Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 56
1.
Front Immunol ; 13: 934624, 2022.
Article En | MEDLINE | ID: mdl-35990659

Adult mammalian hematopoiesis is a dynamic cellular process that provides a continuous supply of myeloid, lymphoid, erythroid/megakaryocyte cells for host survival. This process is sustained by regulating hematopoietic stem cells (HSCs) quiescence, proliferation and activation under homeostasis and stress, and regulating the proliferation and differentiation of downstream multipotent progenitor (MPP) and more committed progenitor cells. Inhibitor of DNA binding (ID) proteins are small helix-loop-helix (HLH) proteins that lack a basic (b) DNA binding domain present in other family members, and function as dominant-negative regulators of other bHLH proteins (E proteins) by inhibiting their transcriptional activity. ID proteins are required for normal T cell, B cell, NK and innate lymphoid cells, dendritic cell, and myeloid cell differentiation and development. However, recent evidence suggests that ID proteins are important regulators of normal and leukemic hematopoietic stem and progenitor cells (HSPCs). This chapter will review our current understanding of the function of ID proteins in HSPC development and highlight future areas of scientific investigation.


Immunity, Innate , Lymphocytes , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA , Hematopoiesis/genetics , Lymphocytes/metabolism , Mammals/genetics
2.
J Clin Invest ; 132(13)2022 07 01.
Article En | MEDLINE | ID: mdl-35775482

Defining mechanism(s) that maintain tissue stem quiescence is important for improving tissue regeneration, cell therapies, aging, and cancer. We report here that genetic ablation of Id2 in adult hematopoietic stem cells (HSCs) promotes increased HSC activation and differentiation, which results in HSC exhaustion and bone marrow failure over time. Id2Δ/Δ HSCs showed increased cycling, ROS production, mitochondrial activation, ATP production, and DNA damage compared with Id2+/+ HSCs, supporting the conclusion that Id2Δ/Δ HSCs are less quiescent. Mechanistically, HIF-1α expression was decreased in Id2Δ/Δ HSCs, and stabilization of HIF-1α in Id2Δ/Δ HSCs restored HSC quiescence and rescued HSC exhaustion. Inhibitor of DNA binding 2 (ID2) promoted HIF-1α expression by binding to the von Hippel-Lindau (VHL) protein and interfering with proteasomal degradation of HIF-1α. HIF-1α promoted Id2 expression and enforced a positive feedback loop between ID2 and HIF-1α to maintain HSC quiescence. Thus, sustained ID2 expression could protect HSCs during stress and improve HSC expansion for gene editing and cell therapies.


Hematopoietic Stem Cells , Mitochondria , Hematopoietic Stem Cells/metabolism , Mitochondria/metabolism
3.
Nat Commun ; 13(1): 1751, 2022 04 01.
Article En | MEDLINE | ID: mdl-35365640

The interaction between tumor suppressor BRCA2 and DSS1 is essential for RAD51 recruitment and repair of DNA double stand breaks (DSBs) by homologous recombination (HR). We have generated mice with a leucine to proline substitution at position 2431 of BRCA2, which disrupts this interaction. Although a significant number of mutant mice die during embryogenesis, some homozygous and hemizygous mutant mice undergo normal postnatal development. Despite lack of radiation induced RAD51 foci formation and a severe HR defect in somatic cells, mutant mice are fertile and exhibit normal RAD51 recruitment during meiosis. We hypothesize that the presence of homologous chromosomes in close proximity during early prophase I may compensate for the defect in BRCA2-DSS1 interaction. We show the restoration of RAD51 foci in mutant cells when Topoisomerase I inhibitor-induced single strand breaks are converted into DSBs during DNA replication. We also partially rescue the HR defect by tethering the donor DNA to the site of DSBs using streptavidin-fused Cas9. Our findings demonstrate that the BRCA2-DSS1 complex is dispensable for RAD51 loading when the homologous DNA is close to the DSB.


DNA Breaks, Double-Stranded , Rad51 Recombinase , Animals , DNA , DNA Repair/genetics , Homologous Recombination , Mice , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism
5.
J Biol Chem ; 296: 100491, 2021.
Article En | MEDLINE | ID: mdl-33662400

Serine palmitoyltransferase complex (SPT) mediates the first and rate-limiting step in the de novo sphingolipid biosynthetic pathway. The larger subunits SPTLC1 and SPTLC2/SPTLC3 together form the catalytic core while a smaller third subunit either SSSPTA or SSSPTB has been shown to increase the catalytic efficiency and provide substrate specificity for the fatty acyl-CoA substrates. The in vivo biological significance of these smaller subunits in mammals is still unknown. Here, using two null mutants, a conditional null for ssSPTa and a null mutant for ssSPTb, we show that SSSPTA is essential for embryogenesis and mediates much of the known functions of the SPT complex in mammalian hematopoiesis. The ssSPTa null mutants are embryonic lethal at E6.5 much like the Sptlc1 and Sptlc2 null alleles. Mx1-Cre induced deletion of ssSPTa leads to lethality and myelopoietic defect. Chimeric and competitive bone marrow transplantation experiments show that the defect in myelopoiesis is accompanied by an expansion of the Lin-Sca1+c-Kit+ stem and progenitor compartment. Progenitor cells that fail to differentiate along the myeloid lineage display evidence of endoplasmic reticulum stress. On the other hand, ssSPTb null mice are homozygous viable, and analyses of the bone marrow cells show no significant difference in the proliferation and differentiation of the adult hematopoietic compartment. SPTLC1 is an obligatory subunit for the SPT function, and because Sptlc1-/- and ssSPTa-/- mice display similar defects during development and hematopoiesis, we conclude that an SPT complex that includes SSSPTA mediates much of its developmental and hematopoietic functions in a mammalian model.


Acyl Coenzyme A/metabolism , Bone Marrow Cells/cytology , Hematopoiesis/physiology , Serine C-Palmitoyltransferase/genetics , Sphingolipids/biosynthesis , Animals , Bone Marrow Cells/metabolism , Catalytic Domain , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Serine C-Palmitoyltransferase/metabolism , Substrate Specificity
6.
Nat Commun ; 11(1): 5836, 2020 11 17.
Article En | MEDLINE | ID: mdl-33203851

Several genes implicated in autism spectrum disorder (ASD) are chromatin regulators, including POGZ. The cellular and molecular mechanisms leading to ASD impaired social and cognitive behavior are unclear. Animal models are crucial for studying the effects of mutations on brain function and behavior as well as unveiling the underlying mechanisms. Here, we generate a brain specific conditional knockout mouse model deficient for Pogz, an ASD risk gene. We demonstrate that Pogz deficient mice show microcephaly, growth impairment, increased sociability, learning and motor deficits, mimicking several of the human symptoms. At the molecular level, luciferase reporter assay indicates that POGZ is a negative regulator of transcription. In accordance, in Pogz deficient mice we find a significant upregulation of gene expression, most notably in the cerebellum. Gene set enrichment analysis revealed that the transcriptional changes encompass genes and pathways disrupted in ASD, including neurogenesis and synaptic processes, underlying the observed behavioral phenotype in mice. Physiologically, Pogz deficiency is associated with a reduction in the firing frequency of simple and complex spikes and an increase in amplitude of the inhibitory synaptic input in cerebellar Purkinje cells. Our findings support a mechanism linking heterochromatin dysregulation to cerebellar circuit dysfunction and behavioral abnormalities in ASD.


Autism Spectrum Disorder/etiology , Behavior, Animal , Brain/physiopathology , DNA Transposable Elements/genetics , Purkinje Cells/physiology , Transposases/metabolism , Animals , Autism Spectrum Disorder/genetics , Brain/anatomy & histology , Brain/embryology , Brain/growth & development , Cognition Disorders/genetics , Disease Models, Animal , Female , Gene Expression Regulation , HEK293 Cells , Humans , Learning , Male , Mice, Inbred ICR , Mice, Knockout , Microcephaly/genetics , Motor Activity/genetics , Neurogenesis/genetics , Pregnancy , Purkinje Cells/pathology , Social Behavior , Transcription, Genetic , Transposases/deficiency
7.
Curr Opin Hematol ; 27(4): 225-231, 2020 07.
Article En | MEDLINE | ID: mdl-32398455

PURPOSE OF REVIEW: Hematopoietic stem cells (HSCs) are defined by their ability to self-renew and differentiate to replenish all blood lineages throughout adult life. Under homeostasis, the majority of HSCs are quiescent, and few stem cells are cycling to sustain hematopoiesis. However, HSCs can be induced to proliferate and differentiate in response to stress signals produced during infection, inflammation, chemotherapy, radiation, bone marrow transplantation, and aging. Recent evidence suggests that acute and chronic stress impact the number and function of HSCs including their ability to repopulate and produce mature cells. This review will focus on how chronic stress affects HSC biology and methods to mitigate HSC loss during chronic hematopoietic stress. RECENT FINDINGS: Quiescent HSCs exit dormancy, divide, and differentiate to maintain steady-state hematopoiesis. Under conditions of acute stress including infection or blood loss some HSCs are pushed into division by cytokines and proinflammatory stimuli to differentiate and provide needed myeloid and erythroid cells to protect and reconstitute the host; after which, hematopoiesis returns to steady-state with minimal loss of HSC function. However, under conditions of chronic stress including serial bone marrow transplantation (BMT), chronic inflammation, and genotoxic stress (chemotherapy) and aging, HSCs are continuously induced to proliferate and undergo accelerated exhaustion. Recent evidence demonstrates that ablation of inhibitor of DNA binding 1 (Id1) gene can protect HSCs from exhaustion during chronic proliferative stress by promoting HSC quiescence. SUMMARY: Increasing our understanding of the molecular processes that protect HSCs from chronic proliferative stress could lead to therapeutic opportunities to prevent accelerated HSC exhaustion during physiological stress, genotoxic stress, BMT, and aging.


Aging/metabolism , Cell Differentiation , Cell Proliferation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Stress, Physiological , Aging/pathology , Hematopoietic Stem Cells/pathology , Humans
8.
PLoS One ; 15(4): e0232025, 2020.
Article En | MEDLINE | ID: mdl-32353019

The actin cytoskeleton plays a central role in establishing cell polarity and shape during embryonic morphogenesis. Daam1, a member of the Formin family of actin cytoskeleton regulators, is a Dvl2-binding protein that functions in the Wnt/Planar Cell Polarity (PCP) pathway. To examine the role of the Daam proteins in mammalian development, we generated Daam-deficient mice by gene targeting and found that Daam1, but not Daam2, is necessary for fetal survival. Embryonic development of Daam1 mutants was delayed most likely due to functional defects in the labyrinthine layer of the placenta. Examination of Daam2 and Daam1/2 double mutants revealed that Daam1 and Daam2 are functionally redundant during placental development. Of note, neural tube closure defects (NTD), which are observed in several mammalian PCP mutants, are not observed in Wnt5a or Daam1 single mutants, but arise in Daam1;Wnt5a double mutants. These findings demonstrate a unique function for Daam genes in placental development and are consistent with a role for Daam1 in the Wnt/PCP pathway in mammals.


Microfilament Proteins/genetics , Placentation/genetics , rho GTP-Binding Proteins/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/metabolism , Cell Polarity , Cytoskeleton/metabolism , Embryonic Development , Female , Formins/genetics , Formins/metabolism , Gene Expression Regulation, Developmental/genetics , Male , Mice/embryology , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Placenta/embryology , Pregnancy , Wnt Signaling Pathway , rho GTP-Binding Proteins/metabolism
9.
Cell Rep ; 31(4): 107572, 2020 04 28.
Article En | MEDLINE | ID: mdl-32348770

Investigating mechanisms that regulate endothelial cell (EC) growth and survival is important for understanding EC homeostasis and how ECs maintain stem cell niches. We report here that targeted loss of Id genes in adult ECs results in dilated, leaky sinusoids and a pro-inflammatory state that increases in severity over time. Disruption in sinusoidal integrity leads to increased hematopoietic stem cell (HSC) proliferation, differentiation, migration, and exhaustion. Mechanistically, sinusoidal ECs (SECs) show increased apoptosis because of reduced Bcl2-family gene expression following Id gene ablation. Furthermore, Id1-/-Id3-/- SECs and upstream type H vessels show increased expression of cyclin-dependent kinase inhibitors p21 and p27 and impaired ability to proliferate, which is rescued by reducing E2-2 expression. Id1-/-Id3-/- mice do not survive sublethal irradiation because of impaired vessel regeneration and hematopoietic failure. Thus, Id genes are required for the survival and regeneration of BM SECs during homeostasis and stress to maintain HSC development.


Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Proteins/metabolism , Animals , Cell Survival/physiology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/physiology , Female , Hematopoiesis/physiology , Humans , Male , Mice , Mice, Inbred C57BL , Regeneration/physiology
10.
Immunity ; 52(1): 83-95.e4, 2020 01 14.
Article En | MEDLINE | ID: mdl-31882362

Lymphoid tissue inducer (LTi) cells are regarded as a subset of innate lymphoid cells (ILCs). However, these cells are not derived from the ILC common progenitor, which generates other ILC subsets and is defined by the expression of the transcription factor PLZF. Here, we examined transcription factor(s) determining the fate of LTi progenitors versus non-LTi ILC progenitors. Conditional deletion of Gata3 resulted in the loss of PLZF+ non-LTi progenitors but not the LTi progenitors that expressed the transcription factor RORγt. Consistently, PLZF+ non-LTi progenitors expressed high amounts of GATA3, whereas GATA3 expression was low in RORγt+ LTi progenitors. The generation of both progenitors required the transcriptional regulator Id2, which defines the common helper-like innate lymphoid progenitor (ChILP), but not cytokine signaling. Nevertheless, low GATA3 expression was necessary for the generation of functionally mature LTi cells. Thus, differential expression of GATA3 determines the fates and functions of distinct ILC progenitors.


GATA3 Transcription Factor/biosynthesis , Stem Cells/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Lineage/immunology , Cells, Cultured , GATA3 Transcription Factor/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/biosynthesis , Programmed Cell Death 1 Receptor/biosynthesis , Promyelocytic Leukemia Zinc Finger Protein/biosynthesis , Stem Cells/immunology , T-Lymphocyte Subsets/immunology
11.
Blood Adv ; 3(22): 3635-3649, 2019 11 26.
Article En | MEDLINE | ID: mdl-31751474

Serine palmitoyltransferase (SPT) long-chain base subunit 1 (SPTLC1) is 1 of the 2 main catalytic subunits of the SPT complex, which catalyzes the first and rate-limiting step of sphingolipid biosynthesis. Here, we show that Sptlc1 deletion in adult bone marrow (BM) cells results in defective myeloid differentiation. In chimeric mice from noncompetitive BM transplant assays, there was an expansion of the Lin- c-Kit+ Sca-1+ compartment due to increased multipotent progenitor production, but myeloid differentiation was severely compromised. We also show that defective biogenesis of sphingolipids in the endoplasmic reticulum (ER) leads to ER stress that affects myeloid differentiation. Furthermore, we demonstrate that transient accumulation of fatty acid, a substrate for sphingolipid biosynthesis, could be partially responsible for the ER stress. Independently, we find that ER stress in general, such as that induced by the chemical thapsigargin or the fatty acid palmitic acid, compromises myeloid differentiation in culture. These results identify perturbed sphingolipid metabolism as a source of ER stress, which may produce diverse pathological effects related to differential cell-type sensitivity.


Cell Differentiation/genetics , Hematopoiesis/genetics , Homeostasis , Myeloid Cells/cytology , Myeloid Cells/metabolism , Serine C-Palmitoyltransferase/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Computational Biology/methods , Gene Deletion , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Spleen/cytology , Spleen/metabolism
12.
Cell Stem Cell ; 23(2): 252-265.e8, 2018 Aug 02.
Article En | MEDLINE | ID: mdl-30082068

Defining mechanisms that maintain tissue stem cells during homeostasis, stress, and aging is important for improving tissue regeneration and repair and enhancing cancer therapies. Here, we show that Id1 is induced in hematopoietic stem cells (HSCs) by cytokines that promote HSC proliferation and differentiation, suggesting that it functions in stress hematopoiesis. Genetic ablation of Id1 increases HSC self-renewal in serial bone marrow transplantation (BMT) assays, correlating with decreases in HSC proliferation, mitochondrial biogenesis, and reactive oxygen species (ROS) production. Id1-/- HSCs have a quiescent molecular signature and harbor less DNA damage than control HSCs. Cytokines produced in the hematopoietic microenvironment after γ-irradiation induce Id1 expression. Id1-/- HSCs display a blunted proliferative response to such cytokines and other inducers of chronic proliferation including genotoxic and inflammatory stress and aging, protecting them from chronic stress and exhaustion. Thus, targeting Id1 may be therapeutically useful for improving HSC survival and function during BMT, chronic stress, and aging.


Aging/metabolism , Hematopoietic Stem Cells/metabolism , Inhibitor of Differentiation Protein 1/deficiency , Stress, Physiological , Animals , Cells, Cultured , Inhibitor of Differentiation Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
13.
Cell Rep ; 23(11): 3236-3248, 2018 06 12.
Article En | MEDLINE | ID: mdl-29898395

Fetal globin genes are transcriptionally silenced during embryogenesis through hemoglobin switching. Strategies to derepress fetal globin expression in the adult could alleviate symptoms in sickle cell disease and ß-thalassemia. We identified a zinc-finger protein, pogo transposable element with zinc-finger domain (POGZ), expressed in hematopoietic progenitor cells. Targeted deletion of Pogz in adult hematopoietic cells in vivo results in persistence of embryonic ß-like globin expression without affecting erythroid development. POGZ binds to the Bcl11a promoter and erythroid-specific intragenic regulatory regions. Pogz+/- mice show elevated embryonic ß-like globin expression, suggesting that partial reduction of Pogz expression results in persistence of embryonic ß-like globin expression. Knockdown of POGZ in primary human CD34+ progenitor cell-derived erythroblasts reduces BCL11A expression, a known repressor of embryonic ß-like globin expression, and increases fetal hemoglobin expression. These findings are significant, since new therapeutic targets and strategies are needed to treat ß-globin disorders.


Fetal Hemoglobin/metabolism , Transposases/genetics , beta-Globins/genetics , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Differentiation , Embryo, Mammalian/metabolism , Embryonic Development , Erythroblasts/cytology , Erythroblasts/metabolism , Fetal Hemoglobin/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins , Transposases/antagonists & inhibitors , Transposases/metabolism , beta-Globins/metabolism
14.
Cancer Cell ; 31(4): 501-515.e8, 2017 04 10.
Article En | MEDLINE | ID: mdl-28399408

Targeting the tumor vasculature with antibody-drug conjugates (ADCs) is a promising anti-cancer strategy that in order to be realized must overcome several obstacles, including identification of suitable targets and optimal warheads. Here, we demonstrate that the cell-surface protein CD276/B7-H3 is broadly overexpressed by multiple tumor types on both cancer cells and tumor-infiltrating blood vessels, making it a potentially ideal dual-compartment therapeutic target. In preclinical studies CD276 ADCs armed with a conventional MMAE warhead destroyed CD276-positive cancer cells, but were ineffective against tumor vasculature. In contrast, pyrrolobenzodiazepine-conjugated CD276 ADCs killed both cancer cells and tumor vasculature, eradicating large established tumors and metastases, and improving long-term overall survival. CD276-targeted dual-compartment ablation could aid in the development of highly selective broad-acting anti-cancer therapies.


B7 Antigens/genetics , B7 Antigens/metabolism , Immunoconjugates/pharmacology , Neoplasms/blood supply , Animals , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , B7 Antigens/immunology , Benzodiazepines/pharmacology , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Line, Tumor , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Immunoconjugates/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Targeted Therapy/methods , Neoplasms/pathology , Neoplasms/therapy , Oligopeptides/pharmacology , Pyrroles/pharmacology , Rabbits
15.
PLoS One ; 12(2): e0171510, 2017.
Article En | MEDLINE | ID: mdl-28158249

Thymic epithelial cells (TEC), as part of thymic stroma, provide essential growth factors/cytokines and self-antigens to support T cell development and selection. Deletion of Rb family proteins in adult thymic stroma leads to T cell hyperplasia in vivo. To determine whether deletion of Rb specifically in keratin (K) 18 positive TEC was sufficient for thymocyte hyperplasia, we conditionally inactivated Rb and its family members p107 and p130 in K18+ TEC in genetically engineered mice (TgK18GT121; K18 mice). We found that thymocyte hyperproliferation was induced in mice with Rb inactivation in K18+ TEC, while normal T cell development was maintained; suggesting that inactivation of Rb specifically in K18+ TEC was sufficient and responsible for the phenotype. Transplantation of wild type bone marrow cells into mice with Rb inactivation in K18+ TEC resulted in donor T lymphocyte hyperplasia confirming the non-cell autonomous requirement for Rb proteins in K18+ TEC in regulating T cell proliferation. Our data suggests that thymic epithelial cells play an important role in regulating lymphoid proliferation and thymus size.


Cell Proliferation , Keratin-18/metabolism , Retinoblastoma Protein/physiology , T-Lymphocytes/cytology , Thymus Gland/cytology , Animals , Female , Gene Expression , Male , Mice , Mice, Transgenic , Retinoblastoma Protein/genetics , Stromal Cells/metabolism , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/metabolism , Transgenes
16.
Oncotarget ; 7(51): 85109-85123, 2016 Dec 20.
Article En | MEDLINE | ID: mdl-27835883

c-Kit is a tyrosine kinase receptor important for gametogenesis, hematopoiesis, melanogenesis and mast cell biology. Dysregulation of c-Kit function is oncogenic and its expression in the stem cell niche of a number of tissues has underlined its relevance for regenerative medicine and hematopoietic stem cell biology. Yet, very little is known about the mechanisms that control c-Kit protein levels. Here we show that the RanBPM/RanBP9 scaffold protein binds to c-Kit and is necessary for normal c-Kit protein expression in the mouse testis and subset lineages of the hematopoietic system. RanBPM deletion causes a reduction in c-Kit protein but not its mRNA suggesting a posttranslational mechanism. This regulation is specific to the c-Kit receptor since RanBPM reduction does not affect other membrane proteins examined. Importantly, in both mouse hematopoietic system and testis, RanBPM deficiency causes defects consistent with c-Kit loss of expression suggesting that RanBPM is an important regulator of c-Kit function. The finding that this regulatory mechanism is also present in human cells expressing endogenous RanBPM and c-Kit suggests a potential new strategy to target oncogenic c-Kit in malignancies.


Adaptor Proteins, Signal Transducing/metabolism , Bone Marrow Cells/metabolism , Cytoskeletal Proteins/metabolism , Germ Cells/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/metabolism , Testis/pathology , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Marrow Cells/pathology , Cell Differentiation , Cytoskeletal Proteins/genetics , Gene Expression Regulation , Germ Cells/pathology , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Protein Binding , Proto-Oncogene Proteins c-kit/genetics , RNA, Small Interfering/genetics , Stem Cells/pathology
17.
Nat Commun ; 7: 12425, 2016 08 08.
Article En | MEDLINE | ID: mdl-27498558

Poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) olaparib has been approved for treatment of advanced ovarian cancer associated with BRCA1 and BRCA2 mutations. BRCA1- and BRCA2-mutated cells, which are homologous recombination (HR) deficient, are hypersensitive to PARPi through the mechanism of synthetic lethality. Here we examine the effect of PARPi on HR-proficient cells. Olaparib pretreatment, PARP1 knockdown or Parp1 heterozygosity of Brca2(cko/ko) mouse embryonic stem cells (mESCs), carrying a null (ko) and a conditional (cko) allele of Brca2, results in viable Brca2(ko/ko) cells. PARP1 deficiency does not restore HR in Brca2(ko/ko) cells, but protects stalled replication forks from MRE11-mediated degradation through its impaired recruitment. The functional consequence of Parp1 heterozygosity on BRCA2 loss is demonstrated by a significant increase in tumorigenesis in Brca2(cko/cko) mice. Thus, while olaparib efficiently kills BRCA2-deficient cells, we demonstrate that it can also contribute to the synthetic viability if PARP is inhibited before BRCA2 loss.


BRCA2 Protein/deficiency , Poly (ADP-Ribose) Polymerase-1/deficiency , Animals , BRCA2 Protein/metabolism , Cell Survival/drug effects , DNA Replication/drug effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Homologous Recombination/drug effects , Humans , Integrases/metabolism , MRE11 Homologue Protein/metabolism , Mice , Models, Biological , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
18.
Stem Cells ; 34(4): 1068-82, 2016 Apr.
Article En | MEDLINE | ID: mdl-27095138

Folliculin (FLCN) is an autosomal dominant tumor suppressor gene that modulates diverse signaling pathways required for growth, proliferation, metabolism, survival, motility, and adhesion. FLCN is an essential protein required for murine embryonic development, embryonic stem cell (ESC) commitment, and Drosophila germline stem cell maintenance, suggesting that Flcn may be required for adult stem cell homeostasis. Conditional inactivation of Flcn in adult hematopoietic stem/progenitor cells (HSPCs) drives hematopoietic stem cells (HSC) into proliferative exhaustion resulting in the rapid depletion of HSPC, loss of all hematopoietic cell lineages, acute bone marrow (BM) failure, and mortality after 40 days. HSC that lack Flcn fail to reconstitute the hematopoietic compartment in recipient mice, demonstrating a cell-autonomous requirement for Flcn in HSC maintenance. BM cells showed increased phosphorylation of Akt and mTorc1, and extramedullary hematopoiesis was significantly reduced by treating mice with rapamycin in vivo, suggesting that the mTorc1 pathway was activated by loss of Flcn expression in hematopoietic cells in vivo. Tfe3 was activated and preferentially localized to the nucleus of Flcn knockout (KO) HSPCs. Tfe3 overexpression in HSPCs impaired long-term hematopoietic reconstitution in vivo, recapitulating the Flcn KO phenotype, and supporting the notion that abnormal activation of Tfe3 contributes to the Flcn KO phenotype. Flcn KO mice develop an acute histiocytic hyperplasia in multiple organs, suggesting a novel function for Flcn in macrophage development. Thus, Flcn is intrinsically required to maintain adult HSC quiescence and homeostasis, and Flcn loss leads to BM failure and mortality in mice.


Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Estrone/genetics , Hematopoietic Stem Cells/pathology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/pathology , Cell Lineage/genetics , Cell Proliferation/genetics , Embryonic Development/genetics , Hematopoietic Stem Cells/metabolism , Homeostasis/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout
19.
Stem Cells ; 34(7): 1934-46, 2016 07.
Article En | MEDLINE | ID: mdl-26990002

Mesenchymal stem cells (MSCs) are multipotent stromal cells residing in the bone marrow. MSCs have the potential to differentiate to adipocytes, chondrocytes, and other types of cells. In this study, we investigated the molecular mechanism that controls MSC cell fate decisions for differentiation. We found that Vav1, a guanine nucleotide exchange factor for Rho GTPase, was highly expressed in MSCs. Interestingly, loss of Vav1 in MSCs led to spontaneous adipogenic but impaired chondrogenic differentiation, and accordingly Vav1 null mice displayed an increase in fat content and a decrease in cartilage. Conversely, ectopic expression of Vav1 in MSCs reversed this phenotype, and led to enhanced MSC differentiation into chondrocyte but retarded adipogenesis. Mechanistically, loss of Vav1 reduced the level of Sirt1, which was responsible for an increase of acetylated PPARγ. As acetylation activates PPARγ, it increased C/EBPα expression and promoted adipogenesis. On the other hand, loss of Vav1 resulted in an increase of acetylated Sox9, a target of Sirt1. As acetylation represses Sox9 activity, it led to a dramatic reduction of collagen 2α1, a key regulator in chondrocyte differentiation. Finally, we found that Vav1 regulates Sirt1 in MSCs through Creb. Together this study reveals a novel function of Vav1 in regulating MSC cell fate decisions for differentiation through Sirt1. Sirt1 deacetylates PPARγ and Sox9, two key mediators that control adipocyte and chondrocyte differentiation. The acetylation status of PPARγ and Sox9 has opposite effects on its activity, thereby controlling cell fate decision. Stem Cells 2016;34:1934-1946.


Adipocytes/cytology , Cell Differentiation , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Sirtuin 1/metabolism , Adipocytes/metabolism , Adipogenesis/genetics , Adiposity , Animals , Cell Differentiation/genetics , Chondrocytes/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Gene Deletion , Mice, Inbred C57BL , Transcription Factors/metabolism
20.
Hum Mol Genet ; 25(10): 1934-1945, 2016 05 15.
Article En | MEDLINE | ID: mdl-26920070

The breast cancer gene, BRCA2, is essential for viability, yet patients with Fanconi anemia-D1 subtype are born alive with biallelic mutations in this gene. The hypomorphic nature of the mutations is believed to support viability, but this is not always apparent. One such mutation is IVS7+2T>G, which causes premature protein truncation due to skipping of exon 7. We previously identified a transcript lacking exons 4-7, which restores the open-reading frame, encodes a DNA repair proficient protein and is expressed in IVS7+2T>G carriers. However, because the exons 4-7 encoded region contains several residues required for normal cell-cycle regulation and cytokinesis, this transcript's ability to support viability can be argued. To address this, we generated a Brca2 knock-in mouse model lacking exons 4-7 and demonstrated that these exons are dispensable for viability as well as tumor-free survival. This study provides the first in vivo evidence of the functional significance of a minor transcript of BRCA2 that can play a major role in the survival of humans who are homozygous for a clearly pathogenic mutation. Our results highlight the importance of assessing protein function restoration by premature truncating codon bypass by alternative splicing when evaluating the functional significance of variants such as nonsense and frame-shift mutations that are assumed to be clearly pathogenic. Our findings will impact not only the assessment of variants that map to this region, but also influence counseling paradigms and treatment options for such mutation carriers.


BRCA2 Protein/genetics , Breast Neoplasms/genetics , Fanconi Anemia/genetics , Genetic Predisposition to Disease , Alternative Splicing/genetics , Animals , Breast Neoplasms/pathology , Exons/genetics , Fanconi Anemia/pathology , Gene Knock-In Techniques , Germ-Line Mutation , Humans , Mice , Mutation , Pedigree , RNA Splice Sites
...