Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Front Immunol ; 14: 1197490, 2023.
Article in English | MEDLINE | ID: mdl-37398663

ABSTRACT

The Haematopoietically expressed homeobox transcription factor (Hhex) is a transcriptional repressor that is of fundamental importance across species, as evident by its evolutionary conservation spanning fish, amphibians, birds, mice and humans. Indeed, Hhex maintains its vital functions throughout the lifespan of the organism, beginning in the oocyte, through fundamental stages of embryogenesis in the foregut endoderm. The endodermal development driven by Hhex gives rise to endocrine organs such as the pancreas in a process which is likely linked to its role as a risk factor in diabetes and pancreatic disorders. Hhex is also required for the normal development of the bile duct and liver, the latter also importantly being the initial site of haematopoiesis. These haematopoietic origins are governed by Hhex, leading to its crucial later roles in definitive haematopoietic stem cell (HSC) self-renewal, lymphopoiesis and haematological malignancy. Hhex is also necessary for the developing forebrain and thyroid gland, with this reliance on Hhex evident in its role in endocrine disorders later in life including a potential role in Alzheimer's disease. Thus, the roles of Hhex in embryological development throughout evolution appear to be linked to its later roles in a variety of disease processes.


Subject(s)
Genes, Homeobox , Transcription Factors , Humans , Animals , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Gene Expression Regulation , Liver/metabolism , Digestive System/metabolism
3.
Nat Immunol ; 24(5): 814-826, 2023 05.
Article in English | MEDLINE | ID: mdl-36997670

ABSTRACT

Missense mutations in PLCG2 can cause autoinflammation with phospholipase C gamma 2-associated antibody deficiency and immune dysregulation (APLAID). Here, we generated a mouse model carrying an APLAID mutation (p.Ser707Tyr) and found that inflammatory infiltrates in the skin and lungs were only partially ameliorated by removing inflammasome function via the deletion of caspase-1. Also, deleting interleukin-6 or tumor necrosis factor did not fully prevent APLAID mutant mice from autoinflammation. Overall, these findings are in accordance with the poor response individuals with APLAID have to treatments that block interleukin-1, JAK1/2 or tumor necrosis factor. Cytokine analysis revealed increased granulocyte colony-stimulating factor (G-CSF) levels as the most distinct feature in mice and individuals with APLAID. Remarkably, treatment with a G-CSF antibody completely reversed established disease in APLAID mice. Furthermore, excessive myelopoiesis was normalized and lymphocyte numbers rebounded. APLAID mice were also fully rescued by bone marrow transplantation from healthy donors, associated with reduced G-CSF production, predominantly from non-hematopoietic cells. In summary, we identify APLAID as a G-CSF-driven autoinflammatory disease, for which targeted therapy is feasible.


Subject(s)
Bone Marrow Transplantation , Granulocyte Colony-Stimulating Factor , Animals , Mice , Cytokines , Interleukin-1 , Tumor Necrosis Factor-alpha/genetics , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism
4.
Sci Immunol ; 7(68): eabi6763, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35148201

ABSTRACT

Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αß) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.


Subject(s)
Immunity, Innate/immunology , Interleukins/immunology , eIF-2 Kinase/immunology , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , eIF-2 Kinase/deficiency
5.
J Biol Chem ; 297(2): 100905, 2021 08.
Article in English | MEDLINE | ID: mdl-34157287

ABSTRACT

Phosphatidylinositol-specific phospholipase Cγ2 (PLCγ2) is a critical signaling molecule activated downstream from a variety of cell surface receptors that contain an intracellular immunoreceptor tyrosine-based activation motif. These receptors recruit kinases such as Syk, BTK, and BLNK to phosphorylate and activate PLCγ2, which then generates 1D-myo-inositol 1,4,5-trisphosphate and diacylglycerol. These well-known second messengers are required for diverse membrane functionality including cellular proliferation, endocytosis, and calcium flux. As a result, PLCγ2 dysfunction is associated with a variety of diseases including cancer, neurodegeneration, and immune disorders. The diverse pathologies associated with PLCγ2 are exemplified by distinct genetic variants. Inherited mutations at this locus cause PLCγ2-associated antibody deficiency and immune dysregulation, in some cases with autoinflammation. Acquired mutations at this locus, which often arise as a result of BTK inhibition to treat chronic lymphocytic leukemia, result in constitutive downstream signaling and lymphocyte proliferation. Finally, a third group of PLCγ2 variants actually has a protective effect in a variety of neurodegenerative disorders, presumably by increased uptake and degradation of deleterious neurological aggregates. Therefore, manipulating PLCγ2 activity either up or down could have therapeutic benefit; however, we require a better understanding of the signaling pathways propagated by these variants before such clinical utility can be realized. Here, we review the signaling roles of PLCγ2 in hematopoietic cells to help understand the effect of mutations driving immune disorders and cancer and extrapolate from this to roles which may relate to protection against neurodegeneration.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/immunology , Calcium Signaling , Immune System Diseases/pathology , Neoplasms/pathology , Neurodegenerative Diseases/pathology , Phospholipase C gamma/metabolism , Syk Kinase/immunology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/metabolism , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Syk Kinase/metabolism
6.
Leukemia ; 35(8): 2205-2219, 2021 08.
Article in English | MEDLINE | ID: mdl-33483615

ABSTRACT

The majority of cases of T-cell acute lymphoblastic leukemia (T-ALL) contain chromosomal abnormalities that drive overexpression of oncogenic transcription factors. However, whether these initiating oncogenes are required for leukemia maintenance is poorly understood. To address this, we developed a tetracycline-regulated mouse model of T-ALL driven by the oncogenic transcription factor Lmo2. This revealed that whilst thymus-resident pre-Leukemic Stem Cells (pre-LSCs) required continuous Lmo2 expression, the majority of leukemias relapsed despite Lmo2 withdrawal. Relapse was associated with a mature phenotype and frequent mutation or loss of tumor suppressor genes including Ikzf1 (Ikaros), with targeted deletion Ikzf1 being sufficient to transform Lmo2-dependent leukemias to Lmo2-independence. Moreover, we found that the related transcription factor TAL1 was dispensable in several human T-ALL cell lines that contain SIL-TAL1 chromosomal deletions driving its overexpression, indicating that evolution to oncogene independence can also occur in human T-ALL. Together these results indicate an evolution of oncogene addiction in murine and human T-ALL and show that loss of Ikaros is a mechanism that can promote self-renewal of T-ALL lymphoblasts in the absence of an initiating oncogenic transcription factor.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Gene Expression Regulation, Leukemic , Ikaros Transcription Factor/physiology , LIM Domain Proteins/physiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism
7.
Blood ; 137(21): 2920-2934, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33512466

ABSTRACT

OBF1 is a specific coactivator of the POU family transcription factors OCT1 and OCT2. OBF1 and OCT2 are B cell-specific and indispensable for germinal center (GC) formation, but their mechanism of action is unclear. Here, we show by chromatin immunoprecipitation-sequencing that OBF1 extensively colocalizes with OCT1 and OCT2. We found that these factors also often colocalize with transcription factors of the ETS family. Furthermore, we showed that OBF1, OCT2, and OCT1 bind widely to the promoters or enhancers of genes involved in GC formation in mouse and human GC B cells. Short hairpin RNA knockdown experiments demonstrated that OCT1, OCT2, and OBF1 regulate each other and are essential for proliferation of GC-derived lymphoma cell lines. OBF1 downregulation disrupts the GC transcriptional program: genes involved in GC maintenance, such as BCL6, are downregulated, whereas genes related to exit from the GC program, such as IRF4, are upregulated. Ectopic expression of BCL6 does not restore the proliferation of GC-derived lymphoma cells depleted of OBF1 unless IRF4 is also depleted, indicating that OBF1 controls an essential regulatory node in GC differentiation.


Subject(s)
Germinal Center/metabolism , Octamer Transcription Factor-1/physiology , Octamer Transcription Factor-2/therapeutic use , Trans-Activators/therapeutic use , Transcription, Genetic/genetics , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Ontology , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Octamer Transcription Factor-1/deficiency , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-2/deficiency , Octamer Transcription Factor-2/genetics , Proto-Oncogene Protein c-ets-1/analysis , RNA Interference , RNA, Small Interfering/genetics , Recombinant Proteins/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics
9.
Nat Immunol ; 21(12): 1574-1584, 2020 12.
Article in English | MEDLINE | ID: mdl-33077975

ABSTRACT

A classical view of blood cell development is that multipotent hematopoietic stem and progenitor cells (HSPCs) become lineage-restricted at defined stages. Lin-c-Kit+Sca-1+Flt3+ cells, termed lymphoid-primed multipotent progenitors (LMPPs), have lost megakaryocyte and erythroid potential but are heterogeneous in their fate. Here, through single-cell RNA sequencing, we identify the expression of Dach1 and associated genes in this fraction as being coexpressed with myeloid/stem genes but inversely correlated with lymphoid genes. Through generation of Dach1-GFP reporter mice, we identify a transcriptionally and functionally unique Dach1-GFP- subpopulation within LMPPs with lymphoid potential with low to negligible classic myeloid potential. We term these 'lymphoid-primed progenitors' (LPPs). These findings define an early definitive branch point of lymphoid development in hematopoiesis and a means for prospective isolation of LPPs.


Subject(s)
Biomarkers , Eye Proteins/metabolism , Genomics , Lymphoid Progenitor Cells/metabolism , Single-Cell Analysis , Animals , Cells, Cultured , Computational Biology/methods , Eye Proteins/genetics , Gene Expression Profiling , Genomics/methods , Hematopoiesis/genetics , High-Throughput Nucleotide Sequencing , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Knockout , Mice, Transgenic , Proteomics , Single-Cell Analysis/methods
10.
Cell Rep ; 33(3): 108285, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33086067

ABSTRACT

Hhex encodes a homeobox transcriptional regulator important for embryonic development and hematopoiesis. Hhex is highly expressed in NK cells, and its germline deletion results in significant defects in lymphoid development, including NK cells. To determine if Hhex is intrinsically required throughout NK cell development or for NK cell function, we generate mice that specifically lack Hhex in NK cells. NK cell frequency is dramatically reduced, while NK cell differentiation, IL-15 responsiveness, and function at the cellular level remain largely normal in the absence of Hhex. Increased IL-15 availability fails to fully reverse NK lymphopenia following conditional Hhex deletion, suggesting that Hhex regulates developmental pathways extrinsic to those dependent on IL-15. Gene expression and functional genetic approaches reveal that Hhex regulates NK cell survival by directly binding Bcl2l11 (Bim) and repressing expression of this key apoptotic mediator. These data implicate Hhex as a transcriptional regulator of NK cell homeostasis and immunity.


Subject(s)
Homeodomain Proteins/metabolism , Killer Cells, Natural/metabolism , Transcription Factors/metabolism , Animals , Apoptosis/genetics , Cell Differentiation/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Female , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Interleukin-15/genetics , Interleukin-15/immunology , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/physiology
11.
Eur J Immunol ; 50(7): 959-971, 2020 07.
Article in English | MEDLINE | ID: mdl-32090320

ABSTRACT

The transcription factor Hhex (hematopoietically expressed homeobox gene) is critical for development of multiple lymphoid lineages beyond the common lymphoid progenitor. In addition, Hhex regulates hematopoietic stem cell (HSC) self-renewal, emergency hematopoiesis, and acute myeloid leukemia initiation and maintenance. Hhex mediates its effects on HSCs and acute myeloid leukemia stem cells via repression of the Cdkn2a tumor suppressor locus. However, we report here that loss of Cdkn2a does not rescue the failure of lymphoid development caused by loss of Hhex. As loss of Hhex causes apoptosis of lymphoid progenitors associated with impaired Bcl2 expression and defective Stat5b signaling, we tested the effects of rescuing these pathways using transgenic mice. Expression of the anti-apoptotic factor Bcl2, but not activated Stat5, rescued the development of T-, B-, and NK-cell lineages in the absence of Hhex. These results indicate that Bcl2 expression, but not Stat5b signaling or loss of Cdkn2a, can overcome the lymphoid deficiencies caused by the absence of Hhex, suggesting that the primary role of this transcription factor is to promote survival of lymphoid progenitors during early lymphoid development.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/immunology , Homeodomain Proteins/immunology , Lymphoid Progenitor Cells/immunology , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , Transcription Factors/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Cell Survival/genetics , Cell Survival/immunology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Homeodomain Proteins/genetics , Lymphoid Progenitor Cells/cytology , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/immunology , STAT5 Transcription Factor/genetics , Signal Transduction/genetics , Transcription Factors/genetics
12.
Leukemia ; 33(8): 1868-1880, 2019 08.
Article in English | MEDLINE | ID: mdl-30700838

ABSTRACT

T cell acute lymphoblastic leukaemia (T-ALL) cases include subfamilies that overexpress the TAL1/LMO, TLX1/3 and HOXA transcription factor oncogenes. While it has been shown that TAL1/LMO transcription factors induce self-renewal of thymocytes, whether this is true for other transcription factor oncogenes is unknown. To address this, we have studied NUP98-HOXD13-transgenic (NHD13-Tg) mice, which overexpress HOXA transcription factors throughout haematopoiesis and develop both myelodysplastic syndrome (MDS) progressing to acute myeloid leukaemia (AML) as well as T-ALL. We find that thymocytes from preleukaemic NHD13-Tg mice can serially transplant, demonstrating that they have self-renewal capacity. Transcriptome analysis shows that NHD13-Tg thymocytes exhibit a stem cell-like transcriptional programme closely resembling that induced by Lmo2, including Lmo2 itself and its critical cofactor Lyl1. To determine whether Lmo2/Lyl1 are required for NHD13-induced thymocyte self-renewal, NHD13-Tg mice were crossed with Lyl1 knockout mice. This showed that Lyl1 is essential for expression of the stem cell-like gene expression programme in thymocytes and self-renewal. Surprisingly however, NHD13 transgenic mice lacking Lyl1 showed accelerated T-ALL and absence of transformation to AML, associated with a loss of multipotent progenitors in the bone marrow. Thus multiple T cell oncogenes induce thymocyte self-renewal via Lmo2/Lyl1; however, NHD13 can also promote T-ALL via an alternative pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Basic Helix-Loop-Helix Transcription Factors/physiology , Homeodomain Proteins/genetics , LIM Domain Proteins/physiology , Neoplasm Proteins/physiology , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Thymocytes/physiology , Transcription Factors/genetics , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
13.
Blood Adv ; 2(4): 347-360, 2018 02 27.
Article in English | MEDLINE | ID: mdl-29453249

ABSTRACT

The hematopoietically expressed homeobox (Hhex) transcription factor is overexpressed in human myeloid leukemias. Conditional knockout models of murine acute myeloid leukemia indicate that Hhex maintains leukemia stem cell self-renewal by enabling Polycomb-mediated epigenetic repression of the Cdkn2a tumor suppressor locus, encoding p16Ink4a and p19Arf However, whether Hhex overexpression also affects hematopoietic differentiation is unknown. To study this, we retrovirally overexpressed Hhex in hematopoietic progenitors. This enabled serial replating of myeloid progenitors, leading to the rapid establishment of interleukin-3 (IL-3)-dependent promyelocytic cell lines. Use of a Hhex-ERT2 fusion protein demonstrated that continuous nuclear Hhex is required for transformation, and structure function analysis demonstrated a requirement of the DNA-binding and N-terminal-repressive domains of Hhex for promyelocytic transformation. This included the N-terminal promyelocytic leukemia protein (Pml) interaction domain, although deletion of Pml failed to prevent Hhex-induced promyelocyte transformation, implying other critical partners. Furthermore, deletion of p16Ink4a or p19Arf did not promote promyelocyte transformation, indicating that repression of distinct Hhex target genes is required for this process. Indeed, transcriptome analysis showed that Hhex overexpression resulted in repression of several myeloid developmental genes. To test the potential for Hhex overexpression to contribute to leukemic transformation, Hhex-transformed promyelocyte lines were rendered growth factor-independent using a constitutively active IL-3 receptor common ß subunit (ßcV449E). The resultant cell lines resulted in a rapid promyelocytic leukemia in vivo. Thus, Hhex overexpression can contribute to myeloid leukemia via multiple mechanisms including differentiation blockade and enabling epigenetic repression of the Cdkn2a locus.


Subject(s)
Cell Self Renewal , Granulocyte Precursor Cells/cytology , Homeodomain Proteins/physiology , Intercellular Signaling Peptides and Proteins , Leukemia, Myeloid/etiology , Transcription Factors/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation, Leukemic , Homeodomain Proteins/metabolism , Mice , Transcription Factors/metabolism
14.
Stem Cells ; 35(8): 1948-1957, 2017 08.
Article in English | MEDLINE | ID: mdl-28577303

ABSTRACT

The hematopoietically expressed homeobox transcription factor (Hhex) is important for the maturation of definitive hematopoietic progenitors and B-cells during development. We have recently shown that in adult hematopoiesis, Hhex is dispensable for maintenance of hematopoietic stem cells (HSCs) and myeloid lineages but essential for the commitment of common lymphoid progenitors (CLPs) to lymphoid lineages. Here, we show that during serial bone marrow transplantation, Hhex-deleted HSCs are progressively lost, revealing an intrinsic defect in HSC self-renewal. Moreover, Hhex-deleted mice show markedly impaired hematopoietic recovery following myeloablation, due to a failure of progenitor expansion. In vitro, Hhex-null blast colonies were incapable of replating, implying a specific requirement for Hhex in immature progenitors. Transcriptome analysis of Hhex-null Lin- Sca+ Kit+ cells showed that Hhex deletion leads to derepression of polycomb repressive complex 2 (PRC2) and PRC1 target genes, including the Cdkn2a locus encoding the tumor suppressors p16Ink 4a and p19Arf . Indeed, loss of Cdkn2a restored the capacity of Hhex-null blast colonies to generate myeloid progenitors in vitro, as well as hematopoietic reconstitution following myeloablation in vivo. Thus, HSCs require Hhex to promote PRC2-mediated Cdkn2a repression to enable continued self-renewal and response to hematopoietic stress. Stem Cells 2017;35:1948-1957.


Subject(s)
Cell Self Renewal , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Stress, Physiological , Transcription Factors/metabolism , Animals , Cell Proliferation , Gene Deletion , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Mice, Inbred C57BL , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism
15.
Genes Dev ; 30(1): 78-91, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728554

ABSTRACT

Unlike clustered HOX genes, the role of nonclustered homeobox gene family members in hematopoiesis and leukemogenesis has not been extensively studied. Here we found that the hematopoietically expressed homeobox gene Hhex is overexpressed in acute myeloid leukemia (AML) and is essential for the initiation and propagation of MLL-ENL-induced AML but dispensable for normal myelopoiesis, indicating a specific requirement for Hhex for leukemic growth. Loss of Hhex leads to expression of the Cdkn2a-encoded tumor suppressors p16(INK4a) and p19(ARF), which are required for growth arrest and myeloid differentiation following Hhex deletion. Mechanistically, we show that Hhex binds to the Cdkn2a locus and directly interacts with the Polycomb-repressive complex 2 (PRC2) to enable H3K27me3-mediated epigenetic repression. Thus, Hhex is a potential therapeutic target that is specifically required for AML stem cells to repress tumor suppressor pathways and enable continued self-renewal.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/physiopathology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Gene Deletion , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Inbred C57BL , Protein Binding , Transcription Factors/genetics
16.
Blood ; 125(5): 803-14, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25472970

ABSTRACT

The hematopoietically expressed homeobox gene, Hhex, is a transcription factor that is important for development of definitive hematopoietic stem cells (HSCs) and B cells, and that causes T-cell leukemia when overexpressed. Here, we have used an Hhex inducible knockout mouse model to study the role of Hhex in adult hematopoiesis. We found that loss of Hhex was tolerated in HSCs and myeloid lineages, but resulted in a progressive loss of B lymphocytes in the circulation. This was accompanied by a complete loss of B-cell progenitors in the bone marrow and of transitional B-cell subsets in the spleen. In addition, transplantation and in vitro culture experiments demonstrated an almost complete failure of Hhex-null HSCs to contribute to lymphoid lineages beyond the common lymphoid precursor stage, including T cells, B cells, NK cells, and dendritic cells. Gene expression analysis of Hhex-deleted progenitors demonstrated deregulated expression of a number of cell cycle regulators. Overexpression of one of these, cyclin D1, could rescue the B-cell developmental potential of Hhex-null lymphoid precursors. Thus, Hhex is a key regulator of early lymphoid development, functioning, at least in part, via regulation of the cell cycle.


Subject(s)
Cell Cycle Proteins/genetics , Cyclin D1/genetics , Hematopoiesis/genetics , Homeodomain Proteins/genetics , Lymphopoiesis/genetics , Precursor Cells, B-Lymphoid/pathology , Transcription Factors/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Cycle Proteins/immunology , Cell Differentiation , Cell Proliferation , Cyclin D1/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Deletion , Gene Expression Regulation , Genetic Complementation Test , Hematopoiesis/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Lymphocyte Count , Lymphocyte Depletion , Lymphopoiesis/immunology , Mice , Mice, Knockout , Precursor Cells, B-Lymphoid/immunology , Spleen/immunology , Spleen/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcription Factors/deficiency , Transcription Factors/immunology , Transcription, Genetic
17.
Blood ; 122(12): 2093-103, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23926305

ABSTRACT

Lmo2 is an oncogenic transcription factor that is frequently overexpressed in T-cell acute lymphoblastic leukemia (T-ALL), including early T-cell precursor ALL (ETP-ALL) cases with poor prognosis. Lmo2 must be recruited to DNA by binding to the hematopoietic basic helix-loop-helix factors Scl/Tal1 or Lyl1. However, it is unknown which of these factors can mediate the leukemic activity of Lmo2. To address this, we have generated Lmo2-transgenic mice lacking either Scl or Lyl1 in the thymus. We show that although Scl is dispensable for Lmo2-driven leukemia, Lyl1 is critical for all oncogenic functions of Lmo2, including upregulation of a stem cell-like gene signature, aberrant self-renewal of thymocytes, and subsequent generation of T-cell leukemia. Lyl1 expression is restricted to preleukemic and leukemic stem cell populations in this model, providing a molecular explanation for the stage-specific expression of the Lmo2-induced gene expression program. Moreover, LMO2 and LYL1 are coexpressed in ETP-ALL patient samples, and LYL1 is required for growth of ETP-ALL cell lines. Thus, the LMO2-LYL1 interaction is a promising therapeutic target for inhibiting self-renewing cancer stem cells in T-ALL, including poor-prognosis ETP-ALL cases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , LIM Domain Proteins/genetics , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , LIM Domain Proteins/metabolism , Mice , Mice, Transgenic , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/mortality , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , T-Cell Acute Lymphocytic Leukemia Protein 1 , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thymocytes/metabolism , Thymocytes/pathology
18.
Blood ; 121(9): 1574-83, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23297132

ABSTRACT

Antiviral immunity and cross-presentation is mediated constitutively through CD8α+ and CD103+ DCs. Development of these DC subsets is thought to require the transcription factors Irf8, Id2, Nfil3, and Batf3, although how this network is regulated is poorly defined. We addressed the nature of the differentiation blocks observed in the absence of these factors and found that although all 4 factors are required for CD103+ DC development, only Irf8 is essential for CD8α+ DCs. CD8α+ DCs emerged in the absence of Id2, Nfil3 and Batf3 in short-term bone marrow reconstitution. These "induced" CD8α+ DCs exhibit several hallmarks of classic CD8α+ DCs including the expression of CD24, Tlr3, Xcr1, Clec9A, and the capacity to cross-present soluble, cell-associated antigens and viral antigens even in the absence of Batf3. Collectively, these results uncover a previously undescribed pathway by which CD8α+ DCs emerge independent of Id2, Nfil3, and Batf3, but dependent on Irf8.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , CD8 Antigens/metabolism , Cell Differentiation , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/physiology , Repressor Proteins/physiology , Animals , Antigens, Viral/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Cross-Priming/genetics , Cross-Priming/immunology , Cross-Priming/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Herpesvirus 1, Human/immunology , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/physiology
19.
Phys Biol ; 8(6): 066002, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21946082

ABSTRACT

The epidermal growth factor receptor (EGFR) is a member of the erbB tyrosine kinase family of receptors. Structural studies have revealed two distinct conformations of the ectodomain of the EGFR: a compact, tethered, conformation and an untethered extended conformation. In the context of a monomer-dimer transition model, ligand binding is thought to untether the monomeric receptor leading to exposure of a dimerization arm which then facilitates receptor dimerization, kinase activation and signaling. For receptors directed orthogonal to the local plane of the membrane surface, this would lead to a large change in the distance of the receptor N-terminus from the membrane surface. To investigate this experimentally, we produced stable BaF/3 cell lines expressing a biochemically functional yellow fluorescent protein (YFP)-EGFR chimera and determined the vertical separation of the N-terminal YFP tag from the membrane using fluorescence resonance energy transfer (FRET) techniques. Homo-FRET/rFLIM was employed to determine the presence of unliganded dimers and to measure the average distance between the N-terminal tags in those dimers. The results suggest that EGF-induced activation occurs within or between pre-formed and extended dimers with very little change in the extension of the N-terminii from the membrane surface. These results provide constraints on possible models for EGFR activation.


Subject(s)
Bacterial Proteins/chemistry , ErbB Receptors/chemistry , Luminescent Proteins/chemistry , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fluorescence Resonance Energy Transfer , Gene Expression , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Models, Molecular , Protein Binding , Protein Conformation , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
EMBO J ; 30(13): 2690-704, 2011 May 17.
Article in English | MEDLINE | ID: mdl-21587207

ABSTRACT

Dendritic cells (DCs) have critical roles in the induction of the adaptive immune response. The transcription factors Id2, Batf3 and Irf-8 are required for many aspects of murine DC differentiation including development of CD8α(+) and CD103(+) DCs. How they regulate DC subset specification is not completely understood. Using an Id2-GFP reporter system, we show that Id2 is broadly expressed in all cDC subsets with the highest expression in CD103(+) and CD8α(+) lineages. Notably, CD103(+) DCs were the only DC able to constitutively cross-present cell-associated antigens in vitro. Irf-8 deficiency affected loss of development of virtually all conventional DCs (cDCs) while Batf3 deficiency resulted in the development of Sirp-α(-) DCs that had impaired survival. Exposure to GM-CSF during differentiation induced expression of CD103 in Id2-GFP(+) DCs. It did not restore cross-presenting capacity to Batf3(-/-) or CD103(-)Sirp-α(-)DCs in vitro. Thus, Irf-8 and Batf3 regulate distinct stages in DC differentiation during the development of cDCs. Genetic mapping DC subset differentiation using Id2-GFP may have broad implications in understanding the interplay of DC subsets during protective and pathological immune responses.


Subject(s)
Antigens, CD/metabolism , CD8 Antigens/metabolism , Cell Lineage/genetics , Dendritic Cells/physiology , Inhibitor of Differentiation Protein 2/genetics , Integrin alpha Chains/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression/physiology , Genes, cdc/physiology , Inhibitor of Differentiation Protein 2/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological
SELECTION OF CITATIONS
SEARCH DETAIL
...