Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Mol Cell Biol ; 21(21): 7509-22, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11585930

RESUMO

Despite the identification of PBC proteins as cofactors that provide DNA affinity and binding specificity for the HOX homeodomain proteins, HOX proteins do not demonstrate robust activity in transient-transcription assays and few authentic downstream targets have been identified for these putative transcription factors. During a search for additional cofactors, we established that each of the 14 HOX proteins tested, from 11 separate paralog groups, binds to CBP or p300. All six isolated homeodomain fragments tested bind to CBP, suggesting that the homeodomain is a common site of interaction. Surprisingly, CBP-p300 does not form DNA binding complexes with the HOX proteins but instead prevents their binding to DNA. The HOX proteins are not substrates for CBP histone acetyltransferase (HAT) but instead inhibit the activity of CBP in both in vitro and in vivo systems. These mutually inhibitory interactions are reflected by the inability of CBP to potentiate the low levels of gene activation induced by HOX proteins in a range of reporter assays. We propose two models for HOX protein function: (i) HOX proteins may function without CBP HAT to regulate transcription as cooperative DNA binding molecules with PBX, MEIS, or other cofactors, and (ii) the HOX proteins may inhibit CBP HAT activity and thus function as repressors of gene transcription.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteínas de Homeodomínio/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Transativadores/antagonistas & inibidores , Motivos de Aminoácidos , Animais , Glutationa Transferase/metabolismo , Luciferases/metabolismo , Modelos Biológicos , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Transcrição Gênica , Transfecção
3.
Dev Dyn ; 218(4): 636-47, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10906782

RESUMO

HOX homeodomain proteins are master developmental regulators, which are now thought to function as transcription factors by forming cooperative DNA binding complexes with PBX or other protein partners. Although PBX proteins exhibit regulated subcellular localization and function in the nucleus in other tissues, little data exists on HOX and PBX protein localization during skin development. We now show that the HOXB6 protein is expressed in the suprabasal layer of the early developing epidermis and throughout the upper layers of late fetal and adult human skin. HOXB6 signal is cytoplasmic throughout fetal epidermal development, but substantially nuclear in normal adult skin. HOXB6 protein is also partially nuclear in hyperproliferative skin conditions, but appears to be cytoplasmic in basal and squamous cell carcinomas. Although all three PBX genes are expressed in fetal epidermis, none of the three PBX proteins exhibit nuclear co-localization with HOXB6 in either fetal or adult epidermis. RNA and protein data suggest that a truncated HOXB6 protein, lacking the homeodomain, is expressed in undifferentiated keratinocytes and that the full-length protein is induced by differentiation. GFP-fusion proteins were used to demonstrate that the full-length HOXB6 protein is localized to the nucleus while the truncated protein is largely cytoplasmic. Taken together, these data suggest that during epidermal development the truncated HOXB6 isoform may function by a mechanism other than as DNA binding protein, and that most of the nuclear, homeodomain-containing HOXB6 protein does not utilize PBX proteins as DNA binding partners in the skin. Published 2000 Wiley-Liss, Inc.


Assuntos
Células Epidérmicas , Epiderme/embriologia , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/química , Adulto , Cálcio/farmacologia , Diferenciação Celular , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Proteínas de Ligação a DNA/biossíntese , Relação Dose-Resposta a Droga , Humanos , Imuno-Histoquímica , Hibridização In Situ , Queratinócitos/citologia , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/biossíntese , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transcrição Gênica , Transfecção
4.
J Biol Chem ; 275(34): 26172-7, 2000 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-10825160

RESUMO

PBX1 is a homeodomain protein that functions in complexes with other homeodomain-containing proteins to regulate gene expression during developmental and/or differentiation processes. A yeast two-hybrid screen of a fetal liver-hematopoietic cDNA library using PBX1a as bait led to the discovery of a novel non-homeodomain-containing protein that interacts with PBX1 as well as PBX2 and PBX3. RNA analysis revealed it to be expressed in CD34(+) hematopoietic cell populations enriched in primitive progenitors, as is PBX1; search of the expressed sequence tag data base indicated that it is also expressed in other early embryonic as well as adult tissues. The full-length cDNA encodes a 731-amino acid protein that has no significant homology to known proteins. This protein that we have termed hematopoietic PBX-interacting protein (HPIP) is mainly localized in the cytosol and in small amounts in the nucleus. The region of PBX that interacts with HPIP includes both the homeodomain and immediate N-terminal flanking sequences. Strikingly, electrophoretic mobility shift assays revealed that HPIP inhibits the ability of PBX-HOX heterodimers to bind to target sequences. Moreover, HPIP strongly inhibits the transactivation activity of E2A-PBX. Together these findings suggest that HPIP is a new regulator of PBX function.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Transformação Celular Neoplásica , Clonagem Molecular , Proteínas Correpressoras , Hematopoese , Camundongos , Dados de Sequência Molecular , Fator de Transcrição 1 de Leucemia de Células Pré-B , Fatores de Transcrição/química , Ativação Transcricional/efeitos dos fármacos , Transfecção
5.
Leukemia ; 13(12): 1993-9, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10602420

RESUMO

There is increasing evidence that HOX homeobox genes play a role in leukemogenesis. Recent studies have demonstrated that enforced co-expression of HOXA9 and MEIS1 in murine marrow leads to rapid development of myeloid leukemia, and that these proteins exhibit cooperative DNA binding. However, it is unclear whether co-activation of HOXA9 and MEIS genes is a common occurrence in human leukemias. We surveyed expression of HOXA9 and MEIS1 in 24 leukemic cell lines and 80 patient samples, using RNase protection analyses and immunohistochemistry. We demonstrate that the expression of HOXA9 and MEIS1 in leukemia cells is uniquely myeloid, and that these genes are commonly co-expressed in myeloid cell lines and in samples of acute myelogenous leukemia (AML) of all subtypes except in promyelocytic leukemia. While HOXA9 is expressed in most cases of chronic myelogenous leukemia, MEIS1 is weakly expressed or not at all. Immunohistochemical staining of selected AML samples showed moderate to high levels of HOXA9 protein, primarily cytoplasmic, in leukemic myeloblasts, with weaker and primarily nuclear staining for MEIS1. These data support the concept that co-activation of HOXA9 and MEIS1 is a common event in AML, and may represent a common pathway of many different oncogenic mutations.


Assuntos
Genes Homeobox , Proteínas de Homeodomínio/genética , Leucemia Mieloide/genética , Proteínas de Neoplasias/genética , Proteínas de Homeodomínio/análise , Humanos , Imuno-Histoquímica , Proteína Meis1 , Proteínas de Neoplasias/análise , Células Tumorais Cultivadas
6.
J Invest Dermatol ; 113(5): 796-801, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10571736

RESUMO

Terminal differentiation of epidermal keratinocytes is linked to transmembrane ion flux. Previously, we have shown that amiloride, an inhibitor of epithelial sodium channels, blocks synthesis of differentiation-specific proteins in normal human keratinocytes. Here, we have identified the specific subunits of amiloride-sensitive human epithelial sodium channels in relation to differentiation of cultured human keratinocytes, as well as to epidermal development. As assessed by northern hybridization, RNase protection assay, and reverse transcription-polymerase chain reaction, transcripts encoding functional alpha and regulatory beta subunits of human epithelial sodium channels were expressed both in cultured keratinocytes and in epidermis at levels comparable with the kidney. The mRNA expression of both human epithelial sodium channel-alpha and -beta increased during calcium-induced keratinocyte differentiation. Whereas the beta subunit of human epithelial sodium channel was induced by elevated concentrations of calcium, the alpha subunit increased with duration of culture. The regulatory gamma subunit was less abundant but also expressed in epidermis. Both human epithelial sodium channel-alpha and -beta were localized throughout the nucleated layers of human adult epidermis, but these channels were not detected in early stages of fetal epidermal development. This co-ordinated expression of subunits suggests that epithelial sodium channels may play an important part in both epidermal differentiation and skin development, presumably by modulating ion transport required for epidermal terminal differentiation.


Assuntos
Células Epiteliais/química , Queratinócitos/citologia , Canais de Sódio/fisiologia , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Humanos , Hibridização In Situ , Recém-Nascido , Masculino , Pele/crescimento & desenvolvimento , Regulação para Cima
7.
Mol Cell Biol ; 19(4): 3051-61, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10082572

RESUMO

Aberrant activation of the HOX, MEIS, and PBX homeodomain protein families is associated with leukemias, and retrovirally driven coexpression of HOXA9 and MEIS1 is sufficient to induce myeloid leukemia in mice. Previous studies have demonstrated that HOX-9 and HOX-10 paralog proteins are unique among HOX homeodomain proteins in their capacity to form in vitro cooperative DNA binding complexes with either the PBX or MEIS protein. Furthermore, PBX and MEIS proteins have been shown to form in vivo heterodimeric DNA binding complexes with each other. We now show that in vitro DNA site selection for MEIS1 in the presence of HOXA9 and PBX yields a consensus PBX-HOXA9 site. MEIS1 enhances in vitro HOXA9-PBX protein complex formation in the absence of DNA and forms a trimeric electrophoretic mobility shift assay (EMSA) complex with these proteins on an oligonucleotide containing a PBX-HOXA9 site. Myeloid cell nuclear extracts produce EMSA complexes which appear to contain HOXA9, PBX2, and MEIS1, while immunoprecipitation of HOXA9 from these extracts results in coprecipitation of PBX2 and MEIS1. In myeloid cells, HOXA9, MEIS1, and PBX2 are all strongly expressed in the nucleus, where a portion of their signals are colocalized within nuclear speckles. However, cotransfection of HOXA9 and PBX2 with or without MEIS1 minimally influences transcription of a reporter gene containing multiple PBX-HOXA9 binding sites. Taken together, these data suggest that in myeloid leukemia cells MEIS1 forms trimeric complexes with PBX and HOXA9, which in turn can bind to consensus PBX-HOXA9 DNA targets.


Assuntos
Células da Medula Óssea/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Sítios de Ligação , Núcleo Celular/metabolismo , Núcleo Celular/ultraestrutura , Dimerização , Humanos , Leucemia Mieloide/patologia , Camundongos , Proteína Meis1 , Testes de Precipitina , Ligação Proteica , Frações Subcelulares/metabolismo , Fatores de Transcrição , Transcrição Gênica , Células U937
8.
Oncogene ; 16(26): 3403-12, 1998 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-9692548

RESUMO

The products of PBX homeobox genes, which were initially discovered in reciprocal translocations occurring in human leukemias, have been shown to cooperate in the in vitro DNA binding with HOX proteins. Despite the growing body of data implicating Hox genes in the development of various cancers, little is known about the role of HOX-PBX interactions in the regulation of proliferation and induction of transformation of mammalian cells. We build on the existing model of Hox-induced transformation of Rat-1 cells to show that both cellular transformation and proliferation induced by Hoxb4 and Hoxb3 are greatly modulated by the levels of available PBX1 present in these cells. Furthermore, we show that the transforming capacity of these two HOX proteins depends on their conserved tetrapeptide and homeodomain regions which mediate binding to PBX and DNA, respectively. Taken together, results of this study demonstrate that cooperation between HOX and PBX proteins modulates cellular proliferation and strongly suggest that cooperative DNA binding by these two groups of proteins represent the basis for Hox-induced cellular transformation.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Xenopus , Sequência de Aminoácidos , Animais , Testes de Carcinogenicidade , Divisão Celular , Sequência Conservada , Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Neoplasias Experimentais , Fator de Transcrição 1 de Leucemia de Células Pré-B , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Ratos , Proteínas Recombinantes/metabolismo , Fatores de Transcrição/genética
9.
Blood ; 92(2): 383-93, 1998 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-9657735

RESUMO

Hox homeobox genes play a crucial role in specifying the embryonic body pattern. However, a role for Hox genes in T-cell development has not been explored. The Hoxa-9 gene is expressed in normal adult and fetal thymuses. Fetal thymuses of mice homozygous for an interruption of the Hoxa-9 gene are one eighth normal size and have a 25-fold decrease in the number of primitive thymocytes expressing the interleukin-2 receptor (IL-2R, CD25). Progression to the double positive (CD4+CD8+) stage is dramatically retarded in fetal thymic organ cultures. This aberrant development is associated with decreased amounts of intracellular CD3 and T-cell receptor beta (TCRbeta) and reduced surface expression of IL-7R and E-cadherin. Mutant thymocytes show a significant increase in apoptotic cell death and premature downregulation of bcl-2 expression. A similar phenotype is seen in primitive thymocytes from adult Hoxa-9-/- mice and from mice transplanted with Hoxa-9-/- marrow. Hoxa-9 appears to play a previously unsuspected role in T-cell ontogeny by modulating cell survival of early thymocytes and by regulating their subsequent differentiation.


Assuntos
Apoptose/genética , Genes Homeobox , Proteínas de Homeodomínio/genética , Linfócitos T/patologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Timo/embriologia , Timo/patologia
10.
J Invest Dermatol ; 111(1): 57-63, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9665387

RESUMO

Scarless healing of cutaneous wounds occurs in humans during the first two trimesters of development, but by birth all wounds are repaired with scar formation. To search for transcriptional regulatory genes that might mediate fetal tissue regeneration, we surveyed homeobox gene expression in proliferating fetal fibroblasts and in wounded and unwounded skin. Two novel human homeobox genes, PRX-2 and HOXB13, were identified that were differentially expressed during fetal versus adult wound healing. Both genes were predominantly expressed in proliferating fetal fibroblasts and developing dermis, and PRX-2 was downregulated in adult skin. In a model of scarless fetal skin regeneration, PRX-2 expression was strongly increased compared with unwounded skin and the signal was localized to the wounded dermis, the site of scarless repair. Conversely, in adult skin weak epidermal PRX-2 expression was observed, mRNA levels were not increased by wounding, and no dermal expression was detected. HOXB13 expression was decreased in wounded fetal tissue relative to unwounded fetal controls or wounded adult skin. Thus both HOXB13 and PRX-2 are expressed in patterns consistent with roles in fetal skin development and cutaneous regeneration.


Assuntos
Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas de Homeodomínio/genética , Pele/metabolismo , Cicatrização , Células Cultivadas , Feminino , Humanos , Pessoa de Meia-Idade , Gravidez
11.
J Invest Dermatol ; 110(2): 110-5, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9457903

RESUMO

The spatial and temporal deployment of HOX homeobox genes along the spinal axis and in limb buds during fetal development is a key program in embryonic pattern formation. Although we have previously reported that several of the HOX homeobox genes are expressed during murine skin development, there is no information about developmental expression of HOX genes in human skin. We have now used reverse transcriptase polymerase chain reaction, in conjunction with a set of degenerate oligonucleotide primers, to identify a subset of HOX genes that are expressed during human fetal skin development. In situ hybridization analyses demonstrated that there were temporal and spatial shifts in expression of these genes. Strong HOXA4 expression was detected in the basal cell layers of 10 wk fetal epidermis and throughout the epidermis and dermis of 17 wk skin, whereas weak signal was present in the granular layer of newborn and adult skin. The expression patterns of HOXA5 and HOXA7 were similar, but their expression was weaker. In situ hybridization analysis also revealed strong HOXC4 and weaker HOXB7 expression throughout fetal development, whereas HOXB4 was expressed at barely detectable levels. Differential HOX gene expression was also observed in developing hair follicles, and sebaceous and sweat glands. None of the HOX genes examined were detected in the adult dermis.


Assuntos
Expressão Gênica/fisiologia , Genes Homeobox/fisiologia , Pele/embriologia , Adulto , Envelhecimento/fisiologia , Desenvolvimento Embrionário e Fetal , Feto/fisiologia , Idade Gestacional , Folículo Piloso/embriologia , Folículo Piloso/fisiologia , Humanos , Recém-Nascido , Reação em Cadeia da Polimerase , Glândulas Sebáceas/embriologia , Glândulas Sebáceas/fisiologia , Glândulas Sudoríparas/embriologia , Glândulas Sudoríparas/fisiologia , Fatores de Tempo , Transcrição Gênica
12.
Plast Reconstr Surg ; 101(1): 12-9, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9427911

RESUMO

Fetal mammals heal skin wounds through the second trimester of development without evidence of scar. We have investigated the role of bone morphogenetic protein 2 (BMP-2), which is a member of the TGF-beta superfamily, in normal skin development and fetal wound healing. We first used RNA in situ hybridization to demonstrate that BMP-2 was expressed at low levels in the developing hair follicles and in the epidermis of normal human fetal skin. We then created an in vivo model to test how exogenous BMP-2 would affect fetal skin development and wound healing. Fifty micrograms of BMP-2 was implanted into the subcutis of five 70-day-old fetal lambs through a full-thickness linear incision. The BMP-2 was placed beneath the right half of the wound, whereas the left half served as an untreated control. In two of the five animals 1 microgram of TGF-beta was placed into the same position in addition to the 50 micrograms of BMP-2. Twenty days later (90 days gestation, term = 140 days) all the fetal wounds were examined for evidence of cellular hyperproliferation and scar formation. BMP-2 induced massive dermal and epidermal growth when compared with controls. This finding was characterized by marked epidermal thickening and keratinization, a dramatic increase in the number of hair follicles, and more than 50 percent thickening of the dermis. The dermal thickening was the result of both increased cellularity and deposition of large irregular collagen bundles. Wounds treated with both BMP-2 and TGF-beta healed also with an adult-like pattern of scar formation. Surprisingly, the wounds with BMP-2 alone healed with an equal pattern of scar, indicating that there was not an additive effect of combining BMP-2 and TGF-beta. We conclude that BMP-2 is a pleomorphic growth factor that induces cellular growth, maturation, and fibroplasia in both the dermis and epidermis. Further analysis of this growth factor in both fetal and adult wound healing may lead to important discoveries regarding the control of scar formation and fibrosis in many adult tissues.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Cicatriz/fisiopatologia , Feto/fisiologia , Pele/crescimento & desenvolvimento , Fator de Crescimento Transformador beta/fisiologia , Cicatrização/fisiologia , Adulto , Animais , Proteína Morfogenética Óssea 2 , Células Cultivadas , Feminino , Fibroblastos , Humanos , Hibridização In Situ , Gravidez , Segundo Trimestre da Gravidez/fisiologia , Ovinos
13.
Differentiation ; 62(1): 33-41, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9373945

RESUMO

In order to identify homeobox genes which may regulate skin development and possibly mediate scarless fetal wound healing we have screened amplified human fetal skin cDNAs by polymerase chain reaction (PCR) using degenerate oligonucleotide primers designed against highly conserved regions within the homeobox. We identified three non-HOX homeobox genes, MSX-1, MSX-2, and MOX-1, which were differentially expressed in fetal and adult human skin. MSX-1 and MSX-2 were detected in the epidermis, hair follicles, and fibroblasts of the developing fetal skin by in situ hybridization. In contrast, MSX-1 and MSX-2 expression in adult skin was confined to epithelially derived structures. Immunohistochemical analysis of these two genes suggested that their respective homeoproteins may be differentially regulated. While Msx-1 was detected in the cell nucleus of both fetal and adult skin; Msx-2 was detected as a diffuse cytoplasmic signal in fetal epidermis and portions of the hair follicle and dermis, but was localized to the nucleus in adult epidermis. MOX-1 was expressed in a pattern similar to MSX early in gestation but then was restricted exclusively to follicular cells in the innermost layer of the outer root sheath by 21 weeks of development. Furthermore, MOX-1 expression was completely absent in adult cutaneous tissue. These data imply that each of these homeobox genes plays a specific role in skin development.


Assuntos
Proteínas de Ligação a DNA/genética , Epiderme/embriologia , Proteínas de Homeodomínio/genética , Pele/embriologia , Fatores de Transcrição , Epiderme/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Humanos , Imuno-Histoquímica , Fator de Transcrição MSX1 , Pessoa de Meia-Idade , Pele/crescimento & desenvolvimento , Fenômenos Fisiológicos da Pele
14.
J Cell Physiol ; 173(2): 168-77, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9365517

RESUMO

The burgeoning number of articles concerning the role of HOX genes and hematopoiesis ensures that this will continue to be an area of very active research. It seems clear that HOX genes are expressed in stage- and lineage-specific patterns during early stages of hematopoietic development and differentiation. Several lines of evidence suggest that multiple genes of the HOXB (B2, B4, B6-B9), HOXC (C6, C8), and HOXA (A5) are involved in erythropoiesis. Similarly, a number of genes of the HOXA, HOXB, and HOXC appear to play a role in lymphoid cells. Furthermore, several genes, such as A9, A10, B3, B7, and B8, may control myelomonocytic differentiation. The question arises as to whether such a multiplicity of HOX genes reflects redundancy or indicates subtlety of the regulatory machinary. A similar complexity has been observed for hematopoietic cytokines, and the current view is that, although multiple molecules may have similar or overlapping effects, each factor has a specific function and regulatory combinations appear to play a critical role in controlling hematopoietic cell processes (99). One challenge for the future is to delineate in more detail the precise expression patterns of these genes in the many distinct subpopulations of blood cells and during fetal development. Overexpression of HOX genes in hematopoietic cells can dramatically perturb the differentiation of various cell lineages and can contribute to leukemogenesis. Future studies may involve the overexpression of alternatively spliced versions of different HOX genes or of truncated versions of HOX genes to ascertain the functional domains of the proteins that mediate the biologic effects. The findings in HOX knockout mice confirm a role for these genes in normal blood cell development. Further work in this area will require careful examination of fetal hematopoiesis and of animals bearing multiple HOX gene knockouts. Involvement of HOX genes in leukemia is just beginning to be appreciated. Establishing the true extent of HOX gene mutations in human disease will require strategies such as comparative genomic hybridization (100) and analysis of high density oligonucleotide arrays (101). The holy grail of homeobox work is to discover the physiologic processes and specific target genes regulated by HOX proteins. Given the broad range of tissues in which HOX genes are expressed, they would appear to be involved in very basic cellular processes, e.g., cell proliferation and death, adhesion, and migration, etc., rather than the direct regulation of tissue-specific genes. The search for target genes may be made easier by the further characterization of cooperative DNA binding between HOX proteins and other transcription factors. We speculate that HOX proteins do not behave as conventional transcriptional activators or inhibitors but rather may mark genes for potential future activation, i.e., they may establish competency to execute specific differentiation programs, with the actual activation being accomplished by transcriptional pathways triggered by exogenous signals. This proposed function may be an architectural one, involving changes in the conformation of DNA and/or altering interactions between DNA and histones, thus making areas of the genome more or less accessible to other protein factors (102). If this is the case, we may need to develop new assays to discern the molecular action of HOX proteins. The ease of manipulating the hematopoietic systems would appear to make it a very attractive model for explicating the general functions of this remarkable family of genes.


Assuntos
Células Sanguíneas/citologia , Genes Homeobox/fisiologia , Animais , Diferenciação Celular/fisiologia , Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia/genética
15.
Mol Cell Biol ; 17(11): 6448-58, 1997 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9343407

RESUMO

Recent studies show that Hox homeodomain proteins from paralog groups 1 to 10 gain DNA binding specificity and affinity through cooperative binding with the divergent homeodomain protein Pbx1. However, the AbdB-like Hox proteins from paralogs 11, 12, and 13 do not interact with Pbx1a, raising the possibility of different protein partners. The Meis1 homeobox gene has 44% identity to Pbx within the homeodomain and was identified as a common site of viral integration in myeloid leukemias arising in BXH-2 mice. These integrations result in constitutive activation of Meis1. Furthermore, the Hoxa-9 gene is frequently activated by viral integration in the same BXH-2 leukemias, suggesting a biological synergy between these two distinct classes of homeodomain proteins in causing malignant transformation. We now show that the Hoxa-9 protein physically interacts with Meis1 proteins by forming heterodimeric binding complexes on a DNA target containing a Meis1 site (TGACAG) and an AbdB-like Hox site (TTTTACGAC). Hox proteins from the other AbdB-like paralogs, Hoxa-10, Hoxa-11, Hoxd-12, and Hoxb-13, also form DNA binding complexes with Meis1b, while Hox proteins from other paralogs do not appear to interact with Meis1 proteins. DNA binding complexes formed by Meis1 with Hox proteins dissociate much more slowly than DNA complexes with Meis1 alone, suggesting that Hox proteins stabilize the interactions of Meis1 proteins with their DNA targets.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas de Drosophila , Proteínas de Homeodomínio/classificação , Proteínas de Homeodomínio/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Transformação Celular Neoplásica/genética , Leucemia Mieloide/genética , Camundongos , Proteína Meis1 , Ligação Proteica
16.
FEBS Lett ; 414(1): 140-5, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9305748

RESUMO

Alternatively spliced forms of the alpha subunit of the cGMP-gated channel have been cloned from human keratinocytes. One form encodes a complete channel which is almost identical to the rod photoreceptor. A second spliced variant would encode a protein missing a portion of the intracellular hydrophilic domain and the putative first transmembrane domain. Both complete and spliced variants of the channel also were found in epidermis. The expression of the complete form of the channel was induced by levels of extracellular calcium which promote keratinocyte differentiation. The cGMP-gated channel may play an important role in calcium induced keratinocyte differentiation by mediating Ca2+ entry.


Assuntos
Processamento Alternativo , Canais de Cálcio/genética , GMP Cíclico/metabolismo , Ativação do Canal Iônico , Queratinócitos/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Northern Blotting , Cálcio/metabolismo , Canais de Cálcio/química , Canais de Cálcio/fisiologia , Diferenciação Celular , Clonagem Molecular , Humanos , Queratinócitos/citologia , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , RNA Mensageiro/análise , RNA Mensageiro/genética , Alinhamento de Sequência , Análise de Sequência de DNA
17.
Blood ; 89(6): 1922-30, 1997 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9058712

RESUMO

Several homeobox genes of the HOXA and HOXB clusters are expressed in primitive blood cells, suggesting a role for HOX genes in normal hematopoiesis. The HOXA9 gene is expressed in CD34+ marrow cells and in developing lymphocytes. We examined blood-forming organs of mice homozygous for an interrupted HOXA9 allele to determine if loss of HOX gene function is deleterious to hematopoiesis. HOXA9-/- mice have approximately 30% to 40% reductions in total leukocytes and lymphocytes (P < .001) and a blunted granulocytic response to granulocyte colony-stimulating factor (G-CSF). Homozygous mice have significantly smaller spleens and thymuses. Myeloid/erythroid and pre-B progenitors in the marrow are significantly reduced, but no significant decreases are noted in mixed colonies, day 12 colony-forming units-spleen (CFU-S), or long-term culture-initiating cells (LTC-IC), suggesting little or no perturbation in earlier progenitors. Heterozygous animals display no hematopoietic defects. The abnormalities in leukocyte production are transplantable, indicating that the defect resides in the hematopoietic cells. These studies demonstrate a physiologic role for a HOX gene in blood cell differentiation, with the greatest apparent influence of HOXA9 at the level of the committed progenitor.


Assuntos
Medula Óssea/patologia , Células Precursoras Eritroides/patologia , Deleção de Genes , Genes Homeobox , Hematopoese/genética , Linfócitos/patologia , Animais , Linfócitos B/patologia , Medula Óssea/metabolismo , Transplante de Medula Óssea/patologia , Diferenciação Celular/genética , Células Precursoras Eritroides/metabolismo , Feminino , Regulação da Expressão Gênica , Granulócitos , Humanos , Contagem de Linfócitos , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/patologia , Timo/patologia
18.
J Biol Chem ; 272(13): 8198-206, 1997 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-9079637

RESUMO

Previous studies showed that the Hox homeodomain proteins from paralog groups 1-8 display cooperative DNA binding with the non-Hox homeodomain protein Pbx, mediated by a canonical YPWM. Although the Abd-B-like Hox proteins in paralogs 9-13 lack this sequence, Hoxb-9 and Hoxa-10 were reported to bind with Pbx1a to DNA. We show that these interactions require a tryptophan 6 amino acids N-terminal to the homeodomain. Binding site selection for Hoxb-9 with Pbx1a yielded ATGATTTACGAC, containing a novel TTAC Hox-binding site adjacent to a Pbx site. In the presence of Pbx1a, Hoxb-9 and Hoxa-10 bound to targets containing either TTAC or TTAT. These data extend previous findings that interactions with Pbx define a Hox protein binding code for different DNA sequences across paralog groups 1 through 10. Members of the 11, 12, and 13 paralogs do not cooperatively bind DNA with Pbx1a, despite the presence of tryptophan residues N-terminal to the homeodomain in Hoxd-12 and Hoxd-13. Hoxa-11, Hoxd-12, or Hoxd-13, in the presence of Pbx1a, selected a TTAC Hox site but lacking a Pbx1a site. These data suggest that Abd-B-like Hox proteins bind to a novel TTAC site and can be divided by their cooperative binding to DNA with Pbx1a.


Assuntos
Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Proteínas de Drosophila , Proteínas de Homeodomínio/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas de Xenopus , Sequência de Aminoácidos , Sequência de Bases , Proteínas de Ligação a DNA/química , Genes Homeobox , Proteínas Homeobox A10 , Proteínas de Homeodomínio/química , Humanos , Dados de Sequência Molecular , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/química , Relação Estrutura-Atividade , TATA Box , Fatores de Transcrição/metabolismo , Triptofano
19.
Mol Cell Biol ; 17(1): 495-505, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8972230

RESUMO

Multiple members of the A, B, and C clusters of Hox genes are expressed in hematopoietic cells. Several of these Hox genes have been found to display distinctive expression patterns, with genes located at the 3' side of the clusters being expressed at their highest levels in the most primitive subpopulation of human CD34+ bone marrow cells and genes located at the 5' end having a broader range of expression, with downregulation at later stages of hematopoietic differentiation. To explore if these patterns reflect different functional activities, we have retrovirally engineered the overexpression of a 5'-located gene, HOXA10, in murine bone marrow cells and demonstrate effects strikingly different from those induced by overexpression of a 3'-located gene, HOXB4. In contrast to HOXB4, which causes selective expansion of primitive hematopoietic cells without altering their differentiation, overexpression of HOXA10 profoundly perturbed myeloid and B-lymphoid differentiation. The bone marrow of mice reconstituted with HOXA10-transduced bone marrow cells contained in high frequency a unique progenitor cell with megakaryocytic colony-forming ability and was virtually devoid of unilineage macrophage and pre-B-lymphoid progenitor cells derived from the transduced cells. Moreover, and again in contrast to HOXB4, a significant proportion of HOXA10 mice developed a transplantable acute myeloid leukemia with a latency of 19 to 50 weeks. These results thus add to recognition of Hox genes as important regulators of hematopoiesis and provide important new evidence of Hox gene-specific functions that may correlate with their normal expression pattern.


Assuntos
Proteínas de Ligação a DNA/genética , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Homeodomínio , Leucemia Mieloide/genética , Doença Aguda , Animais , Linfócitos B , Células da Medula Óssea , Transplante de Medula Óssea , Diferenciação Celular , Feminino , Técnicas de Transferência de Genes , Genes Homeobox/genética , Proteínas Homeobox A10 , Humanos , Tecido Linfoide/citologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise , Retroviridae/genética
20.
Immunity ; 6(1): 13-22, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9052833

RESUMO

HOXB3 mRNA levels are high in the earliest CD34+ lineage- bone marrow cells and low to undetectable in later CD34+/CD34- cells. To gain some insight into the role this gene may play in hematopoiesis, HOXB3 was overexpressed in murine bone marrow cells using retroviral gene transfer. Thymi of HOXB3 marrow recipients were reduced in size compared with control transplant recipients, with a 24-fold decrease in the absolute number of CD4+ CD8+ cells and a 3-fold increase in the number of CD4- CD8- thymocytes that contained a high proportion of gammadelta TCR+ cells. B cell differentiation was also perturbed in these mice, as indicated by the virtual absence of transduced IL-7-responsive pre-B clonogenic progenitors. Recipients of HOXB3-transduced cells also had elevated numbers of mature granulocyte macrophage colony-forming cells in their bone marrow and spleen. Together these results suggest roles for HOXB3 in proliferation and differentiation processes of both early myeloid and lymphoid developmental pathways.


Assuntos
Linfócitos B/citologia , Genes Homeobox , Granulócitos/citologia , Hematopoese , Proteínas de Homeodomínio/genética , Transtornos Mieloproliferativos/genética , Linfócitos T/citologia , Proteínas de Xenopus , Animais , Antígenos CD34/análise , Células da Medula Óssea , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Baço/citologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...