Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Development ; 136(16): 2825-35, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19633173

RESUMO

The glycosaminoglycan hyaluronan (HA) is a structural component of extracellular matrices and also interacts with cell surface receptors to directly influence cell behavior. To explore functions of HA in limb skeletal development, we conditionally inactivated the gene for HA synthase 2, Has2, in limb bud mesoderm using mice that harbor a floxed allele of Has2 and mice carrying a limb mesoderm-specific Prx1-Cre transgene. The skeletal elements of Has2-deficient limbs are severely shortened, indicating that HA is essential for normal longitudinal growth of all limb skeletal elements. Proximal phalanges are duplicated in Has2 mutant limbs indicating an involvement of HA in patterning specific portions of the digits. The growth plates of Has2-deficient skeletal elements are severely abnormal and disorganized, with a decrease in the deposition of aggrecan in the matrix and a disruption in normal columnar cellular relationships. Furthermore, there is a striking reduction in the number of hypertrophic chondrocytes and in the expression domains of markers of hypertrophic differentiation in the mutant growth plates, indicating that HA is necessary for the normal progression of chondrocyte maturation. In addition, secondary ossification centers do not form in the central regions of Has2 mutant growth plates owing to a failure of hypertrophic differentiation. In addition to skeletal defects, the formation of synovial joint cavities is defective in Has2-deficient limbs. Taken together, our results demonstrate that HA has a crucial role in skeletal growth, patterning, chondrocyte maturation and synovial joint formation in the developing limb.


Assuntos
Desenvolvimento Ósseo/fisiologia , Condrócitos/fisiologia , Extremidades , Inativação Gênica , Glucuronosiltransferase , Ácido Hialurônico/metabolismo , Articulações , Agrecanas/metabolismo , Animais , Padronização Corporal/fisiologia , Proliferação de Células , Condrócitos/citologia , Colágeno Tipo X/genética , Colágeno Tipo X/metabolismo , Matriz Extracelular/química , Matriz Extracelular/metabolismo , Extremidades/embriologia , Extremidades/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Hialuronan Sintases , Ácido Hialurônico/genética , Articulações/anormalidades , Articulações/embriologia , Articulações/crescimento & desenvolvimento , Deformidades Congênitas dos Membros/genética , Mesoderma/fisiologia , Camundongos , Camundongos Knockout
2.
Dev Dyn ; 240(5): 1289-302, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21465622

RESUMO

Sonic hedgehog (Shh) signaling in the limb plays a central role in coordination of limb patterning and outgrowth. Shh expression in the limb is limited to the cells of the zone of polarizing activity (ZPA), located in posterior limb bud mesoderm. Shh is not expressed by limb ectoderm or apical ectodermal ridge (AER), but recent studies suggest a role for AER-Shh signaling in limb patterning. Here, we have examined the effects of activation of Shh signaling in the AER. We find that targeted expression of Shh in the AER activates constitutive Shh signaling throughout the AER and subjacent limb mesoderm, and causes a range of limb patterning defects with progressive severity from mild polydactyly, to polysyndactyly with proximal defects, to severe oligodactyly with phocomelia and partial limb ventralization. Our studies emphasize the importance of control of the timing, level and location of Shh pathway signaling for limb anterior-posterior, proximal-distal, and dorsal-ventral patterning.


Assuntos
Extremidades/embriologia , Proteínas Hedgehog/metabolismo , Botões de Extremidades/metabolismo , Animais , Ectoderma/embriologia , Ectoderma/metabolismo , Proteínas Hedgehog/genética , Botões de Extremidades/embriologia , Camundongos , Camundongos Transgênicos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
3.
J Cell Physiol ; 224(3): 664-71, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20432462

RESUMO

Treatment of common and debilitating degenerative cartilage diseases particularly osteoarthritis is a clinical challenge because of the limited capacity of the tissue for self-repair. Because of their unlimited capacity for self-renewal and ability to differentiate into multiple lineages, human embryonic stem cells (hESCs) are a potentially powerful tool for repair of cartilage defects. The primary objective of the present study was to develop culture systems and conditions that enable hESCs to directly and uniformly differentiate into the chondrogenic lineage without prior embryoid body (EB) formation, since the inherent cellular heterogeneity of EBs hinders obtaining homogeneous populations of chondrogenic cells that can be used for cartilage repair. To this end, we have subjected undifferentiated pluripotent hESCs to the high density micromass culture conditions we have extensively used to direct the differentiation of embryonic limb bud mesenchymal cells into chondrocytes. We report that micromass cultures of pluripotent hESCs undergo direct, rapid, progressive, and substantially uniform chondrogenic differentiation in the presence of BMP2 or a combination of BMP2 and TGF-beta1, signaling molecules that act in concert to regulate chondrogenesis in the developing limb. The gene expression profiles of hESC-derived cultures harvested at various times during the progression of their differentiation has enabled us to identify cultures comprising cells in different phases of the chondrogenic lineage ranging from cultures just entering the lineage to well differentiated chondrocytes. Thus, we are poised to compare the abilities of hESC-derived progenitors in different phases of the chondrogenic lineage for cartilage repair.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula , Condrócitos/fisiologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 2/metabolismo , Técnicas de Cultura de Células , Células Cultivadas , Condrócitos/citologia , Condrogênese/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/fisiologia , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Fator de Crescimento Transformador beta1/metabolismo
4.
Int J Dev Biol ; 50(8): 709-13, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17051482

RESUMO

The onset of elbow joint formation in the developing limb is characterized morphologically by the conversion of differentiated chondrocytes at the site of incipient joint formation into the densely packed flattened cells of the joint interzone. However, experimental studies have indicated that the elbow joint is specified well before joint interzone formation by a distinctive population of precursor cells located at the site in the developing limb bud at which the elbow joint will subsequently form. Here we show that during specification of the elbow joint in the chick limb bud, the homeodomain transcription factors Dlx5 and Dlx6 are highly expressed by a discrete group of cells that encompass the prospective elbow joint. The Dlx5- and Dlx6-expressing cells at the prospective elbow joint are located where the differentiating humerus branches into the radius and ulna. Thus, Dlx5 and Dlx6 are the earliest molecular markers of the presumptive elbow joint yet described. The onset of Dlx5 expression in the region of the presumptive elbow joint is shortly followed by the initiation of expression amongst the Dlx5-expressing cells of Gdf5, which encodes a secreted signaling molecule that is involved in regulating the onset of joint formation. These results suggest that Dlx genes may be involved in specification of the elbow joint and/or in providing positional information that specifies the site at which the elbow joint will form.


Assuntos
Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Articulações/embriologia , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Osso e Ossos/metabolismo , Embrião de Galinha , Condrócitos/metabolismo , Marcadores Genéticos , Fator 5 de Diferenciação de Crescimento , Hibridização In Situ , Botões de Extremidades/embriologia , Fatores de Tempo
5.
Matrix Biol ; 25(1): 27-39, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16226436

RESUMO

Bone morphogenetic proteins (BMPs) are involved in multiple aspects of limb development including regulation of cartilage differentiation. Several BMPs bind strongly to heparin, and heparan sulfate proteoglycans (HSPGs) at the cell surface or in the extracellular matrix have recently been implicated as modulators of BMP signaling in some developing systems. Here we have explored the role of HSPGs in regulating BMP activity during limb chondrogenesis by evaluating the effects of exogenous heparan sulfate (HS), heparitinase treatment, and overexpression of the HSPG syndecan-3 on the ability of BMP2 to modulate the chondrogenic differentiation of limb mesenchymal cells in micromass culture. Exogenous HS dramatically enhances the ability of BMP2 to stimulate chondrogenesis and cartilage specific gene expression, and reduces the concentration of BMP2 needed to stimulate chondrogenesis. Furthermore, HS stimulates BMP2-mediated phosphorylation of Smad1, Smad5, and Smad8, transcriptional mediators of BMP2 signaling, indicating that HS enhances the interaction of BMP2 with its receptors. Pretreatment of micromass cultures with heparitinase to degrade endogenous HSPGs also enhances the chondrogenic activity of BMP2, and reduces the concentration of BMP2 needed to promote chondrogenesis. Taken together these results indicate that exogenous HS or heparitinase enhance the chondrogenic activity of BMP2 by interfering with its interaction with endogenous HSPGs that would normally restrict its interaction with its receptors. Consistent with the possibility that HSPGs are negative modulators of BMP signaling during chondrogenesis, we have found that overexpression of syndecan-3, which is one of the major HSPGs normally expressed during chondrogenesis, greatly impairs the ability of BMP2 to promote cartilage differentiation. Furthermore, retroviral overexpression of syndecan-3 inhibits BMP2-mediated Smad phosphorylation in the regions of the cultures in which chondrogenesis is inhibited and in which ectopic syndecan-3 protein is highly expressed. These results indicate that syndecan-3 interferes with the interaction of BMP2 with its receptors, and that this interference results in an inhibition of chondrogenesis. Taken together these results indicate that HSPGs including syndecan-3 normally modulate the strength of BMP signaling during limb cartilage differentiation by limiting the effective concentration of BMP available for signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Cartilagem/fisiologia , Diferenciação Celular/fisiologia , Condrogênese/fisiologia , Extremidades/crescimento & desenvolvimento , Proteoglicanas de Heparan Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteoglicanas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 2 , Células Cultivadas , Embrião de Galinha , Condrócitos/citologia , Condrócitos/fisiologia , Extremidades/anatomia & histologia , Heparitina Sulfato/metabolismo , Mesoderma/citologia , Mesoderma/fisiologia , Proteínas Smad/metabolismo , Sindecana-3
6.
Int J Dev Biol ; 46(7): 957-61, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12455634

RESUMO

Adrenomedullin (AM) is a multifunctional peptide that exhibits discrete domains of expression during mouse embryogenesis consistent with a role in regulating growth and differentiation during morphogenesis. Here we report that AM immunoreactivity is present at high levels throughout the apical ectodermal ridge (AER) of the chick limb bud as the AER is directing the outgrowth and patterning of underlying limb mesoderm. Immunostaining is particularly strong along the surfaces of the contiguous cells of the AER. AM immunoreactivity attenuates as the AER regresses and is absent from the distal apical ectoderm of stage 20 limbless mutant limb buds which fail to develop an AER. To explore the possible role of AM in AER activity, we examined the effect of exogenous AM and an AM inhibitor on the in vitro morphogenesis of limb mesoderm, cultured in the presence and absence of the AER. Although exogenous AM cannot substitute for the AER in promoting outgrowth of limb mesoderm in vitro, a specific AM antagonist, AM(22-52), impairs the outgrowth and proliferation of limb mesoderm cultured in the presence of the AER. This is consistent with the possibility that inhibition of endogenous AM activity in the AER impairs the ability of the AER to promote limb morphogenesis. Taken together, these studies suggest that an AM-like molecule may function in an autocrine fashion to regulate some aspect of AER activity.


Assuntos
Embrião de Galinha/metabolismo , Extremidades/embriologia , Peptídeos/metabolismo , Adrenomedulina , Animais , Imuno-Histoquímica , Botões de Extremidades , Mutação , Peptídeos/genética , Peptídeos/imunologia
7.
J Bone Miner Res ; 17(6): 1008-14, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12054155

RESUMO

Our laboratory and others have shown that a homeodomain protein binding site plays an important role in transcription of the Collal gene in osteoblasts. This suggests that homeodomain proteins have an important role in osteoblast differentiation. We have investigated the role of Dlx5 in osteoblastic differentiation. In situ hybridization studies indicated that Dlx5 is expressed in chick calvarial osteoblasts (cCOB) in vivo. Northern blot analysis indicated that Dlx5 expression in cultured cCOBs is induced concurrently with osteoblastic markers. To study the effect of overexpression of Dlx5 on osteoblast differentiation, we infected primary osteoblast cultures from 15-day-old embryonal chicken calvaria with replication competent retroviral vectors [RCASBP(A)] expressing Dlx5 or control replication competent avian splice acceptor brianhightiter polymerase subtype A [RCASBP(A)]. Expression of Collal, osteopontin, alkaline phosphatase, and osteocalcin messenger RNA (mRNA) occurred sooner and at higher levels in cultures infected with RCASBP(A)DLX5 than in RCASBP(A)-infected cultures. Mineralization of Dlx5-expressing cultures was evident by days 12-14, and RCAS-infected control osteoblasts did not begin to mineralize until day 17. Dlx5 also stimulated osteoblastic differentiation of calvarial cells that do not normally undergo osteoblastic differentiation in vitro. Our results suggest that Dlx5 plays an important role in inducing calvarial osteoblast differentiation.


Assuntos
Diferenciação Celular/genética , Proteínas de Homeodomínio/genética , Osteoblastos/citologia , Crânio/metabolismo , Animais , Northern Blotting , Calcificação Fisiológica , Embrião de Galinha , Colágeno Tipo I/genética , Vetores Genéticos , Osteopontina , RNA Mensageiro/genética , Retroviridae/genética , Sialoglicoproteínas/genética , Crânio/citologia
8.
Dev Biol ; 305(2): 411-20, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17362908

RESUMO

Hyaluronan (HA) is a large glycosaminoglycan that is not only a structural component of extracellular matrices, but also interacts with cell surface receptors to promote cell proliferation, migration, and intracellular signaling. HA is a major component of the extracellular matrix of the distal subapical mesenchymal cells of the developing limb bud that are undergoing proliferation, directed migration, and patterning in response to the apical ectodermal ridge (AER), and has the functional potential to be involved in these processes. Here we show that the HA synthase Has2 is abundantly expressed by the distal subridge mesodermal cells of the chick limb bud and also by the AER itself. Has2 expression and HA production are downregulated in the proximal central core of the limb bud during the formation of the precartilage condensations of the skeletal elements, suggesting that downregulation of HA may be necessary for the close juxtaposition of cells and the resulting cell-cell interactions that trigger cartilage differentiation during condensation. Overexpression of Has2 in the mesoderm of the chick limb bud in vivo results in the formation of shortened and severely malformed limbs that lack one or more skeletal elements. Skeletal elements that do form in limbs overexpressing Has2 are reduced in length, exhibit abnormal morphology, and are positioned inappropriately. We also demonstrate that sustained HA production in micromass cultures of limb mesenchymal cells inhibits formation of precartilage condensations and subsequent chondrogenesis, indicating that downregulation of HA is indeed necessary for formation of the precartilage condensations that trigger cartilage differentiation. Taken together these results suggest involvement of HA in various aspects of limb morphogenesis.


Assuntos
Proteínas Aviárias/biossíntese , Glucuronosiltransferase/biossíntese , Ácido Hialurônico/biossíntese , Ácido Hialurônico/fisiologia , Asas de Animais/embriologia , Animais , Proteínas Aviárias/antagonistas & inibidores , Proteínas Aviárias/genética , Proteínas Aviárias/fisiologia , Cartilagem/citologia , Cartilagem/embriologia , Cartilagem/enzimologia , Diferenciação Celular/genética , Células Cultivadas , Embrião de Galinha , Regulação para Baixo/genética , Ectoderma/citologia , Ectoderma/enzimologia , Ectoderma/metabolismo , Glucuronosiltransferase/antagonistas & inibidores , Glucuronosiltransferase/genética , Glucuronosiltransferase/fisiologia , Hialuronan Sintases , Ácido Hialurônico/antagonistas & inibidores , Botões de Extremidades , Mesoderma/citologia , Mesoderma/enzimologia , Mesoderma/metabolismo , Asas de Animais/enzimologia , Asas de Animais/metabolismo
9.
Dev Growth Differ ; 49(6): 515-21, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17555518

RESUMO

The homeodomain transcription factor Dlx5 has been implicated in the regulation of chondrocyte and osteoblast differentiation during endochondral ossification in the developing limb. In a gain-of-function approach to directly investigate the role of Dlx5 in chondrocyte maturation, we have used cartilage-specific Col2a1-Dlx5 promoter/enhancer constructs to target overexpression of Dlx5 to the differentiating cartilage models of the limbs of transgenic mice. Targeted overexpression of Dlx5 in cartilage rudiments results in the formation of shortened skeletal elements containing excessive numbers of hypertrophic chondrocytes and expanded domains of expression of Ihh and type X collagen, molecular markers of hypertrophic maturation. This suggests that hypertrophic differentiation is enhanced in response to Dlx5 misexpression. Skeletal elements overexpressing Dlx5 also exhibit a marked reduction in the zone of proliferation, indicating that overexpression of Dlx5 reduces chondrocyte proliferation concomitant with promoting hypertrophic maturation. Taken together these results indicate that Dlx5 is a positive regulator of chondrocyte maturation during endochondral ossification, and suggest that it regulates the process at least in part by promoting the conversion of immature proliferating chondrocytes into hypertrophying chondrocytes; a critical step in the maturation process.


Assuntos
Desenvolvimento Ósseo/fisiologia , Diferenciação Celular/fisiologia , Condrócitos/citologia , Extremidades/embriologia , Proteínas de Homeodomínio/fisiologia , Animais , Desenvolvimento Ósseo/genética , Diferenciação Celular/genética , Condrócitos/patologia , Colágeno Tipo II/genética , Extremidades/anatomia & histologia , Proteínas de Homeodomínio/genética , Hipertrofia , Deformidades Congênitas dos Membros/embriologia , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
10.
Dev Biol ; 252(2): 257-70, 2002 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-12482714

RESUMO

The process of endochondral ossification in which the bones of the limb are formed after generation of cartilage models is dependent on a precisely regulated program of chondrocyte maturation. Here, we show that the homeobox-containing gene Dlx5 is expressed at the onset of chondrocyte maturation during the conversion of immature proliferating chondrocytes into postmitotic hypertrophying chondrocytes, a critical step in the maturation process. Moreover, retroviral misexpression of Dlx5 during differentiation of the skeletal elements of the chick limb in vivo results in the formation of severely shortened skeletal elements that contain excessive numbers of hypertrophying chondrocytes which extend into ectopic regions, including sites normally occupied by immature chondrocytes. The expansion in the extent of hypertrophic maturation detectable histologically is accompanied by expanded and upregulated domains of expression of molecular markers of chondrocyte maturation, particularly type X collagen and osteopontin, and by expansion of mineralized cartilage matrix, which is characteristic of terminal hypertrophic differentiation. Furthermore, Dlx5 misexpression markedly reduces chondrocyte proliferation concomitant with promoting hypertrophic maturation. Taken together, these results indicate that Dlx5 is a positive regulator of chondrocyte maturation and suggest that it regulates the process at least in part by promoting conversion of immature proliferating chondrocytes into hypertrophying chondrocytes. Retroviral misexpression of Dlx5 also enhances formation of periosteal bone, which is derived from the Dlx5-expressing perichondrium that surrounds the diaphyses of the cartilage models. This suggests that Dlx5 may be involved in regulating osteoblast differentiation, as well as chondrocyte maturation, during endochondral ossification.


Assuntos
Desenvolvimento Ósseo/fisiologia , Diferenciação Celular/fisiologia , Condrócitos/citologia , Proteínas de Homeodomínio/fisiologia , Animais , Diferenciação Celular/genética , Divisão Celular , Embrião de Galinha , Proteínas de Homeodomínio/genética
11.
Dev Biol ; 243(1): 44-54, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11846476

RESUMO

Joint formation, the onset of which is characterized by the segmentation of continuous skeletal rudiments into two or more separate elements, is a fundamental aspect of limb pattern formation, playing a critical role in determining the size, shape, and number of individual skeletal elements. Joint formation is initiated by conversion of differentiated chondrocytes at sites of presumptive joints into densely packed nonchondrogenic cells of the joint interzone. This conversion is accompanied by loss of Alcian blue-staining cartilage matrix and downregulation of cartilage-specific gene expression. Here, we report that Cux1, which encodes a transcription factor containing a homeodomain and other DNA-binding motifs, is highly expressed at all of the discrete sites of incipient joint formation in the developing limb concomitant with conversion of differentiated chondrocytes into interzone tissue. Moreover, differentiated limb chondrocytes in micromass cultures infected with a Cux1 retroviral expression vector are converted into nonchondrogenic cells which exhibit loss of Alcian blue cartilage matrix and downregulation of cartilage-specific gene expression as occurs at the onset of normal joint formation. These results suggest that Cux1 is involved in regulating the onset of joint formation by facilitating conversion of chondrocytes into nonchondrogenic cells of the interzone.


Assuntos
Extremidades/embriologia , Articulações/embriologia , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Animais , Embrião de Galinha , Embrião não Mamífero/fisiologia , Extremidades/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/fisiologia , Articulações/fisiologia , Morfogênese , Transdução de Sinais
12.
Dev Biol ; 269(1): 109-22, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15081361

RESUMO

Several bone morphogenetic proteins (BMPs) are expressed in the apical ectodermal ridge (AER), a critical signaling center that directs the outgrowth and patterning of limb mesoderm, but little is known about their function. To study the functions of apical ectodermal BMPs, an AER-specific promoter element from the Msx2 gene was used to target expression of the potent BMP antagonist noggin to the apical ectoderm of the limbs of transgenic mice. Msx2-noggin mutant mice have severely malformed limbs characterized by syndactyly, postaxial polydactyly, and dorsal transformations of ventral structures indicated by absence of ventral footpads and presence of supernumerary ventral nails. Mutant limb buds exhibit a dorsoventral (DV) and anteroposterior (AP) expansion in the extent of the AER. AER activity persists longer than normal and is maintained in regions of the apical ectoderm where its activity normally ceases. Mutant limbs possess a broad band of mesodermal tissue along the distal periphery that is absent from normal limbs and which fails to undergo the apoptosis that normally occurs in the subectodermal mesoderm. Taken together, our results suggest that apical ectodermal BMPs may delimit the boundaries of the AER by preventing adjacent nonridge ectodermal cells from becoming AER cells; negatively modulate AER activity and thus fine-tune the strength of AER signaling; and regulate the apoptosis of the distal subectodermal mesoderm that occurs as AER activity attenuates, an event that is essential for normal limb development. Our results also confirm that ectodermal BMP signaling regulates DV patterning.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Ectoderma/fisiologia , Extremidades/embriologia , Animais , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas de Transporte , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Extremidades/fisiologia , Proteínas de Homeodomínio , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/metabolismo , Camundongos , Mutação , Proteínas/genética , Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA