Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 30
1.
Cancer Cell Int ; 24(1): 72, 2024 Feb 12.
Article En | MEDLINE | ID: mdl-38347567

BACKGROUND: Glioblastoma (GBM) is the most lethal primary brain tumor in adult, characterized by highly aggressive and infiltrative growth. The current therapeutic management of GBM includes surgical resection followed by ionizing radiations and chemotherapy. Complex and dynamic interplay between tumor cells and tumor microenvironment drives the progression and contributes to therapeutic resistance. Extracellular vesicles (EVs) play a crucial role in the intercellular communication by delivering bioactive molecules in the surrounding milieu modulating tumor microenvironment. METHODS: In this study, we isolated by ultracentrifugation EVs from GBM stem-like cell (GSC) lines and human microvascular endothelial cells (HMVECs) exposed or not to ionizing irradiation. After counting and characterization, we evaluated the effects of exposure of GSCs to EVs isolated from endothelial cells and vice versa. The RNA content of EVs isolated from GSC lines and HMVECs exposed or not to ionizing irradiation, was analyzed by RNA-Seq. Periostin (POSTN) and Filamin-B (FLNB) emerged in gene set enrichment analysis as the most interesting transcripts enriched after irradiation in endothelial cell-derived EVs and GSC-derived EVs, respectively. POSTN and FLNB expression was modulated and the effects were analyzed by in vitro assays. RESULTS: We confirmed that ionizing radiations increased EV secretion by GSCs and normal endothelial cells, affected the contents of and response to cellular secreted EVs. Particularly, GSC-derived EVs decreased radiation-induced senescence and promoted migration in HMVECs whereas, endothelial cell-derived EVs promoted tumorigenic properties and endothelial differentiation of GSCs. RNA-Seq analysis of EV content, identified FLNB and POSTN as transcripts enriched in EVs isolated after irradiation from GSCs and HMVECs, respectively. Assays performed on POSTN overexpressing GSCs confirmed the ability of POSTN to mimic the effects of endothelial cell-derived EVs on GSC migration and clonogenic abilities and transdifferentiation potential. Functional assays performed on HMVECs after silencing of FLNB supported its role as mediator of the effects of GSC-derived EVs on senescence and migration. CONCLUSION: In this study, we identified POSTN and FLNB as potential mediators of the effects of EVs on GSC and HMVEC behavior confirming that EVs play a crucial role in the intercellular communication by delivering bioactive molecules in the surrounding milieu modulating tumor microenvironment.

2.
Biology (Basel) ; 12(10)2023 Sep 29.
Article En | MEDLINE | ID: mdl-37887007

Bisphosphonates (BPs) are successfully used to cure a number of diseases characterized by a metabolic reduction in bone density, such as Osteoporosis, or a neoplastic destruction of bone tissue, such as multiple myeloma and bone metastases. These drugs exert their therapeutic effect by causing a systemic osteoclast depletion that, in turn, is responsible for reduced bone resorption. Unfortunately, in addition to their beneficial activity, BPs can also determine a frightening side effect known as osteonecrosis of the jaw (ONJ). It is generally believed that the inability of osteoclasts to dispose of inflamed/necrotic bone represents the main physiopathological aspect of ONJ. In principle, a therapeutic strategy able to elicit a local re-activation of osteoclast production could counteract ONJ and promote the healing of its lesions. Using an experimental model of Vitamin D3-dependent osteoclastogenesis, we have previously demonstrated that Magnesium is a powerful inducer of osteoclast differentiation. Here we show that, surprisingly, this effect is greatly enhanced by the presence of Zoledronate, chosen for our study because it is the most effective and dangerous of the BPs. This finding allows us to hypothesize that Magnesium might play an important role in the topical therapy of ONJ.

3.
Cancer Lett ; 567: 216262, 2023 07 28.
Article En | MEDLINE | ID: mdl-37307894

Aberrant splicing events are associated with colorectal cancer (CRC) and provide new opportunities for tumor diagnosis and treatment. The expression of the splice variants of NF-YA, the DNA binding subunit of the transcription factor NF-Y, is deregulated in multiple cancer types compared to healthy tissues. NF-YAs and NF-YAl isoforms differ in the transactivation domain, which may result in distinct transcriptional programs. In this study, we demonstrated that the NF-YAl transcript is higher in aggressive mesenchymal CRCs and predicts shorter patients' survival. In 2D and 3D conditions, CRC cells overexpressing NF-YAl (NF-YAlhigh) exhibit reduced cell proliferation, rapid single cell amoeboid-like migration, and form irregular spheroids with poor cell-to-cell adhesion. Compared to NF-YAshigh, NF-YAlhigh cells show changes in the transcription of genes involved in epithelial-mesenchymal transition, extracellular matrix and cell adhesion. NF-YAl and NF-YAs bind similarly to the promoter of the E-cadherin gene, but oppositely regulate its transcription. The increased metastatic potential of NF-YAlhigh cells in vivo was confirmed in zebrafish xenografts. These results suggest that the NF-YAl splice variant could be a new CRC prognostic factor and that splice-switching strategies may reduce metastatic CRC progression.


Colonic Neoplasms , Colorectal Neoplasms , Animals , Humans , Zebrafish/genetics , Transcription Factors , Colonic Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix , Sorbitol , Cell Movement/genetics , Colorectal Neoplasms/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
5.
Genes Dev ; 33(5-6): 258-275, 2019 03 01.
Article En | MEDLINE | ID: mdl-30824532

Pbx genes encode transcription factors that belong to the TALE (three-amino-acid loop extension) superclass of homeodomain proteins. We have witnessed a surge in information about the roles of this gene family as leading actors in the transcriptional control of development. PBX proteins represent a clear example of how transcription factors can regulate developmental processes by combinatorial properties, acting within multimeric complexes to implement activation or repression of transcription depending on their interaction partners. Here, we revisit long-emphasized functions of PBX transcription factors as cofactors for HOX proteins, major architects of the body plan. We further discuss new knowledge on roles of PBX proteins in different developmental contexts as upstream regulators of Hox genes-as factors that interact with non-HOX proteins and can work independently of HOX-as well as potential pioneer factors. Committed to building a perfect body, PBX proteins govern regulatory networks that direct essential morphogenetic processes and organogenesis in vertebrate development. Perturbations of PBX-dependent networks can cause human congenital disease and cancer.


Gene Expression Regulation, Developmental , Gene Regulatory Networks/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Organogenesis/genetics , Vertebrates/embryology , Vertebrates/genetics , Animals , Genes, Homeobox/genetics , Humans
7.
J Dev Biol ; 5(2)2017 May 10.
Article En | MEDLINE | ID: mdl-29615563

This Special Issue of Journal of Developmental Biology (JDB) covers an indeed very "special" (at least to me) family of highly evolutionarily conserved genes, the Hox genes.[...].

8.
Oncotarget ; 8(3): 4747-4759, 2017 Jan 17.
Article En | MEDLINE | ID: mdl-27902469

OBJECTIVE: To explore the possible misexpression of the microRNA miR-196b in colorectal cancer (CRC) and its role in controlling the expression of GATA6, a putative target gene crucial to intestinal cell homeostasis and tumorigenesis. DESIGN: The expression of miR-196b was analysed by qRT-PCR in surgical resection samples from a cohort of sporadic colon cancer patients. Manipulations of miR-196b expression were performed to demonstrate its inhibition of GATA6 protein levels. RESULTS: We found that miR-196b is significantly upregulated in pre-treatment surgical resection samples from a cohort of sporadic colon cancer patients. The upregulation of miR-196b correlates with less severe clinicopathological characteristics, such as early tumor stage and absence of lymph node metastases. We show that in CRC cells, miR-196b targets the mRNA of GATA6, a transcription factor involved in the homeostasis and differentiation of intestinal epithelial cells, and a positive regulator of the Wnt/ß-catenin pathway. We moreover found that the increase of miR-196b correlates with a reduced GATA6 protein expression in colon cancer patients. CONCLUSION: Our results establish miR-196b as a post-transcriptional inhibitor of GATA6 in CRC cells, implicating miR-196b function in gene regulatory pathways crucial to intestinal cell homeostasis and tumorigenesis. Our results furthermore suggest a role of miR-196b expression in CRC, as an antagonist of GATA6 function in tumor cells, thus providing the basis for a potential targeting strategy for the treatment of CRC.


Colorectal Neoplasms/pathology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , MicroRNAs/genetics , Up-Regulation , 3' Untranslated Regions , Caco-2 Cells , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lymphatic Metastasis , Male , Neoplasm Staging , Wnt Signaling Pathway
9.
Cell Cycle ; 15(17): 2275-87, 2016 Sep.
Article En | MEDLINE | ID: mdl-27097363

NUP98 is a recurrent partner gene in translocations causing acute myeloid leukemias and myelodisplastic syndrome. The expression of NUP98 fusion oncoproteins has been shown to induce mitotic spindle defects and chromosome missegregation, which correlate with the capability of NUP98 fusions to cause mitotic checkpoint attenuation. We show that NUP98 oncoproteins physically interact with the APC/C(Cdc20) in the absence of the NUP98 partner protein RAE1, and prevent the binding of the mitotic checkpoint complex to the APC/C(Cdc20). NUP98 oncoproteins require the GLEBS-like domain present in their NUP98 moiety to bind the APC/C(Cdc20). We found that NUP98 wild-type is a substrate of APC/C(Cdc20) prior to mitotic entry, and that its binding to APC/C(Cdc20) is controlled via phosphorylation of a PEST sequence located within its C-terminal portion. We identify S606, within the PEST sequence, as a key target site, whose phosphorylation modulates the capability of NUP98 to interact with APC/C(Cdc20). We finally provide evidence for an involvement of the peptidyl-prolyl isomerase PIN1 in modulating the possible conformational changes within NUP98 that lead to its dissociation from the APC/C(Cdc20) during mitosis. Our results provide novel insight into the mechanisms underlying the aberrant capability of NUP98 oncoproteins to interact with APC/C(Cdc20) and to interfere with its function.


Cdc20 Proteins/metabolism , M Phase Cell Cycle Checkpoints , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , HEK293 Cells , HeLa Cells , Humans , Mad2 Proteins , Mitosis , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Nuclear Pore Complex Proteins/chemistry , Protein Binding , Protein Domains , Protein Stability , Substrate Specificity
10.
Biochim Biophys Acta ; 1849(8): 1066-80, 2015 Aug.
Article En | MEDLINE | ID: mdl-26141604

The miR-196 miRNA gene family located within the Hox gene clusters has been shown to function during embryogenesis and to be aberrantly expressed in various malignancies, including leukaemia, melanoma, and colorectal cancer. Despite its involvement in numerous biological processes, the control of miR-196 expression is still poorly defined. We identified the miR-196b promoter and found that the mature miR-196b originates from a large, non-coding primary transcript, which starts within an autonomous TATA box promoter and is not in physical continuity with either the Hoxa10 or Hoxa9 main primary transcripts. A ~680bp genomic fragment, spanning the pri-miR-196b transcription start site, is sufficient to recapitulate the neural tube expression pattern of miR-196 during embryogenesis. This region contains potential binding sites for Cdx and 5'Hox transcription factors. Two of these sites revealed to be necessary for neural tube expression and were bound in vivo by Cdx2 and Hoxd13. We show that Cdx2 is required for miR-196 expression and that both Cdx2 and 5'Hox, but not 3'Hox, are able to activate the miR-196b promoter. The possible role of Cdx2- and 5'Hox-mediated regulation of miR-196 expression in vertebrate anterior-posterior (AP) axis formation during embryogenesis is discussed.


Embryonic Development/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , MicroRNAs/genetics , Promoter Regions, Genetic , Transcription Factors/metabolism , Animals , Base Sequence , Body Patterning/genetics , CDX2 Transcription Factor , Cells, Cultured , Chick Embryo , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Transcription, Genetic
11.
Biochim Biophys Acta ; 1843(5): 955-64, 2014 May.
Article En | MEDLINE | ID: mdl-24472656

In spite of the numerous reports implicating MafB transcription factor in the molecular control of monocyte-macrophage differentiation, the precise genetic program underlying this activity has been, to date, poorly understood. To clarify this issue, we planned a number of experiments that were mainly conducted on human primary macrophages. In this regard, a preliminary gene function study, based on MafB inactivation and over-expression, indicated MMP9 and IL-7R genes as possible targets of the investigated transcription factor. Bioinformatics analysis of their promoter regions disclosed the presence of several putative MARE elements and a combined approach of EMSA and luciferase assay subsequently demonstrated that expression of both genes is indeed activated by MafB through a direct transcription mechanism. Additional investigation, performed with similar procedures to elucidate the biological relevance of our observation, revealed that MafB is a downstream target of the IL-10/STAT3 signaling pathway, normally inducing the macrophage de-activation process. Taken together our data support the existence of a signaling cascade by which stimulation of macrophages with the IL-10 cytokine determines a sequential activation of STAT3 and MafB transcription factors, in turn leading to an up-regulated expression of MMP9 and IL-7R genes.


Interleukin-10/metabolism , Macrophage Activation , MafB Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Base Sequence , Cell Line , DNA Probes , Gene Silencing , Humans , MafB Transcription Factor/genetics , Matrix Metalloproteinase 7/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Receptors, Interleukin-7/genetics
12.
Cancer Res ; 74(4): 1079-90, 2014 Feb 15.
Article En | MEDLINE | ID: mdl-24371226

NUP98 is a recurrent fusion partner in chromosome translocations that cause acute myelogenous leukemia. NUP98, a nucleoporin, and its interaction partner Rae1, have been implicated in the control of chromosome segregation, but their mechanistic contributions to tumorigenesis have been unclear. Here, we show that expression of NUP98 fusion oncoproteins causes mitotic spindle defects and chromosome missegregation, correlating with the capability of NUP98 fusions to cause premature securin degradation and slippage from an unsatisfied spindle assembly checkpoint (SAC). NUP98 fusions, unlike wild-type NUP98, were found to physically interact with the anaphase promoting complex/cyclosome (APC/C)(Cdc20) and to displace the BubR1 SAC component, suggesting a possible mechanistic basis for their interference with SAC function. In addition, NUP98 oncoproteins displayed a prolonged half-life in cells. We found that NUP98 stability is controlled by a PEST sequence, absent in NUP98 oncoproteins, whose deletion reproduced the aberrant SAC-interfering activity of NUP98 oncoproteins. Together, our findings suggest that NUP98 oncoproteins predispose myeloid cells to oncogenic transformation or malignant progression by promoting whole chromosome instability.


Aneuploidy , M Phase Cell Cycle Checkpoints/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/physiology , Cell Transformation, Neoplastic/genetics , Chromosomal Instability/physiology , Chromosome Segregation/genetics , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Mitosis/genetics , Oncogenes/physiology , Proteolysis , Securin/metabolism , Transcription Factors/genetics
13.
Exp Cell Res ; 319(20): 3201-13, 2013 Dec 10.
Article En | MEDLINE | ID: mdl-23973664

Orosomucoid 1 (ORM1), also named Alpha 1 acid glycoprotein A (AGP-A), is an abundant plasma protein characterized by anti-inflammatory and immune-modulating properties. The present study was designed to identify a possible correlation between ORM1 and Vitamin D3 (1,25(OH)2D3), a hormone exerting a widespread effect on cell proliferation, differentiation and regulation of the immune system. In particular, the data described here indicated that ORM1 is a 1,25(OH)2D3 primary response gene, characterized by the presence of a VDRE element inside the 1kb sequence of its proximal promoter region. This finding was demonstrated with gene expression studies, Chromatin Immunoprecipitation and luciferase transactivation experiments and confirmed by VDR full length and dominant negative over-expression. In addition, several experiments carried out in human normal monocytes demonstrated that the 1,25(OH)2D3--VDR--ORM1 pathway plays a functional role inside the macrophage de-activation process and that ORM1 may be considered as a signaling molecule involved in the maintenance of tissue homeostasis and remodeling.


Macrophage Activation/drug effects , Macrophages/drug effects , Orosomucoid/metabolism , Vitamin D/pharmacology , Gene Expression Profiling , HL-60 Cells , Humans , Macrophages/metabolism , Orosomucoid/genetics , Orosomucoid/isolation & purification , U937 Cells
14.
Hum Mol Genet ; 21(11): 2464-75, 2012 Jun 01.
Article En | MEDLINE | ID: mdl-22373878

Synpolydactyly (SPD) is a distal limb anomaly characterized by incomplete digit separation and the presence of supernumerary digits in the syndactylous web. This phenotype has been associated with mutations in the homeodomain or polyalanine tract of the HOXD13 gene. We identified a novel mutation (G11A) in HOXD13 that is located outside the previously known domains and affects the intracellular half life of the protein. Misexpression of HOXD13(G11A) in the developing chick limb phenocopied the human SPD phenotype. Finally, we demonstrated through in vitro studies that this mutation has a destabilizing effect on GLI3R uncovering an unappreciated mechanism by which HOXD13 determines the patterning of the limb.


Body Patterning/genetics , Homeodomain Proteins/genetics , Mutation , Syndactyly/genetics , Transcription Factors/genetics , Animals , COS Cells , Chick Embryo , Chlorocebus aethiops , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Phenotype , Syndactyly/metabolism , Transcription Factors/metabolism , Transfection , Zinc Finger Protein Gli3
15.
Dev Dyn ; 240(5): 1173-89, 2011 May.
Article En | MEDLINE | ID: mdl-21455939

Genes expressed in the somatopleuric mesoderm, the embryonic domain giving rise to the vertebrate pelvis, appear important for pelvic girdle formation. Among such genes, Pbx family members and Emx2 were found to genetically interact in hindlimb and pectoral girdle formation. Here, we generated compound mutant embryos carrying combinations of mutated alleles for Pbx1, Pbx2, and Pbx3, as well as Pbx1 and Emx2, to examine potential genetic interactions during pelvic development. Indeed, Pbx genes share overlapping functions and Pbx1 and Emx2 genetically interact in pelvic formation. We show that, in compound Pbx1;Pbx2 and Pbx1;Emx2 mutants, pelvic mesenchymal condensation is markedly perturbed, indicative of an upstream control by these homeoproteins. We establish that expression of Tbx15, Prrx1, and Pax1, among other genes involved in the specification and development of select pelvic structures, is altered in our compound mutants. Lastly, we identify potential Pbx1-Emx2-regulated enhancers for Tbx15, Prrx1, and Pax1, using bioinformatics analyses.


Pelvis/embryology , Animals , Computational Biology , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , In Situ Hybridization , Male , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pre-B-Cell Leukemia Transcription Factor 1 , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Dev Dyn ; 240(5): 1063-86, 2011 May.
Article En | MEDLINE | ID: mdl-21416555

Limb development has long provided an excellent model for understanding the genetic principles driving embryogenesis. Studies utilizing chick and mouse have led to new insights into limb patterning and morphogenesis. Recent research has centered on the regulatory networks underlying limb development. Here, we discuss the hierarchical, overlapping, and iterative roles of Pbx family members in appendicular development that have emerged from genetic analyses in the mouse. Pbx genes are essential in determining limb bud positioning, early bud formation, limb axes establishment and coordination, and patterning and morphogenesis of most elements of the limb and girdle. Pbx proteins directly regulate critical effectors of limb and girdle development, including morphogen-encoding genes like Shh in limb posterior mesoderm, and transcription factor-encoding genes like Alx1 in pre-scapular domains. Interestingly, at least in limb buds, Pbx appear to act not only as Hox cofactors, but also in the upstream control of 5' HoxA/D gene expression.


Extremities/embryology , Homeodomain Proteins/metabolism , Animals , Extremities/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Mesoderm/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Protein Binding/genetics , Protein Binding/physiology
17.
Development ; 137(15): 2559-69, 2010 Aug 01.
Article En | MEDLINE | ID: mdl-20627960

The genetic pathways underlying shoulder blade development are largely unknown, as gene networks controlling limb morphogenesis have limited influence on scapula formation. Analysis of mouse mutants for Pbx and Emx2 genes has suggested their potential roles in girdle development. In this study, by generating compound mutant mice, we examined the genetic control of scapula development by Pbx genes and their functional relationship with Emx2. Analyses of Pbx and Pbx1;Emx2 compound mutants revealed that Pbx genes share overlapping functions in shoulder development and that Pbx1 genetically interacts with Emx2 in this process. Here, we provide a biochemical basis for Pbx1;Emx2 genetic interaction by showing that Pbx1 and Emx2 can bind specific DNA sequences as heterodimers. Moreover, the expression of genes crucial for scapula development is altered in these mutants, indicating that Pbx genes act upstream of essential pathways for scapula formation. In particular, expression of Alx1, an effector of scapula blade patterning, is absent in all compound mutants. We demonstrate that Pbx1 and Emx2 bind in vivo to a conserved sequence upstream of Alx1 and cooperatively activate its transcription via this potential regulatory element. Our results establish an essential role for Pbx1 in genetic interactions with its family members and with Emx2 and delineate novel regulatory networks in shoulder girdle development.


DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Mutation , Scapula/embryology , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Crosses, Genetic , DNA-Binding Proteins/genetics , Dimerization , Gene Expression Profiling , Homeodomain Proteins/genetics , Mice , Models, Genetic , Molecular Sequence Data , Pre-B-Cell Leukemia Transcription Factor 1 , Scapula/physiology , Sequence Homology, Nucleic Acid , Shoulder/embryology , Transcription Factors/genetics
18.
Mol Cell Biol ; 29(21): 5775-88, 2009 Nov.
Article En | MEDLINE | ID: mdl-19703996

HOX DNA-binding proteins control patterning during development by regulating processes such as cell aggregation and proliferation. Recently, a possible involvement of HOX proteins in replication origin activity was suggested by results showing that a number of HOX proteins interact with the DNA replication licensing regulator geminin and bind a characterized human origin of replication. The functional significance of these observations, however, remained unclear. We show that HOXD13, HOXD11, and HOXA13 bind in vivo all characterized human replication origins tested. We furthermore show that HOXD13 interacts with the CDC6 loading factor, promotes pre-replication complex (pre-RC) proteins assembly at origins, and stimulates DNA synthesis in an in vivo replication assay. HOXD13 expression in cultured cells accelerates DNA synthesis initiation in correlation with the earlier pre-RC recruitment onto origins during G(1) phase. Geminin, which interacts with HOXD13 as well, blocks HOXD13-mediated assembly of pre-RC proteins and inhibits HOXD13-induced DNA replication. Our results uncover a function for Hox proteins in the regulation of replication origin activity and reveal an unforeseen role for the inhibition of HOX protein activity by geminin in the context of replication origin licensing.


Cell Cycle Proteins/metabolism , DNA Replication , Homeodomain Proteins/metabolism , Replication Origin/genetics , Transcription Factors/metabolism , Animals , Cell Line , Cells, Cultured , DNA/biosynthesis , G1 Phase , Geminin , Humans , Mice , Nuclear Proteins/metabolism , Protein Binding
19.
Hum Mol Genet ; 18(5): 847-60, 2009 Mar 01.
Article En | MEDLINE | ID: mdl-19060004

The 5' members of the HoxD gene cluster (paralogous groups 9-13) are crucial for correct vertebrate limb patterning. Mutations in the HOXD13 gene have been found to cause synpolydactyly (SPD) and other limb malformations in human. We report the identification in a Greek family of a variant form of SPD caused by a novel missense mutation that substitutes glycine for valine in position 220 (G220V) of HOXD13. This mutation represents the first substitution of an amino acid located outside of the HOXD13 homeodomain that causes autopodal limb malformations. We have characterized this mutation at the molecular level and found that the G220V substitution causes a significant impairment of the capacity of HOXD13 to bind DNA and regulate transcription. HOXD13(G220V) was found to be deficient in both activating and repressing transcription through HOXD13-responsive regulatory elements. In accordance with these results, a comparison of the activities of HOXD13 and HOXD13(G220V) in vivo, using retrovirus-mediated misexpression in developing chick limbs, showed that the G220V mutation impairs the capacity of HOXD13 to perturb the development of proximal limb skeletal elements and to ectopically activate the transcription of the Hand2 target gene. We moreover show that the G220V mutation compromises the stability of the HOXD13 protein within cells and causes its partial accumulation in the cytosol in the form of subtle aggregates. Taken together, our results establish that the G220V substitution does not produce a dominant-negative effect or a gain-of-function, but represents a dominant loss-of-function mutation revealing haploinsufficiency of HOXD13 in human.


Amino Acid Substitution , Homeodomain Proteins/chemistry , Homeodomain Proteins/genetics , Limb Deformities, Congenital/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Amino Acid Sequence , Animals , COS Cells , Cell Line, Tumor , Chickens , Chlorocebus aethiops , Homeodomain Proteins/metabolism , Humans , Limb Deformities, Congenital/metabolism , Mice , Molecular Sequence Data , Mutation , Mutation, Missense , Pedigree , Phenotype , Protein Binding , Protein Structure, Tertiary , Sequence Alignment , Transcription Factors/metabolism , Transcription, Genetic
20.
Dev Biol ; 317(2): 497-507, 2008 May 15.
Article En | MEDLINE | ID: mdl-18407260

5' HoxD genes are required for the correct formation of limb skeletal elements. Hoxd13, the most 5'-located HoxD gene, is important for patterning the most distal limb region, and its mutation causes human limb malformation syndromes. The mechanisms underlying the control of developmental processes by Hoxd13, and by Hox genes in general, are still elusive, due to the limited knowledge on their direct downstream target genes. We identified by ChIP-on-chip 248 known gene loci bound invivo by Hoxd13. Genes relevant to limb patterning and skeletogenesis were further analysed. We found that Hoxd13 binds invivo, in developing limbs, the loci of Hand2, a gene crucial to limb AP axis patterning, of Meis1 and Meis2, involved in PD patterning, of the Sfrp1, Barx1, and Fbn1 genes, involved in skeletogenesis, and of the Dach1, Bmp2, Bmp4, andEmx2 genes. We show that Hoxd13 misexpression in developing chick limbs alters the expression of the majority of these genes, supporting the conclusion that Hoxd13 directly regulates their transcription. Our results indicate that 5' Hox proteins regulate directly both key genes for early limb AP and PD axis patterning and genes involved, at later stages, in skeletal patterning.


Bone Development/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Bone Development/physiology , Cell Line , Chick Embryo , Chromatin Immunoprecipitation , Humans , Mice , Microarray Analysis , Reverse Transcriptase Polymerase Chain Reaction
...