Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 108(19): 7956-61, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518888

ABSTRACT

Rearrangements of the MLL (ALL1) gene are very common in acute infant and therapy-associated leukemias. The rearrangements underlie the generation of MLL fusion proteins acting as potent oncogenes. Several most consistently up-regulated targets of MLL fusions, MEIS1, HOXA7, HOXA9, and HOXA10 are functionally related and have been implicated in other types of leukemias. Each of the four genes was knocked down separately in the human precursor B-cell leukemic line RS4;11 expressing MLL-AF4. The mutant and control cells were compared for engraftment in NOD/SCID mice. Engraftment of all mutants into the bone marrow (BM) was impaired. Although homing was similar, colonization by the knockdown cells was slowed. Initially, both types of cells were confined to the trabecular area; this was followed by a rapid spread of the WT cells to the compact bone area, contrasted with a significantly slower process for the mutants. In vitro and in vivo BrdU incorporation experiments indicated reduced proliferation of the mutant cells. In addition, the CXCR4/SDF-1 axis was hampered, as evidenced by reduced migration toward an SDF-1 gradient and loss of SDF-1-augmented proliferation in culture. The very similar phenotype shared by all mutant lines implies that all four genes are involved and required for expansion of MLL-AF4 associated leukemic cells in mice, and down-regulation of any of them is not compensated by the others.


Subject(s)
Genes, Homeobox , Homeodomain Proteins/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Knockdown Techniques , Gene Rearrangement , Histone-Lysine N-Methyltransferase , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Transplantation , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Small Interfering/genetics , Transplantation, Heterologous
2.
Nucleic Acids Res ; 38(16): 5472-8, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20444872

ABSTRACT

HOXA9-mediated up-regulation of miR-155 was noted during an array-based analysis of microRNA expression in Hoxa9(-/-)bone marrow (BM) cells. HOXA9 induction of miR-155 was confirmed in these samples, as well as in wild-type versus Hoxa9-deficient marrow, using northern analysis and qRT-PCR. Infection of wild-type BM with HOXA9 expressing or GFP(+) control virus further confirmed HOXA9-mediated regulation of miR-155. miR-155 expression paralleled Hoxa9 mRNA expression in fractionated BM progenitors, being highest in the stem cell enriched pools. HOXA9 capacity to induce myeloid colony formation was blunted in miR-155-deficient BM cells, indicating that miR-155 is a downstream mediator of HOXA9 function in blood cells. Pu.1, an important regulator of myelopoiesis, was identified as a putative down stream target for miR-155. Although miR-155 was shown to down-regulate the Pu.1 protein, HOXA9 did not appear to modulate Pu.1 expression in murine BM cells.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Animals , Cells, Cultured , Homeodomain Proteins/genetics , Mice , MicroRNAs/biosynthesis , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Up-Regulation
3.
Mol Cell Biol ; 29(18): 5181-92, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19620287

ABSTRACT

While investigating the mechanism of action of the HOXA9 protein, we serendipitously identified Meis1 as a HOXA9 regulatory target. Since HOXA9 and MEIS1 play key developmental roles, are cooperating DNA binding proteins and leukemic oncoproteins, and are important for normal hematopoiesis, the regulation of Meis1 by its partner protein is of interest. Loss of Hoxa9 caused downregulation of the Meis1 mRNA and protein, while forced HOXA9 expression upregulated Meis1. Hoxa9 and Meis1 expression was correlated in hematopoietic progenitors and acute leukemias. Meis1(+/-) Hoxa9(-/-) deficient mice, generated to test HOXA9 regulation of endogenous Meis1, were small and had reduced bone marrow Meis1 mRNA and significant defects in fluorescence-activated cell sorting-enumerated monocytes, mature and pre/pro-B cells, and functional B-cell progenitors. These data indicate that HOXA9 modulates Meis1 during normal murine hematopoiesis. Chromatin immunoprecipitation analysis did not reveal direct binding of HOXA9 to Meis1 promoter/enhancer regions. However, Creb1 and Pknox1, whose protein products have previously been reported to induce Meis1, were shown to be direct targets of HOXA9. Loss of Hoxa9 resulted in a decrease in Creb1 and Pknox1 mRNA, and forced expression of CREB1 in Hoxa9(-/-) bone marrow cells increased Meis1 mRNA almost as well as HOXA9, suggesting that CREB1 may mediate HOXA9 modulation of Meis1 expression.


Subject(s)
Genes, Neoplasm , Hematopoiesis , Homeodomain Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Body Weight , Crosses, Genetic , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Embryo Implantation , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Heterozygote , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid/genetics , Male , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/genetics
4.
Mol Cell Biol ; 28(14): 4609-19, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18474618

ABSTRACT

The PicTar program predicted that microRNA-126 (miR-126), miR-145, and let-7s target highly conserved sites within the Hoxa9 homeobox. There are increased nucleotide constraints in the three microRNA seed sites among Hoxa9 genes beyond that required to maintain protein identity, suggesting additional functional conservation. In preliminary experiments, forced expression of these microRNAs in Hoxa9-immortalized bone marrow cells downregulated the HOXA9 protein and caused loss of biological activity. The microRNAs were shown to target their predicted sites within the homeobox. miR-126 and Hoxa9 mRNA are coexpressed in hematopoietic stem cells and downregulated in parallel during progenitor cell differentiation; however, miR-145 is barely detectable in hematopoietic cells, and let-7s are highly expressed in bone marrow progenitors, suggesting that miR-126 may function in normal hematopoietic cells to modulate HOXA9 protein. In support of this hypothesis, expression of miR-126 alone in MLL-ENL-immortalized bone marrow cells decreased endogenous HOXA9 protein, while inhibition of endogenous miR-126 increased expression of HOXA9 in F9 cells.


Subject(s)
Genes, Homeobox , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Amino Acid Sequence , Animals , Base Sequence , Bone Marrow Cells/metabolism , Cell Line, Tumor , Conserved Sequence , Down-Regulation , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Mice , Molecular Sequence Data , Sequence Alignment
5.
Blood ; 109(11): 4732-8, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17327400

ABSTRACT

The HOXA9 homeoprotein exerts dramatic effects in hematopoiesis. Enforced expression of HOXA9 enhances proliferation of primitive blood cells, expands hematopoietic stem cells (HSCs), and leads to myeloid leukemia. Conversely, loss of HOXA9 inhibits proliferation and impairs HSC function. The pathways by which HOXA9 acts are largely unknown, and although HOXA9 is a transcription factor, few direct target genes have been identified. Our previous study suggested that HOXA9 positively regulates Pim1, an oncogenic kinase. The hematologic phenotypes of Hoxa9- and Pim1-deficient animals are strikingly similar. Here we show that HOXA9 protein binds to the Pim1 promoter and induces Pim1 mRNA and protein in hematopoietic cells. Pim1 protein is diminished in Hoxa9(-/-) cells, and Hoxa9 and Pim1 mRNA levels track together in early hematopoietic compartments. Induction of Pim1 protein by HOXA9 increases the phosphorylation and inactivation of the proapoptotic BAD protein, a target of Pim1. Hoxa9(-/-) cells show increased apoptosis and decreased proliferation, defects that are ameliorated by reintroduction of Pim1. Thus Pim1 appears to be a direct transcriptional target of HOXA9 and a mediator of its antiapoptotic and proproliferative effects in early cells. Since HOXA9 is frequently up-regulated in acute myeloid leukemia, Pim1 may be a therapeutic target in human disease.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Homeodomain Proteins/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Apoptosis , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , K562 Cells , Leukemia, Myeloid/metabolism , Phosphorylation , RNA, Messenger/metabolism , Retroviridae/genetics , Time Factors , Transgenes , U937 Cells
6.
Blood ; 106(12): 3988-94, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16091451

ABSTRACT

The homeobox gene Hoxa-9 is normally expressed in primitive bone marrow cells, and overexpression of Hoxa-9 markedly expands hematopoietic stem cells, suggesting a function in early hematopoiesis. We present evidence for major functional defects in Hoxa-9-/- hematopoietic stem cells. Hoxa-9-/- marrow cells have normal numbers of immunophenotypic stem cells (Lin(-)c-kit(+)flk-2(-)Sca-1+ [KLFS] cells). However, sublethally irradiated Hoxa-9-/- mice develop persistent pancytopenia, indicating unusual sensitivity to ionizing irradiation. In competitive transplantation assays, Hoxa-9-/- cells showed an 8-fold reduction in multilineage long-term repopulating ability, a defect not seen in marrow cells deficient for the adjacent Hoxa-10 gene. Single-cell cultures of KLFS cells showed a 4-fold reduction in large high-proliferation potential colonies. In liquid cultures, Hoxa-9-deficient Lin(-)Sca-1(+) cells showed slowed proliferation (a 5-fold reduction in cell numbers at day 8) and delayed emergence of committed progenitors (a 5-fold decrease in colony-forming cells). Slowing of proliferation was accompanied by a delay in myeloid maturation, with a decrease in Gr-1hiMac-1hi cells at the end of the culture. Retroviral transduction with a Hoxa-9 expression vector dramatically enhanced the cytokine-driven proliferation and in vivo engraftment of Hoxa-9-/- marrow cells. Hoxa-9 appears to be specifically required for normal hematopoietic stem cell function both in vitro and in vivo.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Animals , Cell Movement/physiology , Cell Proliferation , Flow Cytometry , Gene Expression , Hematopoiesis/genetics , Mice , Mice, Mutant Strains
7.
Stem Cells ; 23(5): 644-55, 2005 May.
Article in English | MEDLINE | ID: mdl-15849172

ABSTRACT

There is growing evidence for a role of HOX homeodomain proteins in normal hematopoiesis. Several HOX genes, including HOXA9 and HOXA10, are expressed in primitive hematopoietic cells, implying a role in early hematopoietic differentiation. To identify potential target genes of these two closely related transcription factors, human CD34+ umbilical cord blood cells were transduced with vectors expressing either HOXA9 or HOXA10 and analyzed with cDNA micro-arrays. Statistical analysis using significance analysis of microarrays revealed a common signature of several hundred genes, demonstrating that the transcriptomes of HOXA9 and HOXA10 largely overlap in this cellular context. Seven genes that were upregulated by both HOX proteins were validated by real-time reverse transcription polymerase chain reaction. HOXA9 and HOXA10 showed positive regulation of genes in the Wnt pathway, including Wnt10B and two Wnt receptors Frizzled 1 and Frizzled 5, an important pathway for hematopoietic stem cell (HSC) self-renewal. Other validated genes included v-ets-related gene (ERG), Iroquois 3 (IRX3), aldehyde dehydrogenase 1 (ALDH1), and very long-chain acyl-CoA synthetase homolog 1 (VLCS-H1). GenMAPP (Gene Micro Array Pathway Profiler) analysis indicated that HOXA10 repressed expression of several genes involved in heme biosynthesis and three globin genes, indicating a general suppression of erythroid differentiation. A number of genes regulated by HOXA9 and HOXA10 are expressed in normal HSC populations.


Subject(s)
Antigens, CD34 , Fetal Blood/physiology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/biosynthesis , Up-Regulation/physiology , Animals , Cells, Cultured , Fetal Blood/cytology , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Homeobox A10 Proteins , Humans , Mice , Oligonucleotide Array Sequence Analysis
8.
Methods Mol Biol ; 289: 157-70, 2005.
Article in English | MEDLINE | ID: mdl-15502181

ABSTRACT

HOX homeodomain proteins are thought to be master developmental regulators of tissue patterning during embryogenesis. These DNA binding proteins also have diverse roles in adult cell function, and derangement of HOX genes has been associated with several types of cancer. In this chapter we present protocols for the immunohistochemical localization of HOX proteins in the epidermis. We also provide in situ hybridization protocols for detection of HOX gene mRNA transcripts in the epidermis.


Subject(s)
Epidermal Cells , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , RNA, Messenger/genetics , Animals , Epidermis/metabolism , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Keratinocytes
9.
Blood ; 105(4): 1456-66, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15522959

ABSTRACT

The HOX family of homeobox genes plays an important role in normal and malignant hematopoiesis. Dysregulated HOX gene expression profoundly effects the proliferation and differentiation of hematopoietic stem cells (HSCs) and committed progenitors, and aberrant activation of HOX genes is a common event in human myeloid leukemia. HOXB6 is frequently overexpressed in human acute myeloid leukemia (AML). To gain further insight into the role of HOXB6 in hematopoiesis, we overexpressed HOXB6 in murine bone marrow using retrovirus-mediated gene transfer. We also explored structure-function relationships using mutant HOXB6 proteins unable to bind to DNA or a key HOX-binding partner, pre-B-cell leukemia transcription factor-1 (PBX1). Additionally, we investigated the potential cooperative interaction with myeloid ecotropic viral integration site 1 homolog (MEIS1). In vivo, HOXB6 expanded HSCs and myeloid precursors while inhibiting erythropoiesis and lymphopoiesis. Overexpression of HOXB6 resulted in AML with a median latency of 223 days. Coexpression of MEIS1 dramatically shortened the onset of AML. Cytogenetic analysis of a subset of HOXB6-induced AMLs revealed recurrent deletions of chromosome bands 2D-E4, a region frequently deleted in HOXA9-induced AMLs. In vitro, HOXB6 immortalized a factor-dependent myelomonocytic precursor capable of granulocytic and monocytic differentiation. These biologic effects of HOXB6 were largely dependent on DNA binding but independent of direct interaction with PBX1.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/pathology , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Leukemia, Myeloid/blood , Myeloid Progenitor Cells/metabolism , Myeloid Progenitor Cells/pathology , Acute Disease , Animals , Cell Differentiation/genetics , Cell Line, Transformed , Cell Proliferation , Erythropoiesis/genetics , Female , Homeodomain Proteins/physiology , Karyotyping , Leukemia, Myeloid/genetics , Leukemia, Myeloid/pathology , Lymphopoiesis/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/physiology , Phenotype , Time Factors
10.
FASEB J ; 18(13): 1481-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15466355

ABSTRACT

Over the past three decades the Veterans Affairs (VA) Research program has evolved into a powerful, peer-reviewed funding mechanism for basic and translational research that has resulted in numerous important contributions to medical science and improvements in patient care. Continuity in VA Merit Review funding has fostered and nurtured the scientific careers of a large number of physician-scientists who have remained devoted to the mission of performing creative and innovative research that affects the patient care mission of the VA. VA medical research policies have undergone a major overhaul in the past year. Although many of these changes (de-emphasizing bench research and revamping the peer review process) have recently been reversed, the future direction of VA research remains in flux. The goal of this manuscript is to demonstrate the importance of the Merit Review medical research funding mechanism not just to the VA, but to the entire nation's health care system. To achieve this goal, the opinions of 65 established VA medical investigators were obtained regarding the past success and future direction of VA research. The conclusions reached include the following. 1) Merit Review research funding has been essential to the training, recruitment, and retention of productive VA physician-scientists. 2) The VA research program has contributed both basic and clinical innovations that have led to improvements in medical care. Contributions of VA researchers to excellence in many aspects of patient care at VA hospitals have been extraordinary. 3) Development of initiatives that entice outstanding Ph.D.'s to develop their careers in the VA has been crucial to the success of the program. 4) The VA research program has fostered a mutually beneficial relationship with affiliated medical schools. 5) Better methods to quantify VA research contributions and outcomes are essential for future program development.


Subject(s)
Biomedical Research/standards , Physicians/standards , Research Personnel/standards , Research Support as Topic , United States Department of Veterans Affairs/organization & administration , Biomedical Research/history , Biomedical Research/trends , Data Collection , Delivery of Health Care/history , Delivery of Health Care/standards , Education, Medical/history , Education, Medical/standards , History, 18th Century , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Patient Care/history , Patient Care/standards , Personnel Selection , Physicians/history , Physicians/supply & distribution , Research Personnel/education , Research Personnel/history , Research Personnel/supply & distribution , Research Support as Topic/history , Research Support as Topic/trends , Schools, Medical/organization & administration , United States , United States Department of Veterans Affairs/economics , United States Department of Veterans Affairs/history , Veterans , Workforce
11.
J Biol Chem ; 279(38): 39895-904, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15269212

ABSTRACT

Although HOXB6 and other HOX genes have previously been associated with hematopoiesis and leukemias, the precise mechanism of action of their protein products remains unclear. Here we use a biological model in which HOXB6 represses alpha- and gamma-globin mRNA levels to perform a structure/function analysis for this homeodomain protein. HOXB6 protein represses globin transcript levels in stably transfected K562 cells in a DNA-binding dependent fashion. However, the capacity to form cooperative DNA-binding complexes with the PBX co-factor protein is not required for HOXB6 biological activity. Neither the conserved extreme N-terminal region, a polyglutamic acid region at the protein C terminus, nor the Ser(214) CKII phosphorylation site was required for DNA binding or activity in this model. We have previously reported that HOX proteins can inhibit CREB-binding protein (CBP)-histone acetyltransferase-mediated potentiation of reporter gene transcription. We now show that endogenous CBP is co-precipitated with exogenous HOXB6 from nuclear and cytoplasmic compartments of transfected K562 cells. Furthermore, endogenous CBP co-precipitates with endogenous HOXB6 in day 14.5 murine fetal liver cells during active globin gene expression in this tissue. The CBP interaction motif was localized to the homeodomain but does not require the highly conserved helix 3. Our data suggest that the homeodomain contains most or all of the important structures required for HOXB6 activity in blood cells.


Subject(s)
Globins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Acetyltransferases/metabolism , Animals , CREB-Binding Protein , DNA/metabolism , Gene Expression Regulation, Developmental , Histone Acetyltransferases , Homeodomain Proteins/chemistry , Humans , K562 Cells , Liver/cytology , Mice , Precipitin Tests , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Messenger/metabolism , Serine/genetics , Transfection
12.
Mol Cell Biol ; 24(9): 3827-37, 2004 May.
Article in English | MEDLINE | ID: mdl-15082777

ABSTRACT

HOXA9 expression is a common feature of acute myeloid leukemia, and high-level expression is correlated with poor prognosis. Moreover, HOXA9 overexpression immortalizes murine marrow progenitors that are arrested at a promyelocytic stage of differentiation when cultured and causes leukemia in recipient mice following transplantation of HOXA9 expressing bone marrow. The molecular mechanisms underlying the physiologic functions and transforming properties of HOXA9 are poorly understood. This study demonstrates that HOXA9 is phosphorylated by protein kinase C (PKC) and casein kinase II and that PKC mediates phosphorylation of purified HOXA9 on S204 as well as on T205, within a highly conserved consensus sequence, in the N-terminal region of the homeodomain. S204 in the endogenous HOXA9 protein was phosphorylated in PLB985 myeloid cells, as well as in HOXA9-immortalized murine marrow cells. This phosphorylation was enhanced by phorbol ester, a known inducer of PKC, and was inhibited by a specific PKC inhibitor. PKC-mediated phosphorylation of S204 decreased HOXA9 DNA binding affinity in vitro and the ability of the endogenous HOXA9 to form cooperative DNA binding complexes with PBX. PKC inhibition significantly reduced the phorbol-ester induced differentiation of the PLB985 hematopoietic cell line as well as HOXA9-immortalized murine bone marrow cells. These data suggest that phorbol ester-induced myeloid differentiation is in part due to PKC-mediated phosphorylation of HOXA9, which decreases the DNA binding of the homeoprotein.


Subject(s)
Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Myeloid Cells/physiology , Protein Kinase C/metabolism , Amino Acid Sequence , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Casein Kinase II , Cell Line , Enzyme Activation , Isoenzymes/metabolism , Leukemia, Myeloid , Mice , Molecular Sequence Data , Myeloid Cells/cytology , Phorbol Esters/metabolism , Phosphorylation , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism
13.
Blood ; 103(5): 1676-84, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14604967

ABSTRACT

Hematopoietic defects in HOXA9(-/-) mice demonstrate a key role for this homeoprotein in blood cell development. Conversely, enforced HOXA9 expression is leukemogenic in mice, and HOXA9 is frequently activated in human acute myeloid leukemia (AML). Although HOXA9 is thought to function as a transcription factor, few downstream targets have been identified. We searched for early HOXA9 target genes by using a transient overexpression strategy in 3 hematopoietic cell lines (2 myeloid, 1 lymphoid). cDNA microarray analyses identified 220 genes whose expression was modulated at least 2-fold. Expression signatures in myeloid and lymphoid cells demonstrated that HOXA9 functions as both an activator and repressor of a variety of genes in cell-specific patterns suggesting that the transcriptional effects of HOXA9 are largely dependent on the cell context. Transient transcription assays and target gene expression patterns in HOXA9(-/-) marrow cells imply that we have identified direct physiologic targets. Many target genes are expressed in CD34+ stem cells or are members of gene families involved in proliferation or myeloid differentiation. Expression of 14 HOXA9 target genes correlated with high-level HOXA9 expression in primary AML. These data suggest that many genes identified in this survey may mediate the biologic effects of HOXA9 in normal and leukemic hematopoiesis.


Subject(s)
Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/biosynthesis , Leukemia/metabolism , RNA, Messenger/metabolism , Blotting, Western , Bone Marrow Cells/cytology , Cell Division , DNA/chemistry , DNA, Complementary/metabolism , Down-Regulation , Homeodomain Proteins/genetics , Humans , Jurkat Cells , K562 Cells , Leukemia/genetics , Luciferases/metabolism , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection , U937 Cells , Up-Regulation
14.
Dev Dyn ; 227(2): 192-202, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12761847

ABSTRACT

Substantial evidence suggests that HOX homeobox genes regulate aspects of body development, including hair formation. We initially isolated the HOXB13 gene from human fetal skin in experiments designed to identify candidate genes that regulate scarless fetal wound healing. Although the HOX homeodomain proteins have been proposed to function as transcription factors, we have demonstrated previously that substantial fractions of the HOXB6 and HOXB4 proteins are localized to the cytoplasm throughout epidermal development. The purpose of the current study was to identify HOXB13 protein expression patterns in developing skin to elucidate potential mechanisms by which this protein might regulate aspects of tissue development and healing. HOXB13 protein expression was detected throughout the developing epidermis, with weaker signal observed in the early developing dermis. Epidermal HOXB13 signal was detected over the entire body surface, but surprisingly, essentially all of the signal was cytoplasmic in developing skin. Low-level HOXB13 protein expression was detected in adult skin and within the telogen hair follicle, and a portion of the residual signal in adult epidermis was nuclear. Expression in hyperproliferative skin conditions remained cytoplasmic with the exception of epidermis associated with Kaposi's sarcoma, which showed strong HOXB13 expression that was partially localized to the nucleus.


Subject(s)
Epidermis/embryology , Epidermis/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Animals , Chromosomes , Cytoplasm/metabolism , Gene Expression Regulation, Developmental , Hair Follicle/embryology , Hair Follicle/physiology , Humans , Mice , Mice, Inbred C57BL , Skin Diseases/physiopathology
15.
FASEB J ; 17(10): 1352-4, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12759339

ABSTRACT

In contrast to adult cutaneous wound repair, early gestational fetal cutaneous wounds heal by a process of regeneration, resulting in little or no scarring. Previous studies indicate that down-regulation of HoxB13, a member of the highly conserved family of Hox transcription factors, occurs during fetal scarless wound healing. No down-regulation was noted in adult wounds. Here, we evaluate healing of adult cutaneous wounds in Hoxb13 knockout (KO) mice, hypothesizing that loss of Hoxb13 in adult skin should result in enhanced wound healing. Tensiometry was used to measure the tensile strength of incisional wounds over a 60-day time course; overall, Hoxb13 KO wounds are significantly stronger than wild-type (WT). Histological evaluation of incisional wounds shows that 7-day-old Hoxb13 KO wounds are significantly smaller and that 60-day-old Hoxb13 KO wounds exhibit a more normal collagen architecture compared with WT wounds. We also find that excisional wounds close at a faster rate in Hoxb13 KO mice. Biochemical and histochemical analyses show that Hoxb13 KO skin contains significantly elevated levels of hyaluronan. Because higher levels of hyaluronan and enhanced wound healing are characteristics of fetal skin, we conclude that loss of Hoxb13 produces a more "fetal-like" state in adult skin.


Subject(s)
Homeodomain Proteins/physiology , Hyaluronic Acid/biosynthesis , Skin Physiological Phenomena , Skin/metabolism , Wound Healing , Animals , Dermis/anatomy & histology , Homeodomain Proteins/genetics , Kinetics , Mice , Mice, Knockout , Skin/anatomy & histology
16.
J Invest Dermatol ; 120(1): 135-44, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12535210

ABSTRACT

The phenotype of fibroblasts repopulating experimental wounds in vivo has been shown to influence both wound healing responses and clinical outcome. Recent studies have demonstrated that the human homeobox gene PRX-2 is strongly upregulated in fibroblasts within fetal, but not adult, mesenchymal tissues during healing. Differential homeobox gene expression by fibroblasts may therefore be important in mediating the scarless healing exhibited in early fetal wounds. RNase protection analysis demonstrated that murine Prx-2 expression was involved in fetal but not adult wound healing responses in vitro. Using fibroblasts established from homozygous mutant (Prx-2-/-) and wild-type (Prx-2+/+) murine skin tissues it was demonstrated that Prx-2 affected a number of fetal fibroblastic responses believed to be important in mediating scarless healing in vivo; namely cellular proliferation, extracellular matrix reorganization, and matrix metalloproteinase 2 and hyaluronic acid production. These data demonstrate how Prx-2 may contribute to the regulation of fetal, but not adult, fibroblasts and ultimately the wound healing phenotype. This study provides further evidence for the importance of homeobox transcription factors in the regulation of scarless wound healing. A further understanding of these processes will, it is hoped, enable the targeting of specific therapies in wound healing, both to effect scarless healing and to stimulate healing in chronic, nonhealing wounds such as venous leg ulcers.


Subject(s)
Fetus/physiology , Fibroblasts/physiology , Genes, Homeobox/physiology , Homeodomain Proteins/genetics , Wound Healing/physiology , Animals , Cell Division , Extracellular Matrix/physiology , Gene Expression Regulation, Developmental , Hyaluronic Acid/metabolism , Metalloendopeptidases/analysis , Mice , Tissue Inhibitor of Metalloproteinase-2/analysis
17.
Dev Dyn ; 224(1): 58-68, 2002 May.
Article in English | MEDLINE | ID: mdl-11984874

ABSTRACT

The HOX homeodomain proteins are fundamental regulators of organ and tissue development, where they are thought to function as transcription factors, and HOX gene expression has been associated with numerous types of cancers. Previous studies have demonstrated that enforced expression of the HOXB4 protein transforms cultured fibroblasts and leads to a selective expansion of the hematopoietic stem cell pool, suggesting that this protein might play a role in cellular proliferation. In support of this concept, we now show that enforced expression of HOXB4 in human neonatal keratinocytes results in increased cellular proliferation and colony formation as well as decreased expression of the alpha-2-integrin and CD44 cell surface adhesion molecules. We previously have reported HOXB4 gene expression in the basal and suprabasal layers of developing human skin and now show extensive HOXB4 mRNA in psoriatic skin and basal cell carcinoma. In fetal human skin HOXB4 protein expression was both nuclear and cytoplasmic within epidermal basal cells and in hair follicle inner and outer root sheath cells, whereas strong nuclear signals were observed in the bulge region. In adult skin, HOXB4 protein expression was both nuclear and cytoplasmic, but was predominantly localized to the intermediate and differentiated cell layers. In contrast to the striking gradient patterns of HOX gene and protein expression previously described in developing spinal cord and limb, HOXB4 protein was uniformly detected in all regions of the fetal and adult skin. Although little HOXB4 signal localized to proliferative cell layers, as marked by proliferating cell nuclear antigen (PCNA) staining, in normal adult epidermis, nuclear HOXB4 protein expression substantially overlapped with PCNA-positive cell in a series of samples of hyperproliferative skin. Taken together, these data suggest that nuclear HOXB4 protein may play a role in the regulation of cellular proliferation/adhesion in developing fetal human epidermis and in hyperproliferation conditions, including cancers, in adult epidermis. Published 2002 Wiley-Liss, Inc.


Subject(s)
Epidermis/growth & development , Homeodomain Proteins/metabolism , Keratinocytes/physiology , Skin Diseases/metabolism , Transcription Factors/metabolism , Adult , Antigens, CD/metabolism , Cell Adhesion , Cell Division , Cell Nucleus/metabolism , Epidermal Cells , Epidermis/embryology , Epidermis/metabolism , Genes, Homeobox , Homeodomain Proteins/genetics , Humans , Hyaluronan Receptors/metabolism , In Situ Hybridization , Integrin alpha2 , Keratinocytes/cytology , Proliferating Cell Nuclear Antigen/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...