Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 132(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35775482

ABSTRACT

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.


Subject(s)
Hematopoietic Stem Cells , Mitochondria , Hematopoietic Stem Cells/metabolism , Mitochondria/metabolism
2.
Cancer Res ; 81(23): 5977-5990, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34642183

ABSTRACT

The relationship between cancer and autoimmunity is complex. However, the incidence of solid tumors such as melanoma has increased significantly among patients with previous or newly diagnosed systemic autoimmune disease (AID). At the same time, immune checkpoint blockade (ICB) therapy of cancer induces de novo autoinflammation and exacerbates underlying AID, even without evident antitumor responses. Recently, systemic lupus erythematosus (SLE) activity was found to drive myeloid-derived suppressor cell (MDSC) formation in patients, a known barrier to healthy immune surveillance and successful cancer immunotherapy. Cross-talk between MDSCs and macrophages generally drives immune suppressive activity in the tumor microenvironment. However, it remains unclear how peripheral pregenerated MDSC under chronic inflammatory conditions modulates global macrophage immune functions and the impact it could have on existing tumors and underlying lupus nephritis. Here we show that pathogenic expansion of SLE-generated MDSCs by melanoma drives global macrophage polarization and simultaneously impacts the severity of lupus nephritis and tumor progression in SLE-prone mice. Molecular and functional data showed that MDSCs interact with autoimmune macrophages and inhibit cell surface expression of CD40 and the production of IL27. Moreover, low CD40/IL27 signaling in tumors correlated with high tumor-associated macrophage infiltration and ICB therapy resistance both in murine and human melanoma exhibiting active IFNγ signatures. These results suggest that preventing global macrophage reprogramming induced by MDSC-mediated inhibition of CD40/IL27 signaling provides a precision melanoma immunotherapy strategy, supporting an original and advantageous approach to treat solid tumors within established autoimmune landscapes. SIGNIFICANCE: Myeloid-derived suppressor cells induce macrophage reprogramming by suppressing CD40/IL27 signaling to drive melanoma progression, simultaneously affecting underlying autoimmune disease and facilitating resistance to immunotherapy within preexisting autoimmune landscapes.


Subject(s)
Autoimmunity , CD40 Antigens/metabolism , Interleukin-27/metabolism , Lupus Erythematosus, Systemic/physiopathology , Macrophages/pathology , Melanoma/pathology , Myeloid-Derived Suppressor Cells/pathology , Animals , Immunotherapy , Macrophages/immunology , Macrophages/metabolism , Melanoma/immunology , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Tumor Microenvironment
3.
Cell Rep ; 31(4): 107572, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32348770

ABSTRACT

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.


Subject(s)
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
4.
Blood Adv ; 3(22): 3635-3649, 2019 11 26.
Article in English | MEDLINE | ID: mdl-31751474

ABSTRACT

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.


Subject(s)
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
5.
Cell Stem Cell ; 23(2): 252-265.e8, 2018 Aug 02.
Article in English | MEDLINE | ID: mdl-30082068

ABSTRACT

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.


Subject(s)
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
6.
Cell Rep ; 23(11): 3236-3248, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898395

ABSTRACT

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.


Subject(s)
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
7.
Cancer Cell ; 31(4): 501-515.e8, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399408

ABSTRACT

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.


Subject(s)
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
8.
Oncotarget ; 7(51): 85109-85123, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27835883

ABSTRACT

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.


Subject(s)
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
9.
Nat Commun ; 7: 12425, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27498558

ABSTRACT

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.


Subject(s)
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
10.
Stem Cells ; 34(4): 1068-82, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27095138

ABSTRACT

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.


Subject(s)
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
11.
Hum Mol Genet ; 25(10): 1934-1945, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26920070

ABSTRACT

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.


Subject(s)
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
12.
Blood ; 124(10): 1586-96, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25051963

ABSTRACT

Growth factor independence 1 (Gfi-1) is a part of the transcriptional network that regulates the development of adult hematopoietic stem and progenitor cells. Gfi-1-null (Gfi-1(-/-)) mice have reduced numbers of hematopoietic stem cells (HSCs), impaired radioprotective function of hematopoietic progenitor cells (HPCs), and myeloid and erythroid hyperplasia. We found that the development of HPCs and erythropoiesis, but not HSC function, was rescued by reducing the expression of inhibitor of DNA-binding protein 2 (Id2) in Gfi-1(-/-) mice. Analysis of Gfi-1(-/-);Id2(+/-) mice revealed that short-term HSCs, common myeloid progenitors (CMPs), erythroid burst-forming units, colony-forming units in spleen, and more differentiated red cells were partially restored by reducing Id2 levels in Gfi-1(-/-) mice. Moreover, short-term reconstituting cells, and, to a greater extent, CMP and megakaryocyte-erythroid progenitor development, and red blood cell production (anemia) were rescued in mice transplanted with Gfi-1(-/-);Id2(+/-) bone marrow cells (BMCs) in comparison with Gfi-1(-/-) BMCs. Reduction of Id2 expression in Gfi-1(-/-) mice increased the expression of Gata1, Eklf, and EpoR, which are required for proper erythropoiesis. Reducing the levels of other Id family members (Id1 and Id3) in Gfi-1(-/-) mice did not rescue impaired HPC function or erythropoiesis. These data provide new evidence that Gfi-1 is linked to the erythroid gene regulatory network by repressing Id2 expression.


Subject(s)
DNA-Binding Proteins/physiology , Erythropoiesis/genetics , Gene Regulatory Networks , Hematopoietic Stem Cells/metabolism , Inhibitor of Differentiation Protein 2/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Down-Regulation/genetics , Erythroid Precursor Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Transcription Factors/metabolism , Transcription Factors/physiology
13.
Nat Commun ; 4: 1662, 2013.
Article in English | MEDLINE | ID: mdl-23575666

ABSTRACT

Toll-like receptor 4 (Tlr4) has a pivotal role in innate immune responses, and the transcription factor CCAAT/enhancer binding protein delta (C/EBPδ, Cebpd) is a Tlr4-induced gene. Here we identify a positive feedback loop in which C/EBPδ activates Tlr4 gene expression in macrophages and tumour cells. In addition, we discovered a negative feedback loop whereby the tumour suppressor FBXW7α (FBW7, Cdc4), whose gene expression is inhibited by C/EBPδ, targets C/EBPδ for degradation when C/EBPδ is phosphorylated by GSK-3ß. Consequently, FBXW7α suppresses Tlr4 expression and responses to the ligand lipopolysaccharide. FBXW7α depletion alone is sufficient to augment pro-inflammatory signalling in vivo. Moreover, as inflammatory pathways are known to modulate tumour biology, Cebpd null mammary tumours, which have reduced metastatic potential, show altered expression of inflammation-associated genes. Together, these findings reveal a role for C/EBPδ upstream of Tlr4 signalling and uncover a function for FBXW7α as an attenuator of inflammatory signalling.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/physiology , Down-Regulation , F-Box Proteins/physiology , Inflammation/physiopathology , Signal Transduction/physiology , Toll-Like Receptor 4/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Cell Line, Tumor , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/genetics , Mice , RNA, Messenger/genetics , Ubiquitin-Protein Ligases/genetics
14.
Stem Cells ; 31(8): 1454-66, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23592398

ABSTRACT

Pancreatic cancer is the fourth leading cause of cancer-related mortality in the world. Pancreatic cancer can be localized, locally advanced, or metastatic. The median 1- and 5-year survival rates are 25% and 6%, respectively. Epigenetic modifications such as DNA methylation play a significant role during both normal human development and cancer progression. To investigate epigenetic regulation of genes in the tumor-initiating population of pancreatic cancer cells, which are also termed cancer stem cells (CSCs), we conducted epigenetic arrays in PANC1 and HPAC pancreatic cancer cell lines and compared the global DNA methylation status of CpG promoters in invasive cells, demonstrated to be CSCs, to their noninvasive counterparts, or non-CSCs. Our results suggested that the NF-κB pathway is one of the most activated pathways in pancreatic CSCs. In agreement with this, we determined that upon treatment with NF-κB pathway inhibitors, the stem cell-like properties of cells are significantly disrupted. Moreover, SOX9, demethylated in CSCs, is shown to play a crucial role in the invasion process. Additionally, we found a potential NF-κB binding site located in the SOX9 promoter and determined that the NF-κB subunit p65 positively regulates SOX9 expression by binding to its promoter directly. This interaction can be efficiently blocked by NF-κB inhibitors. Thus, our work establishes a link between the classic NF-κB signaling transduction pathway and the invasiveness of pancreatic CSCs, which may result in the identification of novel signals and molecules that function at an epigenetic level, and could potentially be targeted for pharmaceutical investigations and clinical trials.


Subject(s)
NF-kappa B/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Animals , Cell Line, Tumor , DNA Methylation , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , NF-kappa B/genetics , Neoplasm Invasiveness , Signal Transduction , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism
15.
FASEB J ; 26(1): 309-23, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21990377

ABSTRACT

Obesity is a major health concern that contributes to the development of diabetes, hyperlipidemia, coronary artery disease, and cancer. Id proteins are helix-loop-helix transcription factors that regulate the proliferation and differentiation of cells from multiple tissues, including adipocytes. We screened mouse tissues for the expression of Id1 and found that Id1 protein is highly expressed in brown adipose tissue (BAT) and white adipose tissue (WAT), suggesting a role for Id1 in adipogenesis and cell metabolism. Id1(-/-) mice are viable but show a significant reduction in fat mass (P<0.005) over the life of the animal that was not due to decreased number of adipocytes. Analysis of Id1(-/-) mice revealed higher energy expenditure, increased lipolysis, and fatty acid oxidation, resulting in reduced triglyceride accumulation in WAT compared to Id1(+/+) mice. Serum levels of triglycerides (193.9±32.2 vs. 86.5±33.8, P<0.0005), cholesterol (189.4±33.8 vs. 110.6±8.23, P<0.0005) and leptin (1263±835 vs. 222±260, P<0.005) were significantly lower in aged Id1(-/-) mice compared to Id1(+/+) mice. Id1-deficient mice have higher resting (P<0.005) and total (P<0.05) O(2) consumption and lower respiratory exchange ratio (P<0.005), confirming that Id1(-/-) mice use a higher proportion of lipid as an energy source for the increased energy expenditure. The expression of PGC1α and UCP1 were 2- to 3-fold up-regulated in Id1(-/-) BAT, suggesting that loss of Id1 increases thermogenesis. As a consequence of higher energy expenditure and reduced fat mass, Id1(-/-) mice displayed enhanced insulin sensitivity. Id1 deficiency protected mice against age- and high-fat-diet-induced adiposity, insulin resistance, and hepatosteatosis. Our findings suggest that Id1 plays a critical role in the regulation of energy homeostasis and could be a potential target in the treatment of insulin resistance and fatty liver disease.


Subject(s)
Aging/metabolism , Energy Metabolism/physiology , Fatty Liver/metabolism , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Insulin Resistance/physiology , Adipocytes/cytology , Adipogenesis/physiology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Fatty Acids/metabolism , Fatty Liver/genetics , Fatty Liver/prevention & control , Female , Fibroblasts/cytology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , Pregnancy , Thermogenesis/physiology
16.
Blood ; 116(7): 1060-9, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20453161

ABSTRACT

The development of mature blood cells from hematopoietic stem cells requires coordinated activities of transcriptional networks. Transcriptional repressor growth factor independence 1 (Gfi-1) is required for the development of B cells, T cells, neutrophils, and for the maintenance of hematopoietic stem cell function. However, the mechanisms by which Gfi-1 regulates hematopoiesis and how Gfi-1 integrates into transcriptional networks remain unclear. Here, we provide evidence that Id2 is a transcriptional target of Gfi-1, and repression of Id2 by Gfi-1 is required for B-cell and myeloid development. Gfi-1 binds to 3 conserved regions in the Id2 promoter and represses Id2 promoter activity in transient reporter assays. Increased Id2 expression was observed in multipotent progenitors, myeloid progenitors, T-cell progenitors, and B-cell progenitors in Gfi-1(-/-) mice. Knockdown of Id2 expression or heterozygosity at the Id2 locus partially rescues the B-cell and myeloid development but not the T-cell development in Gfi-1(-/-) mice. These studies demonstrate a role of Id2 in mediating Gfi-1 functions in B-cell and myeloid development and provide a direct link between Gfi-1 and the B-cell transcriptional network by its ability to repress Id2 expression.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/metabolism , DNA-Binding Proteins/physiology , Inhibitor of Differentiation Protein 2/physiology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Transcription Factors/physiology , Animals , Base Sequence , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Colony-Forming Units Assay , Electrophoretic Mobility Shift Assay , Flow Cytometry , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunoenzyme Techniques , Inhibitor of Differentiation Protein 2/antagonists & inhibitors , Luciferases/metabolism , Mice , Mice, Knockout , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Transfection
17.
Blood ; 114(6): 1186-95, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19478045

ABSTRACT

Development of hematopoietic stem cells (HSCs) and their immediate progeny is maintained by the interaction with cells in the microenvironment. We found that hematopoiesis was dysregulated in Id1(-/-) mice. Although the frequency of HSCs in Id1(-/-) bone marrow was increased, their total numbers remained unchanged as the result of decreased bone marrow cellularity. In addition, the ability of Id1(-/-) HSCs to self-renew was normal, suggesting Id1 does not affect HSC function. Id1(-/-) progenitors showed increased cycling in vivo but not in vitro, suggesting cell nonautonomous mechanisms for the increased cycling. Id1(-/-) HSCs developed normally when transplanted into Id1(+/+) mice, whereas the development of Id1(+/+) HSCs was impaired in Id1(-/-) recipients undergoing transplantation and reproduced the hematologic features of Id1(-/-) mice, indicating that the Id1(-/-) microenvironment cannot support normal hematopoietic development. Id1(-/-) stromal cells showed altered production of cytokines in vitro, and cytokine levels were deregulated in vivo, which could account for the Id1(-/-) hematopoietic phenotypes. Thus, Id1 is required for regulating the hematopoietic progenitor cell niche but is dispensable for maintaining HSCs.


Subject(s)
Bone Marrow/metabolism , Cell Cycle/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Animals , Cytokines/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Stromal Cells/cytology , Stromal Cells/metabolism
18.
Hum Mol Genet ; 17(19): 2921-33, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18621876

ABSTRACT

The lamin B receptor (LBR) is an integral nuclear envelope protein that interacts with chromatin and has homology to sterol reductases. Mutations in LBR result in Pelger-Huët anomaly and HEM-Greenberg skeletal dysplasia, whereas in mice Lbr mutations result in ichthyosis. To further understand the function of the LBR and its role in disease, we derived a novel mouse model with a gene-trap insertion into the Lbr locus (Lbr(GT/GT)). Phenotypically, the Lbr(GT/GT) mice are similar to ichthyosis mice. The Lbr(GT/GT) granulocytes lack a mature segmented nucleus and have a block in late maturation. Despite these changes in nuclear morphology, the innate granulocyte immune function in the killing of Staphylococcus aureus bacteria appears to be intact. Granulocyte differentiation requires the transcription factor C/EBPepsilon. We identified C/EBPepsilon binding sites within the Lbr promoter and used EMSAs and luciferase assays to show that Lbr is transcriptionally regulated by C/EBPepsilon. Our findings indicate that the Lbr(GT/GT) mice are a model for Pelger-Huët anomaly and that Lbr, under transcriptional regulation of C/EBPepsilon, is necessary for morphological but not necessarily functional granulocyte maturation.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Neutrophils/cytology , Pelger-Huet Anomaly/genetics , Pelger-Huet Anomaly/physiopathology , Receptors, Cytoplasmic and Nuclear/genetics , Transcription, Genetic , Animals , CCAAT-Enhancer-Binding Proteins/genetics , Cell Differentiation , Cell Nucleus Shape , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutagenesis, Insertional , Neutrophil Activation , Neutrophils/physiology , Pelger-Huet Anomaly/embryology , Pelger-Huet Anomaly/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Staphylococcus aureus/physiology , Lamin B Receptor
19.
Blood ; 112(4): 1068-77, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18523151

ABSTRACT

Inhibitors of DNA binding (Id) family members are key regulators of cellular differentiation and proliferation. These activities are related to the ability of Id proteins to antagonize E proteins and other transcription factors. As negative regulators of E proteins, Id proteins have been implicated in lymphocyte development. Overexpression of Id1, Id2, or Id3 has similar effects on lymphocyte development. However, which Id protein plays a physiologic role during lymphocyte development is not clear. By analyzing Id2 knock-out mice and retroviral transduced hematopoietic progenitors, we demonstrated that Id2 is an intrinsic negative regulator of B-cell development. Hematopoietic progenitor cells overexpressing Id2 did not reconstitute B-cell development in vivo, which resembled the phenotype of E2A null mice. The B-cell population in bone marrow was significantly expanded in Id2 knock-out mice compared with their wild-type littermates. Knock-down of Id2 by shRNA in hematopoietic progenitor cells promoted B-cell differentiation and induced the expression of B-cell lineage-specific genes. These data identified Id2 as a physiologically relevant regulator of E2A during B lymphopoiesis. Furthermore, we identified a novel Id2 function in erythroid development. Overexpression of Id2 enhanced erythroid development, and decreased level of Id2 impaired normal erythroid development. Id2 regulation of erythroid development is mediated via interacting with transcription factor PU.1 and modulating PU.1 and GATA-1 activities. We conclude that Id2 regulates lymphoid and erythroid development via interaction with different target proteins.


Subject(s)
B-Lymphocytes/cytology , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Lineage , Erythroid Cells/cytology , GATA1 Transcription Factor/physiology , Inhibitor of Differentiation Protein 2/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Animals , Bone Marrow , Erythropoiesis , Hematopoietic Stem Cells , Inhibitor of Differentiation Protein 2/genetics , Lymphocytes/cytology , Lymphopoiesis , Mice , Mice, Knockout , Protein Binding/physiology
20.
J Immunol ; 178(3): 1553-63, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17237404

ABSTRACT

IL-7 administration to mice was previously reported to increase the mobilization of progenitor cells from marrow to peripheral sites. We now report that IL-7 increases the number of mature myeloid and monocytic cells in spleen and peripheral blood. This effect required T cells, and we show that IL-7 treatment in vivo induced GM-CSF and IL-3 production by T cells with memory phenotype. However, additional myelopoietic cytokines were shown to be involved because mice deficient in both GM-CSF and IL-3 also responded to IL-7 with increased myelopoiesis. Candidate cytokines included IFN-gamma and Flt3 ligand, which were also produced in response to IL-7. Because IFN-gamma-deficient mice also increased myelopoiesis, it was suggested that IL-7 induced production of redundant myelopoietic cytokines. In support of this hypothesis, we found that the supernatant from IL-7-treated, purified T cells contained myelopoietic activity that required a combination of Abs against GM-CSF, IL-3, and anti-Flt3 ligand to achieve maximum neutralization. IL-7 administration increased the number of splenic erythroid cells in either normal, Rag1 or GM-CSF-IL-3-deficient mice, suggesting that IL-7 might directly act on erythroid progenitors. In support of this theory, we detected a percentage of TER-119(+) erythroid cells that expressed the IL-7Ralpha-chain and common gamma-chain. Bone marrow cells expressing IL-7R and B220 generated erythroid colonies in vitro in response to IL-7, erythropoietin, and stem cell factor. This study demonstrates that IL-7 can promote nonlymphoid hemopoiesis and production of cytokines active in the host defense system in vivo, supporting its possible clinical utility.


Subject(s)
Erythropoiesis/drug effects , Interleukin-7/pharmacology , Myelopoiesis/drug effects , Animals , Blood Cells/cytology , Cytokines/biosynthesis , Cytokines/drug effects , Erythroid Precursor Cells/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interferon-gamma/biosynthesis , Interleukin-3/biosynthesis , Interleukin-7/administration & dosage , Membrane Proteins/biosynthesis , Mice , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...