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










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(50): e2201097119, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36469766

ABSTRACT

Despite the robust healing capacity of the liver, regenerative failure underlies numerous hepatic diseases, including the JAG1 haploinsufficient disorder, Alagille syndrome (ALGS). Cholestasis due to intrahepatic duct (IHD) paucity resolves in certain ALGS cases but fails in most with no clear mechanisms or therapeutic interventions. We find that modulating jag1b and jag2b allele dosage is sufficient to stratify these distinct outcomes, which can be either exacerbated or rescued with genetic manipulation of Notch signaling, demonstrating that perturbations of Jag/Notch signaling may be causal for the spectrum of ALGS liver severities. Although regenerating IHD cells proliferate, they remain clustered in mutants that fail to recover due to a blunted elevation of Notch signaling in the distal-most IHD cells. Increased Notch signaling is required for regenerating IHD cells to branch and segregate into the peripheral region of the growing liver, where biliary paucity is commonly observed in ALGS. Mosaic loss- and-gain-of-function analysis reveals Sox9b to be a key Notch transcriptional effector required cell autonomously to regulate these cellular dynamics during IHD regeneration. Treatment with a small-molecule putative Notch agonist stimulates Sox9 expression in ALGS patient fibroblasts and enhances hepatic sox9b expression, rescues IHD paucity and cholestasis, and increases survival in zebrafish mutants, thereby providing a proof-of-concept therapeutic avenue for this disorder.


Subject(s)
Alagille Syndrome , Bile Ducts, Intrahepatic , Signal Transduction , Animals , Humans , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Mosaicism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Regeneration , Bile Ducts, Intrahepatic/cytology , Bile Ducts, Intrahepatic/pathology , Fibroblasts
2.
Dis Model Mech ; 15(4)2022 04 01.
Article in English | MEDLINE | ID: mdl-35199829

ABSTRACT

An accurate prediction of the intracranial infiltration tendency and drug response of individual glioblastoma (GBM) cells is essential for personalized prognosis and treatment for this disease. However, the clinical utility of mouse patient-derived orthotopic xenograft (PDOX) models remains limited given current technical constraints, including difficulty in generating sufficient sample numbers from small tissue samples and a long latency period for results. To overcome these issues, we established zebrafish GBM xenografts of diverse origin, which can tolerate intracranial engraftment and maintain their unique histological features. Subsequent single-cell RNA-sequencing (scRNA-seq) analysis confirmed significant transcriptional identity to that of invading GBM microtumors observed in the proportionally larger brains of model animals and humans. Endothelial scRNA-seq confirmed that the zebrafish blood-brain barrier is homologous to the mammalian blood-brain barrier. Finally, we established a rapid and efficient zebrafish PDOX (zPDOX) model, which can predict long-term outcomes of GBM patients within 20 days. The zPDOX model provides a novel avenue for precision medicine of GBM, especially for the evaluation of intracranial infiltration tendency and prediction of individual drug sensitivity.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Brain Neoplasms/pathology , Disease Models, Animal , Glioblastoma/pathology , Heterografts , Humans , Mammals , Mice , Xenograft Model Antitumor Assays , Zebrafish
3.
Hepatology ; 75(3): 567-583, 2022 03.
Article in English | MEDLINE | ID: mdl-34569629

ABSTRACT

BACKGROUND AND AIMS: Alagille Syndrome (ALGS) is a congenital disorder caused by mutations in the Notch ligand gene JAGGED1, leading to neonatal loss of intrahepatic duct (IHD) cells and cholestasis. Cholestasis can resolve in certain patients with ALGS, suggesting regeneration of IHD cells. However, the mechanisms driving IHD cell regeneration following Jagged loss remains unclear. Here, we show that cholestasis due to developmental loss of IHD cells can be consistently phenocopied in zebrafish with compound jagged1b and jagged2b mutations or knockdown. APPROACH AND RESULTS: Leveraging the transience of jagged knockdown in juvenile zebrafish, we find that resumption of Jagged expression leads to robust regeneration of IHD cells through a Notch-dependent mechanism. Combining multiple lineage tracing strategies with whole-liver three-dimensional imaging, we demonstrate that the extrahepatic duct (EHD) is the primary source of multipotent progenitors that contribute to the regeneration, but not to the development, of IHD cells. Hepatocyte-to-IHD cell transdifferentiation is possible but rarely detected. Progenitors in the EHD proliferate and migrate into the liver with Notch signaling loss and differentiate into IHD cells if Notch signaling increases. Tissue-specific mosaic analysis with an inducible dominant-negative Fgf receptor suggests that Fgf signaling from the surrounding mesenchymal cells maintains this extrahepatic niche by directly preventing premature differentiation and allocation of EHD progenitors to the liver. Indeed, transcriptional profiling and functional analysis of adult mouse EHD organoids uncover their distinct differentiation and proliferative potential relative to IHD organoids. CONCLUSIONS: Our data show that IHD cells regenerate upon resumption of Jagged/Notch signaling, from multipotent progenitors originating from an Fgf-dependent extrahepatic stem cell niche. We posit that if Jagged/Notch signaling is augmented, through normal stochastic variation, gene therapy, or a Notch agonist, regeneration of IHD cells in patients with ALGS may be enhanced.


Subject(s)
Alagille Syndrome , Bile Ducts, Extrahepatic , Bile Ducts, Intrahepatic , Calcium-Binding Proteins , Jagged-1 Protein , Liver Regeneration/physiology , Receptors, Notch/metabolism , Zebrafish Proteins , Alagille Syndrome/genetics , Alagille Syndrome/metabolism , Animals , Bile Ducts, Extrahepatic/growth & development , Bile Ducts, Extrahepatic/physiology , Bile Ducts, Intrahepatic/growth & development , Bile Ducts, Intrahepatic/physiology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Transdifferentiation , Disease Models, Animal , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Liver/growth & development , Liver/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Dev Dyn ; 251(9): 1439-1455, 2022 09.
Article in English | MEDLINE | ID: mdl-34719843

ABSTRACT

BACKGROUND: The phalanges are the final skeletal elements to form in the vertebrate limb and their identity is regulated by signaling at the phalanx forming region (PFR) located at the tip of the developing digit ray. Here, we seek to explore the relationship between PFR activity and phalanx morphogenesis, which define the most distal limb skeletal elements, and signals associated with termination of limb outgrowth. RESULTS: As Grem1 is extinguished in the distal chick limb mesoderm, the chondrogenesis marker Aggrecan is up-regulated in the metatarsals and phalanges. Fate mapping confirms that subridge mesoderm cells contribute to the metatarsal and phalanges when subridge Grem1 is down-regulated. Grem1 overexpression specifically blocks chick phalanx development by inhibiting PFR activity. PFR activity and digit development are also disrupted following overexpression of a Gli3 repressor, which results in Grem1 expression in the distal limb and downregulation of Bmpr1b. CONCLUSIONS: Based on expression and fate mapping studies, we propose that downregulation of Grem1 in the distal limb marks the transition from metatarsal to phalanx development. This suggests that downregulation of Grem1 in the distal limb mesoderm is necessary for phalanx development. Grem1 downregulation allows for full PFR activity and phalanx progenitor cell commitment to digit fate.


Subject(s)
Gene Expression Regulation, Developmental , Mesoderm , Down-Regulation , Extremities , Limb Buds/metabolism , Mesoderm/metabolism , Signal Transduction
5.
Elife ; 102021 02 05.
Article in English | MEDLINE | ID: mdl-33544077

ABSTRACT

Genetic variants associated with type 2 diabetes (T2D) risk affect gene regulation in metabolically relevant tissues, such as pancreatic islets. Here, we investigated contributions of regulatory programs active during pancreatic development to T2D risk. Generation of chromatin maps from developmental precursors throughout pancreatic differentiation of human embryonic stem cells (hESCs) identifies enrichment of T2D variants in pancreatic progenitor-specific stretch enhancers that are not active in islets. Genes associated with progenitor-specific stretch enhancers are predicted to regulate developmental processes, most notably tissue morphogenesis. Through gene editing in hESCs, we demonstrate that progenitor-specific enhancers harboring T2D-associated variants regulate cell polarity genes LAMA1 and CRB2. Knockdown of lama1 or crb2 in zebrafish embryos causes a defect in pancreas morphogenesis and impairs islet cell development. Together, our findings reveal that a subset of T2D risk variants specifically affects pancreatic developmental programs, suggesting that dysregulation of developmental processes can predispose to T2D.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Epigenome , Intracellular Signaling Peptides and Proteins/genetics , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Transcription Factors/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
6.
Cell Chem Biol ; 28(5): 625-635.e5, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33503403

ABSTRACT

Wnt signaling plays a central role in tissue maintenance and cancer. Wnt activates downstream genes through ß-catenin, which interacts with TCF/LEF transcription factors. A major question is how this signaling is coordinated relative to tissue organization and renewal. We used a recently described class of small molecules that binds tubulin to reveal a molecular cascade linking stress signaling through ATM, HIPK2, and p53 to the regulation of TCF/LEF transcriptional activity. These data suggest a mechanism by which mitotic and genotoxic stress can indirectly modulate Wnt responsiveness to exert coherent control over cell shape and renewal. These findings have implications for understanding tissue morphogenesis and small-molecule anticancer therapeutics.


Subject(s)
Molecular Probes/pharmacology , Protein Serine-Threonine Kinases/metabolism , Small Molecule Libraries/pharmacology , TCF Transcription Factors/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , Animals , Cells, Cultured , Humans , Male , Molecular Probes/chemistry , Small Molecule Libraries/chemistry , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects , Xenopus , Zebrafish , beta Catenin/genetics , beta Catenin/metabolism
7.
Science ; 370(6515): 463-467, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33093109

ABSTRACT

Vertebrate sensory organs arise from epithelial thickenings called placodes. Along with neural crest cells, cranial placodes are considered ectodermal novelties that drove evolution of the vertebrate head. The anterior-most placode generates the endocrine lobe [adenohypophysis (ADH)] of the pituitary, a master gland controlling growth, metabolism, and reproduction. In addition to known ectodermal contributions, we use lineage tracing and time-lapse imaging in zebrafish to identify an endodermal contribution to the ADH. Single-cell RNA sequencing of the adult pituitary reveals similar competency of endodermal and ectodermal epithelia to generate all endocrine cell types. Further, endoderm can generate a rudimentary ADH-like structure in the near absence of ectodermal contributions. The fish condition supports the vertebrate pituitary arising through interactions of an ancestral endoderm-derived proto-pituitary with newly evolved placodal ectoderm.


Subject(s)
Endoderm/embryology , Pituitary Gland, Anterior/embryology , Animals , Cell Lineage , Endoderm/cytology , Pituitary Gland, Anterior/cytology , RNA-Seq , Single-Cell Analysis , Zebrafish
8.
Development ; 146(14)2019 07 24.
Article in English | MEDLINE | ID: mdl-31142539

ABSTRACT

An early step in pancreas development is marked by the expression of the transcription factor Pdx1 within the pancreatic endoderm, where it is required for the specification of all endocrine cell types. Subsequently, Pdx1 expression becomes restricted to the ß-cell lineage, where it plays a central role in ß-cell function. This pivotal role of Pdx1 at various stages of pancreas development makes it an attractive target to enhance pancreatic ß-cell differentiation and increase ß-cell function. In this study, we used a newly generated zebrafish reporter to screen over 8000 small molecules for modulators of pdx1 expression. We found four hit compounds and validated their efficacy at different stages of pancreas development. Notably, valproic acid treatment increased pancreatic endoderm formation, while inhibition of TGFß signaling led to α-cell to ß-cell transdifferentiation. HC toxin, another HDAC inhibitor, enhances ß-cell function in primary mouse and human islets. Thus, using a whole organism screening strategy, this study identified new pdx1 expression modulators that can be used to influence different steps in pancreas and ß-cell development.


Subject(s)
Drug Evaluation, Preclinical/methods , Islets of Langerhans/embryology , Models, Animal , Organogenesis/drug effects , Small Molecule Libraries/analysis , Zebrafish , Animals , Animals, Genetically Modified , COS Cells , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Transdifferentiation/drug effects , Cell Transdifferentiation/genetics , Cells, Cultured , Chlorocebus aethiops , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Histone Deacetylase Inhibitors/isolation & purification , Histone Deacetylase Inhibitors/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Islets of Langerhans/drug effects , Islets of Langerhans/growth & development , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Organogenesis/genetics , Small Molecule Libraries/isolation & purification , Trans-Activators/genetics , Trans-Activators/metabolism , Valproic Acid/isolation & purification , Valproic Acid/pharmacology , Zebrafish/embryology , Zebrafish/genetics
9.
Dev Biol ; 441(1): 127-131, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29964026

ABSTRACT

In mouse, retinoic acid (RA) is required for the early phase of body axis extension controlled by a population of neuromesodermal progenitors (NMPs) in the trunk called expanding-NMPs, but not for the later phase of body axis extension controlled by a population of NMPs in the tail called depleting-NMPs. Recent observations suggest that zebrafish utilize depleting-NMPs but not expanding-NMPs for body axis extension. In zebrafish, a role for RA in body axis extension was not supported by previous studies on aldh1a2 (raldh2) mutants lacking RA synthesis. Here, by treating zebrafish embryos with an RA synthesis inhibitor, we also found that body axis extension and somitogenesis was not perturbed, although loss of pectoral fin and cardiac edema were observed consistent with previous studies. The conclusion that zebrafish diverges from mouse in not requiring RA for body axis extension is consistent with zebrafish lacking early expanding-NMPs to generate the trunk. We suggest that RA control of body axis extension was added to higher vertebrates during evolution of expanding-NMPs.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Nonmammalian/embryology , Mesoderm/embryology , Neural Stem Cells/metabolism , Tretinoin/metabolism , Zebrafish/embryology , Animals , Embryo, Mammalian/cytology , Embryo, Nonmammalian/cytology , Mesoderm/cytology , Mice , Neural Stem Cells/cytology , Species Specificity
10.
Cell Rep ; 23(11): 3146-3151, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898387

ABSTRACT

A standard approach in the identification of transcriptional enhancers is the use of transgenic animals carrying DNA elements joined to reporter genes inserted randomly in the genome. We examined elements near Tbx5, a gene required for forelimb development in humans and other vertebrates. Previous transgenic studies reported a mammalian Tbx5 forelimb enhancer located in intron 2 containing a putative retinoic acid response element and a zebrafish tbx5a forelimb (pectoral fin) enhancer located downstream that is conserved from fish to mammals. We used CRISPR/Cas9 gene editing to knockout the endogenous elements and unexpectedly found that deletion of the intron 2 and downstream elements, either singly or together in double knockouts, resulted in no effect on forelimb development. Our findings show that reporter transgenes may not identify endogenous enhancers and that in vivo genetic loss-of-function studies are required, such as CRISPR/Cas9, which is similar in effort to production of animals carrying reporter transgenes.


Subject(s)
Enhancer Elements, Genetic/genetics , Forelimb/growth & development , Gene Editing , T-Box Domain Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified/metabolism , CRISPR-Cas Systems/genetics , Forelimb/metabolism , Introns , Mice , Zebrafish/metabolism
11.
Nat Commun ; 8(1): 769, 2017 10 03.
Article in English | MEDLINE | ID: mdl-28974684

ABSTRACT

Liver duct paucity is characteristic of children born with Alagille Syndrome (ALGS), a disease associated with JAGGED1 mutations. Here, we report that zebrafish embryos with compound homozygous mutations in two Notch ligand genes, jagged1b (jag1b) and jagged2b (jag2b) exhibit a complete loss of canonical Notch activity and duct cells within the liver and exocrine pancreas, whereas hepatocyte and acinar pancreas development is not affected. Further, animal chimera studies demonstrate that wild-type endoderm cells within the liver and pancreas can rescue Notch activity and duct lineage specification in adjacent cells lacking jag1b and jag2b expression. We conclude that these two Notch ligands are directly and solely responsible for all duct lineage specification in these organs in zebrafish. Our study uncovers genes required for lineage specification of the intrahepatopancreatic duct cells, challenges the role of duct cells as progenitors, and suggests a genetic mechanism for ALGS ductal paucity.The hepatopancreatic duct cells connect liver hepatocytes and pancreatic acinar cells to the intestine, but the mechanism for their lineage specification is unclear. Here, the authors reveal that Notch ligands Jagged1b and Jagged2b induce duct cell lineage in the liver and pancreas of the zebrafish.


Subject(s)
Bile Ducts, Intrahepatic/embryology , Calcium-Binding Proteins/genetics , Endoderm/metabolism , Gene Expression Regulation, Developmental , Jagged-2 Protein/genetics , Pancreatic Ducts/embryology , Zebrafish Proteins/genetics , Alagille Syndrome/genetics , Animals , Cell Lineage , Endoderm/cytology , Zebrafish
12.
Elife ; 62017 04 07.
Article in English | MEDLINE | ID: mdl-28387645

ABSTRACT

The evolutionary origins of the hypoxia-sensitive cells that trigger amniote respiratory reflexes - carotid body glomus cells, and 'pulmonary neuroendocrine cells' (PNECs) - are obscure. Homology has been proposed between glomus cells, which are neural crest-derived, and the hypoxia-sensitive 'neuroepithelial cells' (NECs) of fish gills, whose embryonic origin is unknown. NECs have also been likened to PNECs, which differentiate in situ within lung airway epithelia. Using genetic lineage-tracing and neural crest-deficient mutants in zebrafish, and physical fate-mapping in frog and lamprey, we find that NECs are not neural crest-derived, but endoderm-derived, like PNECs, whose endodermal origin we confirm. We discover neural crest-derived catecholaminergic cells associated with zebrafish pharyngeal arch blood vessels, and propose a new model for amniote hypoxia-sensitive cell evolution: endoderm-derived NECs were retained as PNECs, while the carotid body evolved via the aggregation of neural crest-derived catecholaminergic (chromaffin) cells already associated with blood vessels in anamniote pharyngeal arches.


Subject(s)
Cell Hypoxia , Cell Lineage , Neuroendocrine Cells , Neuroepithelial Cells , Animals , Anura , Biological Evolution , Lampreys , Zebrafish
13.
Development ; 140(13): 2669-79, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23720049

ABSTRACT

Although the liver and ventral pancreas are thought to arise from a common multipotent progenitor pool, it is unclear whether these progenitors of the hepatopancreas system are specified by a common genetic mechanism. Efforts to determine the role of Hnf1b and Wnt signaling in this crucial process have been confounded by a combination of factors, including a narrow time frame for hepatopancreas specification, functional redundancy among Wnt ligands, and pleiotropic defects caused by either severe loss of Wnt signaling or Hnf1b function. Using a novel hypomorphic hnf1ba zebrafish mutant that exhibits pancreas hypoplasia, as observed in HNF1B monogenic diabetes, we show that hnf1ba plays essential roles in regulating ß-cell number and pancreas specification, distinct from its function in regulating pancreas size and liver specification, respectively. By combining Hnf1ba partial loss of function with conditional loss of Wnt signaling, we uncover a crucial developmental window when these pathways synergize to specify the entire ventrally derived hepatopancreas progenitor population. Furthermore, our in vivo genetic studies demonstrate that hnf1ba generates a permissive domain for Wnt signaling activity in the foregut endoderm. Collectively, our findings provide a new model for HNF1B function, yield insight into pancreas and ß-cell development, and suggest a new mechanism for hepatopancreatic specification.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatopancreas/cytology , Hepatopancreas/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Differentiation/physiology , Hepatocyte Nuclear Factor 1-beta/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Wnt Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics
14.
Dev Dyn ; 234(4): 948-60, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16245339

ABSTRACT

We have cloned the chicken and mouse orthologues of the Caenorhabditis elegans heterochronic gene lin-41. During limb development, lin-41 is expressed in three phases over developmental time and most notably is associated with the developing autopod. Using chicken and mouse mutants and bead implantations, we report that lin-41 is genetically and biochemically downstream of both the Shh and Fgf signaling pathways. In C. elegans, it is proposed that lin-41 activity is temporally regulated by miRNAs (let-7 and lin-4) that bind to complementary sites in the lin-41 3'-untranslated region (UTR). Taking a bioinformatics approach, we also report the presence of potential miRNA binding sites in the 3'-UTR of chicken lin-41, including sites for the chicken orthologues of both C. elegans let-7 and lin-4. Finally, we show that these miRNAs and others are expressed in the chick limb consistent with the hypothesis that they regulate chicken Lin-41 activity in vivo.


Subject(s)
Extremities/embryology , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Blotting, Northern , Chick Embryo , Cloning, Molecular , Computational Biology , Gene Components , In Situ Hybridization , Mice , Mice, Mutant Strains , MicroRNAs/genetics , Microspheres , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, DNA , Signal Transduction/genetics , Species Specificity
15.
Development ; 130(3): 527-37, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12490559

ABSTRACT

We have analyzed a new limb mutant in the chicken that we name oligozeugodactyly (ozd). The limbs of this mutant have a longitudinal postaxial defect, lacking the posterior element in the zeugopod (ulna/fibula) and all digits except digit 1 in the leg. Classical recombination experiments show that the limb mesoderm is the defective tissue layer in ozd limb buds. Molecular analysis revealed that the ozd limbs develop in the absence of Shh expression, while all other organs express Shh and develop normally. Neither Ptc1 nor Gli1 are detectable in mutant limb buds. However, Bmp2 and dHAND are expressed in the posterior wing and leg bud mesoderm, although at lower levels than in normal embryos. Activation of Hoxd11-13 occurs normally in ozd limbs but progressively declines with time. Phase III of expression is more affected than phase II, and expression is more severely affected in the more 5' genes. Interestingly, re-expression of Hoxd13 occurs at late stages in the distal mesoderm of ozd leg buds, correlating with formation of digit 1. Fgf8 and Fgf4 expression are initiated normally in the mutant AER but their expression is progressively downregulated in the anterior AER. Recombinant Shh protein or ZPA grafts restore normal pattern to ozd limbs; however, retinoic acid fails to induce Shh in ozd limb mesoderm. We conclude that Shh function is required for limb development distal to the elbow/knee joints, similar to the Shh(-/-) mouse. Accordingly we classify the limb skeletal elements as Shh dependent or independent, with the ulna/fibula and digits other than digit 1 in the leg being Shh dependent. Finally we propose that the ozd mutation is most likely a defect in a regulatory element that controls limb-specific expression of Shh.


Subject(s)
Limb Deformities, Congenital/embryology , Limb Deformities, Congenital/genetics , Mutation , Trans-Activators/deficiency , Animals , Apoptosis , Body Patterning/genetics , Cell Division , Cell Survival , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Hedgehog Proteins , Mesoderm/cytology , Phenotype , Trans-Activators/genetics , Trans-Activators/physiology , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...