Differentiation Factor 2 , Hippo Signaling Pathway , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Models, Biological , Phosphoproteins/metabolism , Receptors, CCR2/metabolism , Receptors, Cell Surface/metabolism , Smad Proteins/metabolism , Transcription Factors , YAP-Signaling Proteins , Zyxin/metabolism
19.
Cancer Res ; 71(10): 3482-93, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21444673

ABSTRACT

Endoglin is an accessory receptor for TGF-ß that has been implicated in prostate cancer cell detachment, migration, and invasiveness. However, the pathophysiologic significance of endoglin with respect to prostate tumorigenesis has yet to be fully established. In this study, we addressed this question by investigation of endoglin-dependent prostate cancer progression in a TRAMP (transgenic adenocarcinoma mouse prostate) mouse model where endoglin was genetically deleted. In this model, endoglin was haploinsufficient such that its allelic deletion slightly increased the frequency of tumorigenesis, yet produced smaller, less vascularized, and less metastatic tumors than TRAMP control tumors. Most strikingly, TRAMP:eng(+/-)-derived tumors lacked the pronounced infiltration of carcinoma-associated fibroblasts (CAF) that characterize TRAMP prostate tumors. Studies in human primary prostate-derived stromal cells (PrSC) confirmed that suppressing endoglin expression decreased cell proliferation, the ability to recruit endothelial cells, and the ability to migrate in response to tumor cell-conditioned medium. We found increased levels of secreted insulin-like growth factor-binding proteins (IGFBP) in the conditioned medium from endoglin-deficient PrSCs and that endoglin-dependent regulation of IGFBP-4 secretion was crucial for stromal cell-conditioned media to stimulate prostate tumor cell growth. Together, our results firmly establish the pathophysiologic involvement of endoglin in prostate cancer progression; furthermore, they show how endoglin acts to support the viability of tumor-infiltrating CAFs in the tumor microenvironment to promote neovascularization and growth.


Subject(s)
Antigens, CD/physiology , Gene Expression Regulation, Neoplastic/genetics , Intracellular Signaling Peptides and Proteins/physiology , Prostatic Neoplasms/genetics , Receptors, Cell Surface/physiology , Stromal Cells/cytology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Endoglin , Fibroblasts/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Prostatic Neoplasms/metabolism , RNA, Small Interfering/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL