Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Ther ; 29(9): 2841-2853, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33940155

ABSTRACT

A primary challenge in lentiviral gene therapy of ß-hemoglobinopathies is to maintain low vector copy numbers to avoid genotoxicity while being reliably therapeutic for all genotypes. We designed a high-titer lentiviral vector, LVß-shα2, that allows coordinated expression of the therapeutic ßA-T87Q-globin gene and of an intron-embedded miR-30-based short hairpin RNA (shRNA) selectively targeting the α2-globin mRNA. Our approach was guided by the knowledge that moderate reduction of α-globin chain synthesis ameliorates disease severity in ß-thalassemia. We demonstrate that LVß-shα2 reduces α2-globin mRNA expression in erythroid cells while keeping α1-globin mRNA levels unchanged and ßA-T87Q-globin gene expression identical to the parent vector. Compared with the first ßA-T87Q-globin lentiviral vector that has received conditional marketing authorization, BB305, LVß-shα2 shows 1.7-fold greater potency to improve α/ß ratios. It may thus result in greater therapeutic efficacy and reliability for the most severe types of ß-thalassemia and provide an improved benefit/risk ratio regardless of the ß-thalassemia genotype.


Subject(s)
Genetic Vectors/administration & dosage , RNA, Small Interfering/genetics , alpha-Globins/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , Cell Line , Cells, Cultured , Down-Regulation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Genotype , Humans , K562 Cells , Lentivirus/genetics , Lentivirus/physiology , MicroRNAs/antagonists & inhibitors , Primary Cell Culture , Viral Load , beta-Thalassemia/therapy
2.
J Clin Invest ; 131(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-33914703

ABSTRACT

Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.


Subject(s)
Antineoplastic Agents/pharmacology , Hematologic Neoplasms/drug therapy , Neoplasm Proteins/agonists , Neoplasms, Experimental/drug therapy , PPAR gamma/agonists , Primary Myelofibrosis/drug therapy , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Disease Models, Animal , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Primary Myelofibrosis/genetics , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
3.
BioDrugs ; 34(5): 625-647, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32897504

ABSTRACT

ß-Globin gene transfer has been used as a paradigm for hematopoietic stem cell (HSC) gene therapy, but is subject to major difficulties, such as the lack of selection of genetically corrected HSCs, the need for high-level expression of the therapeutic gene, and cell-specific transgene expression. It took more than 40 years for scientists and physicians to advance from the cloning of globin gene and discovering globin gene mutations to improving our understanding of the pathophysiological mechanisms involved, the detection of genetic modifiers, the development of animal models and gene transfer vectors, comprehensive animal testing, and demonstrations of phenotypic improvement in clinical trials, culminating in the authorization of the first gene therapy product for ß-thalassemia in 2019. Research has focused mostly on the development of lentiviral gene therapy vectors expressing variants of the ß-globin gene or, more recently, targeting a γ-globin repressor, some of which have entered clinical testing and should soon diversify the available treatments and promote price competition. These results are encouraging, but we have yet to reach the end of the story. New molecular and cellular tools, such as gene editing or the development of induced pluripotent stem cells, are being developed, heralding the emergence of alternative products, the efficacy and safety of which are being studied. Hemoglobin disorders constitute an important model for testing the pros and cons of these advanced technologies, some of which are already in the clinical phase. In this review, we focus on the development of the advanced products and recent technological innovations that could lead to clinical trials in the near future, and provide hope for a definitive cure of these severe conditions.


Subject(s)
Genetic Therapy , beta-Thalassemia , Animals , Gene Editing , Genetic Vectors , Therapies, Investigational , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy
4.
Hum Gene Ther ; 30(10): 1306-1323, 2019 10.
Article in English | MEDLINE | ID: mdl-30848170

ABSTRACT

Recent marketing approval for genetically engineered hematopoietic stem and T cells bears witness to the substantial improvements in lentiviral vectors over the last two decades, but evaluations of the long-term efficacy and toxicity of gene and cell therapy products will, nevertheless, require further studies in nonhuman primate models. Macaca fascicularis monkeys from Mauritius have a low genetic diversity and are particularly useful for reproducible drug testing. In particular, they have a genetically homogeneous class I major histocompatibility complex system that probably mitigates the variability of the response to simian immunodeficiency virus infection. However, the transduction of simian cells with human immunodeficiency virus type 1 (HIV-1)-derived vectors is inefficient due to capsid-specific restriction factors, such as the tripartite motif-containing protein tripartite motif 5α, which prevent infection with non-host-adapted retroviruses. This study introduced the modified capsid of the macaque-trophic HIV-1 clone MN4/LSQD into the packaging system and compared transduction efficiencies between hematopoietic cells transduced with this construct and cells transduced with HIV-1 NL4-3-derived packaging constructs. Capsid modification increased transduction efficiency in all hematopoietic cells tested (by factors of up to 10), including hematopoietic progenitor cells, repopulating cells, and T cells from Mauritian Macaca fascicularis, regardless of vector structure or purification method. The study also established culture conditions similar to those used in clinical practice for the efficient transduction of hematopoietic stem and progenitor CD34+ cells. These results suggest that the procedure is suitable for use in Mauritian Macaca fascicularis, which can therefore be used as a model in preclinical studies for hematopoietic gene and cell therapy.


Subject(s)
Capsid/immunology , Genetic Vectors/metabolism , HIV-1/immunology , Hematopoietic Stem Cells/immunology , Macaca fascicularis/immunology , Transduction, Genetic/methods , Animals , Antigens, CD34/genetics , Antigens, CD34/immunology , Biomarkers/metabolism , Capsid/chemistry , Female , Gene Expression , Genetic Vectors/immunology , HIV-1/genetics , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/virology , Male , Mice , Mice, Inbred NOD , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Transplantation, Heterologous , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/immunology
5.
Exp Hematol ; 64: 12-32, 2018 08.
Article in English | MEDLINE | ID: mdl-29807062

ABSTRACT

The ß-hemoglobinopathies, transfusion-dependent ß-thalassemia and sickle cell disease, are the most prevalent inherited disorders worldwide and affect millions of people. Many of these patients have a shortened life expectancy and suffer from severe morbidity despite supportive therapies, which impose an enormous financial burden to societies. The only available curative therapy is allogeneic hematopoietic stem cell transplantation, although most patients do not have an HLA-matched sibling donor, and those who do still risk life-threatening complications. Therefore, gene therapy by one-time ex vivo modification of hematopoietic stem cells followed by autologous engraftment is an attractive new therapeutic modality. The first proof-of-principle of conversion to transfusion independence by means of a lentiviral vector expressing a marked and anti-sickling ßT87Q-globin gene variant was reported a decade ago in a patient with transfusion-dependent ß-thalassemia. In follow-up multicenter Phase II trials with an essentially identical vector (termed LentiGlobin BB305) and protocol, 12 of the 13 patients with a non-ß0/ß0 genotype, representing more than half of all transfusion-dependent ß-thalassemia cases worldwide, stopped red blood cell transfusions with total hemoglobin levels in blood approaching normal values. Correction of biological markers of dyserythropoiesis was achieved in evaluated patients. In nine patients with ß0/ß0 transfusion-dependent ß-thalassemia or equivalent severity (ßIVS1-110), median annualized transfusion volume decreased by 73% and red blood cell transfusions were stopped in three patients. Proof-of-principle of therapeutic efficacy in the first patient with sickle cell disease was also reported with LentiGlobin BB305. Encouraging results were presented in children with transfusion-dependent ß-thalassemia in another trial with the GLOBE lentiviral vector and several other gene therapy trials are currently open for both transfusion-dependent ß-thalassemia and sickle cell disease. Phase III trials are now under way and should help to determine benefit/risk/cost ratios to move gene therapy toward clinical practice.


Subject(s)
Genetic Vectors/therapeutic use , Hemoglobinopathies/therapy , Lentivirus/genetics , Anemia, Sickle Cell/therapy , Blood Transfusion , Clinical Trials as Topic , Developing Countries , Gene Editing , Genetic Vectors/genetics , Global Burden of Disease , Hematopoietic Stem Cell Transplantation , Hemoglobinopathies/epidemiology , Hemoglobinopathies/genetics , Humans , Iron Overload/etiology , Iron Overload/prevention & control , Mutagenesis, Site-Directed , Prevalence , Recombinant Proteins/genetics , Transplantation Conditioning/methods , beta-Globins/genetics , beta-Thalassemia/therapy
6.
Mol Ther ; 26(2): 480-495, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29221807

ABSTRACT

Although gene transfer to hematopoietic stem cells (HSCs) has shown therapeutic efficacy in recent trials for several individuals with inherited disorders, transduction incompleteness of the HSC population remains a hurdle to yield a cure for all patients with reasonably low integrated vector numbers. In previous attempts at HSC selection, massive loss of transduced HSCs, contamination with non-transduced cells, or lack of applicability to large cell populations has rendered the procedures out of reach for human applications. Here, we fused codon-optimized puromycin N-acetyltransferase to herpes simplex virus thymidine kinase. When expressed from a ubiquitous promoter within a complex lentiviral vector comprising the ßAT87Q-globin gene, viral titers and therapeutic gene expression were maintained at effective levels. Complete selection and preservation of transduced HSCs were achieved after brief exposure to puromycin in the presence of MDR1 blocking agents, suggesting the procedure's suitability for human clinical applications while affording the additional safety of conditional suicide.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hemoglobinopathies/genetics , Hemoglobinopathies/therapy , Transduction, Genetic , beta-Globins/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Disease Models, Animal , Gene Expression , Gene Order , Genes, Transgenic, Suicide , Genetic Therapy/methods , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Mice , Mice, Transgenic , Transgenes
7.
Stem Cells ; 31(10): 2162-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23554255

ABSTRACT

Our understanding of system dynamics of mixed cell populations in whole organisms has benefited from the advent of individual cell marking by nonarrayed DNA barcodes subsequently analyzed by high-throughput DNA sequencing. However, key limitations include statistical biases compromising quantification and the lack of applicability to deconvolute individual cell fate in vivo after pooling single cells differentially exposed to different conditions ex vivo. Here, we have derived an arrayed lentiviral library of DNA barcodes and obtained a proof-of-concept of its resolving capacity by quantifying hematopoietic regeneration after engraftment of mice with genetically modified autologous cells. This method has helped clarify and bridge the seemingly opposed clonal-succession and continuous-recruitment models of hematopoietic stem cell behavior and revealed that myeloid-lymphoid biases are common occurrences in steady-state hematopoiesis. Arrayed lentiviral barcoding should prove a versatile and powerful approach to deconvolute cell dynamics in vivo with applications in hematology, embryology, and cancer biology.


Subject(s)
Hematopoietic Stem Cells/physiology , Lentivirus/genetics , Animals , Cell Tracking/methods , DNA Barcoding, Taxonomic , Genetic Vectors , HEK293 Cells , Hematopoietic Stem Cell Transplantation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
8.
EMBO Mol Med ; 5(4): 548-62, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23526803

ABSTRACT

Neurogenesis decreases during aging and following cranial radiotherapy, causing a progressive cognitive decline that is currently untreatable. However, functional neural stem cells remained present in the subventricular zone of high dose-irradiated and aged mouse brains. We therefore investigated whether alterations in the neurogenic niches are perhaps responsible for the neurogenesis decline. This hypothesis was supported by the absence of proliferation of neural stem cells that were engrafted into the vascular niches of irradiated host brains. Moreover, we observed a marked increase in TGF-ß1 production by endothelial cells in the stem cell niche in both middle-aged and irradiated mice. In co-cultures, irradiated brain endothelial cells induced the apoptosis of neural stem/progenitor cells via TGF-ß/Smad3 signalling. Strikingly, the blockade of TGF-ß signalling in vivo using a neutralizing antibody or the selective inhibitor SB-505124 significantly improved neurogenesis in aged and irradiated mice, prevented apoptosis and increased the proliferation of neural stem/progenitor cells. These findings suggest that anti-TGF-ß-based therapy may be used for future interventions to prevent neurogenic collapse following radiotherapy or during aging.


Subject(s)
Aging/metabolism , Brain/growth & development , Brain/radiation effects , Endothelial Cells/metabolism , Neural Stem Cells/metabolism , Neurogenesis/radiation effects , Stem Cell Niche , Transforming Growth Factor beta/metabolism , Aging/radiation effects , Animals , Brain/cytology , Brain/metabolism , Cell Proliferation , Humans , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/radiation effects , Signal Transduction/radiation effects
9.
Cell Cycle ; 7(13): 1911-5, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18604174

ABSTRACT

Defects in DNA repair pathways have been involved in collapse of early neurogenesis leading to brain development abnormalities and embryonic lethality. However, consequences of DNA repair defects in adult neural stem and progenitor cells and their potential contribution in ageing phenotype are poorly understood. The Fanconi anaemia (FA) pathway, which functions primarily as a DNA damage response system, has been examined in neural stem and progenitor cells during developmental and adult neurogenesis. We have shown that loss of fanca and fancg specifically provokes neural progenitor apoptosis during forebrain development, related to DNA repair defects, which persists in adulthood leading to depletion of the neural stem cell pool with ageing. In addition, neural stem cells from FA mice had a reduced capacity to self-renew in vitro. Here, we expand upon our recent work and give further data examining possible implication of oxidative stress. Therefore, FA phenotype might be interpreted as a premature ageing of stem cells, DNA damages being among the driving forces of ageing.


Subject(s)
Brain/metabolism , DNA Repair , Fanconi Anemia Complementation Group A Protein/metabolism , Fanconi Anemia Complementation Group G Protein/metabolism , Fanconi Anemia/metabolism , Neurons/metabolism , Stem Cells/metabolism , Aging/metabolism , Animals , Apoptosis/physiology , Cell Cycle/physiology , DNA Damage , Homeostasis , Mice , Oxidative Stress
10.
Mol Cell Neurosci ; 38(4): 569-77, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18583149

ABSTRACT

Neurogenesis persists in the adult brain subventricular zone where neural stem/progenitor cells (NSPCs) lie close to brain endothelial cells (BECs). We show in mouse that BECs produce bone morphogenetic proteins (BMPs). Coculture of embryonic and adult NSPCs with BECs activated the canonical BMP/Smad pathway and reduced their proliferation. We demonstrate that coculture with BECs in the presence of EGF and FGF2 induced a reversible cell cycle exit of NSPCs (LeX+) and an increase in the amount of GFAP/LeX-expressing progenitors thought to be stem cells. Levels of the phosphatidylinositol phosphatase PTEN were upregulated in NSPCs after coculture with BECs, or treatment with recombinant BMP4, with a concomitant reduction in Akt phosphorylation. Silencing Smad5 with siRNA or treatment with Noggin, a BMP antagonist, demonstrated that upregulation of PTEN in NSPCs required BMP/Smad signaling and that this pathway regulated cell cycle exit of NSPCs. Therefore, BECs may provide a feedback mechanism to control the proliferation of NSPCs.


Subject(s)
Bone Morphogenetic Proteins/physiology , Brain/metabolism , Cell Proliferation , Endothelial Cells/metabolism , Neurons/physiology , Stem Cells/metabolism , Animals , Brain/cytology , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Stem Cells/cytology
11.
EMBO J ; 27(5): 770-81, 2008 Mar 05.
Article in English | MEDLINE | ID: mdl-18239686

ABSTRACT

Although brain development abnormalities and brain cancer predisposition have been reported in some Fanconi patients, the possible role of Fanconi DNA repair pathway during neurogenesis is unclear. We thus addressed the role of fanca and fancg, which are involved in the activation of Fanconi pathway, in neural stem and progenitor cells during brain development and adult neurogenesis. Fanca(-/-) and fancg(-/-) mice presented with microcephalies and a decreased neuronal production in developing cortex and adult brain. Apoptosis of embryonic neural progenitors, but not that of postmitotic neurons, was increased in the neocortex of fanca(-/-) and fancg(-/-) mice and was correlated with chromosomal instability. In adult Fanconi mice, we showed a reduced proliferation of neural progenitor cells related to apoptosis and accentuated neural stem cells exhaustion with ageing. In addition, embryonic and adult Fanconi neural stem cells showed a reduced capacity to self-renew in vitro. Our study demonstrates a critical role for Fanconi pathway in neural stem and progenitor cells during developmental and adult neurogenesis.


Subject(s)
Brain/cytology , Fanconi Anemia Complementation Group A Protein/deficiency , Fanconi Anemia Complementation Group G Protein/deficiency , Neurons/cytology , Stem Cells/cytology , Animals , Apoptosis , Brain/embryology , Cell Proliferation , Chromosome Aberrations , DNA Repair , Embryonic Development , Fanconi Anemia , Fanconi Anemia Complementation Group A Protein/genetics , Fanconi Anemia Complementation Group G Protein/genetics , Female , Mice , Mice, Knockout , Pregnancy
12.
Mol Cell ; 28(4): 584-97, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-18042454

ABSTRACT

The Maf oncoproteins are b-Zip transcription factors of the AP-1 superfamily. They are involved in developmental, metabolic, and tumorigenic processes. Maf proteins are overexpressed in about 50% of human multiple myelomas. Here, we show that Maf-transforming activity is controlled by GSK-3-dependent phosphorylation and that phosphorylation by GSK-3 can increase the oncogenic activity of a protein. Using microarray analysis, we identify a gene-expression subprogram regulated by GSK-3-mediated Maf phosphorylation involved in extracellular matrix remodeling and relevant to cancer progression. We also demonstrate that GSK-3 triggers MafA sequential phosphorylation on residues S61, T57, T53, and S49, inducing its ubiquitination and degradation. Paradoxically, this phosphorylation increases MafA-transcriptional activity through the recruitment of the coactivator P/CAF. We further demonstrate that P/CAF protects MafA from ubiquitination and degradation, suggesting that, upon the release of the coactivator complex, MafA becomes polyubiquitinated and degraded to allow the response to terminate.


Subject(s)
Cell Transformation, Neoplastic , Glycogen Synthase Kinase 3/metabolism , Maf Transcription Factors, Large/metabolism , Amino Acid Sequence , Animals , COS Cells , Cell Line , Chickens , Chlorocebus aethiops , Humans , Maf Transcription Factors, Large/chemistry , Maf Transcription Factors, Large/genetics , Molecular Sequence Data , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , Protein Processing, Post-Translational , Rats , Transcription, Genetic , Ubiquitination , p300-CBP Transcription Factors/metabolism
13.
J Neurochem ; 99(3): 807-17, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16925596

ABSTRACT

Developing and adult forebrains contain neural stem cells (NSCs) but no marker is available to highly purify them. When analysed by flow cytometry, stem cells from various tissues are enriched in a 'side population' (SP) characterized by the exclusion of the fluorescent dye Hoechst 33342. Here, we characterize the SP in embryonic, neonatal and adult forebrains, as well as in neurosphere cultures and we have determined whether this SP could be a source of enriched NSCs. By using specific inhibitors, we found that the SP from embryonic forebrain results from the activity of the ABCG2 transporter, a characteristic of other stem cells, whereas the SP from adult forebrain probably results from the ABCB1 transporter. SP cells from embryonic and adult forebrains, however, expressed a range of cell surface markers more consistent with a haematopoietic/endothelial origin than with a neural origin; NSC markers were mostly expressed on cells outside the SP (in the main population, MP). Moreover, assays for NSC growth in vitro showed that SP cells from embryonic and adult forebrains did not generate NSC-derived colonies, whereas the MP did. We thus conclude that NSCs from developing and adult forebrains are not contained in the SP contrary to stem cells from other tissues.


Subject(s)
Neurons/physiology , Prosencephalon/cytology , Stem Cells/physiology , Animals , Benzimidazoles , Cell Separation , Clone Cells , DNA/biosynthesis , DNA/genetics , Endothelial Cells/physiology , Female , Flow Cytometry , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pregnancy , Prosencephalon/embryology , RNA/biosynthesis , RNA/genetics
14.
FEBS Lett ; 579(17): 3547-54, 2005 Jul 04.
Article in English | MEDLINE | ID: mdl-15963504

ABSTRACT

Basic-leucine zipper transcription factors of the Maf family are key regulators of various developmental and differentiation processes. We previously reported that the phosphorylation status of MafA is a critical determinant of its biological functions. Using Western blot and mass spectrometry analysis, we now show that MafA is phosphorylated by p38 MAP kinase and identify three phosphoacceptor sites: threonine 113 and threonine 57, evolutionarily conserved residues located in the transcription activating domain, and serine 272. Mutation of these residues severely impaired MafA biological activity. Furthermore, we show that p38 also phosphorylates MafB and c-Maf. Together, these findings suggest that the p38 MAP kinase pathway is a novel regulator of large Maf transcription factors.


Subject(s)
Lens, Crystalline/enzymology , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/physiology , Amino Acid Sequence , Animals , Cell Differentiation/genetics , Chickens , DNA-Binding Proteins/physiology , Humans , Lens, Crystalline/cytology , Mice , Molecular Sequence Data , Mutation , Phosphorylation , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-maf , Quail , Serine/genetics , Threonine/genetics , Transcription Factors/genetics
15.
Gene Expr Patterns ; 4(1): 35-46, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14678826

ABSTRACT

Maf proteins are basic-leucine zipper transcription factors belonging to the AP1 superfamily. Several developmental processes require Maf proteins yet, the redundancy or complementarity of their respective roles in common processes has been only partially investigated. We present for the first time a complete comparative analysis of maf gene expression patterns in vertebrates. Expression of c-maf, mafB/kreisler, mafA/L-maf, mafF, mafG and mafK was analyzed by whole-mount in situ hybridization within chick embryos and their extraembryonic tissues ranging from embryonic day (E) 1 to 7. We carefully examined the extent of overlap between distinct maf genes and report that the developing lens, kidney, pancreas and apoptotic zones of limb buds show sustained co-expression of large maf genes. Small maf genes also exhibit overlap, for example in the dermomyotome. We also describe so far unidentified sites of maf gene expression. mafA is found in the developing neural tube and dorsal root ganglia. c-maf hybridization is detected in the neuroretina, the notochord and the endothelium of extraembryonic blood vessels.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Profiling , Proto-Oncogene Proteins/genetics , Animals , Chick Embryo , Gastrula/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Limb Buds/embryology , Limb Buds/metabolism , MafF Transcription Factor , MafK Transcription Factor , Mesoderm/metabolism , Nuclear Proteins/genetics , Pancreas/embryology , Pancreas/metabolism , Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism , Proto-Oncogene Proteins c-maf , Repressor Proteins/genetics , Retina/embryology , Retina/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...