Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Neurosci ; 41(8): 1650-1664, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33452226

ABSTRACT

Promoting oligodendrocyte (OL) differentiation represents a promising option for remyelination therapy for treating the demyelinating disease multiple sclerosis (MS). The Wnt effector transcription factor 7-like 2 (TCF7l2) was upregulated in MS lesions and had been proposed to inhibit OL differentiation. Recent data suggest the opposite yet underlying mechanisms remain elusive. Here, we unravel a previously unappreciated function of TCF7l2 in controlling autocrine bone morphogenetic protein (BMP)4-mediated signaling. Disrupting TCF7l2 in mice of both sexes results in oligodendroglial-specific BMP4 upregulation and canonical BMP4 signaling activation in vivo Mechanistically, TCF7l2 binds to Bmp4 gene regulatory element and directly represses its transcriptional activity. Functionally, enforced TCF7l2 expression promotes OL differentiation by reducing autocrine BMP4 secretion and dampening BMP4 signaling. Importantly, compound genetic disruption demonstrates that oligodendroglial-specific BMP4 deletion rescues arrested OL differentiation elicited by TCF7l2 disruption in vivo Collectively, our study reveals a novel connection between TCF7l2 and BMP4 in oligodendroglial lineage and provides new insights into augmenting TCF7l2 for promoting remyelination in demyelinating disorders such as MS.SIGNIFICANCE STATEMENT Incomplete or failed myelin repairs, primarily resulting from the arrested differentiation of myelin-forming oligodendrocytes (OLs) from oligodendroglial progenitor cells, is one of the major reasons for neurologic progression in people affected by multiple sclerosis (MS). Using in vitro culture systems and in vivo animal models, this study unraveled a previously unrecognized autocrine regulation of bone morphogenetic protein (BMP)4-mediated signaling by the Wnt effector transcription factor 7-like 2 (TCF7l2). We showed for the first time that TCF7l2 promotes oligodendroglial differentiation by repressing BMP4-mediated activity, which is dysregulated in MS lesions. Our study suggests that elevating TCF7l2 expression may be possible in overcoming arrested oligodendroglial differentiation as observed in MS patients.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Gene Expression Regulation/physiology , Neurogenesis/physiology , Oligodendroglia/cytology , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Autocrine Communication/physiology , Brain/cytology , Brain/metabolism , Cell Differentiation/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism
2.
J Neurosci ; 38(7): 1802-1820, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29335358

ABSTRACT

In the CNS, myelination and remyelination depend on the successful progression and maturation of oligodendroglial lineage cells, including proliferation and differentiation of oligodendroglial progenitor cells (OPCs). Previous studies have reported that Sox2 transiently regulates oligodendrocyte (OL) differentiation in the embryonic and perinatal spinal cord and appears dispensable for myelination in the postnatal spinal cord. However, the role of Sox2 in OL development in the brain has yet to be defined. We now report that Sox2 is an essential positive regulator of developmental myelination in the postnatal murine brain of both sexes. Stage-specific paradigms of genetic disruption demonstrated that Sox2 regulated brain myelination by coordinating upstream OPC population supply and downstream OL differentiation. Transcriptomic analyses further supported a crucial role of Sox2 in brain developmental myelination. Consistently, oligodendroglial Sox2-deficient mice developed severe tremors and ataxia, typical phenotypes indicative of hypomyelination, and displayed severe impairment of motor function and prominent deficits of brain OL differentiation and myelination persisting into the later CNS developmental stages. We also found that Sox2 was required for efficient OPC proliferation and expansion and OL regeneration during remyelination in the adult brain and spinal cord. Together, our genetic evidence reveals an essential role of Sox2 in brain myelination and CNS remyelination, and suggests that manipulation of Sox2 and/or Sox2-mediated downstream pathways may be therapeutic in promoting CNS myelin repair.SIGNIFICANCE STATEMENT Promoting myelin formation and repair has translational significance in treating myelin-related neurological disorders, such as periventricular leukomalacia and multiple sclerosis in which brain developmental myelin formation and myelin repair are severely affected, respectively. In this report, analyses of a series of genetic conditional knock-out systems targeting different oligodendrocyte stages reveal a previously unappreciated role of Sox2 in coordinating upstream proliferation and downstream differentiation of oligodendroglial lineage cells in the mouse brain during developmental myelination and CNS remyelination. Our study points to the potential of manipulating Sox2 and its downstream pathways to promote oligodendrocyte regeneration and CNS myelin repair.


Subject(s)
Brain/growth & development , Brain/physiology , Central Nervous System/physiology , Myelin Sheath/physiology , Oligodendroglia/physiology , SOXB1 Transcription Factors/physiology , Animals , Cell Differentiation/physiology , Cell Proliferation , Demyelinating Diseases/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Movement Disorders/physiopathology , Nerve Regeneration/physiology , Spinal Cord/growth & development , Spinal Cord/physiology , Transcriptome
3.
Dalton Trans ; 46(38): 12981-12987, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28933485

ABSTRACT

A series of new lanthanide-containing peroxoisopolyoxotungstates, K6Na4[H32{Ln4(WO4)(H2O)16[W7O22(O2)2]4}3]·105H2O [Ln = CeIII (1), NdIII (2), SmIII (3), TbIII (4), ErIII (5)], have been successfully synthesized and structurally characterized. All polyanions [Ln(WO4)(H2O)16{W7O22(O2)2}4]14- are isostructural and consist of a central [Ln(WO4)(H2O)16]10+ cluster surrounded by four peripheral [W7O22(O2)2]6- units. They could act as efficient recyclable catalysts for the epoxidation of various alkenes including different cycloalkenes, styrene derivatives, internal and long-chain alkenes. Under optimal conditions, catalyst 2 displays the best catalytic activity for the oxidation of cyclooctene with high cyclooctene conversion (98.3%) and excellent selectivity (up to 99%) and could be reused for three cycles with a negligible decrease in reactivity.

4.
Neurobiol Dis ; 100: 30-38, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28042097

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative condition caused by age-related death of dopaminergic (DA) neurons in the substantia nigra (SN). Mitochondrial DNA (mtDNA) deletions rise exponentially with age in humans and reach their highest levels approaching 60% in dopaminergic neurons of the substantia nigra and overlap with dying neurons. Parkin deletion causes Parkinsonism in humans, presumably through a decrease in mitochondrial quality control, but Parkin knockout mice do not have DA neurodegeneration. We crossed Parkin knockouts to the Twinkle-TG mouse in which mtDNA deletions are increased specifically in substantia nigra to determine the effect of increased deletion mutagenesis in the absence of mitochondrial quality control. These double-mutant 'TwinkPark' mice had 1, the highest mtDNA deletion concentration in SN; 2, the lowest mitochondrial function and membrane potential; 3, the most severe neurobehavioral deficits at 19months; 4, the least dopaminergic neurons in the SN and lowest dopamine levels, i.e. Parkinsonism. This mouse model could provide novel insights into the pathomechanism by which a specific increase in mtDNA deletions with age contribute to dopaminergic neurodegeneration and Parkinson's disease.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/genetics , Parkinsonian Disorders/genetics , Ubiquitin-Protein Ligases/metabolism , Animals , DNA, Mitochondrial/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Mice, Transgenic , Mutation/genetics , Parkinsonian Disorders/metabolism , Substantia Nigra/metabolism , Ubiquitin-Protein Ligases/deficiency
5.
Brain Res ; 1657: 232-244, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28027875

ABSTRACT

Inherited mitochondrial complex 1 deficiency causes Leber's hereditary Optic Neuropathy (LHON) and retinal ganglion cell (RGC) degeneration, and optic neuropathies are common in many inherited mitochondrial diseases. How mitochondrial defects pathomechanistically trigger optic neuropathy remains unclear. We observe that complex 1-deficient Ndufs4-/- mice present with acute vision loss around p30, and this vision loss is coincident with an 'inflammatory wave'. In order to understand what causes the inflammatory wave we explored retinal pathology that occurs from p20-p30. The results indicated that in the period p20-p30 in Ndufs4-/- retinas, there is: significant reduction in bipolar cells, RGC dendritic atrophy, reduced PSD95, increased oxidative stress as manifested by increased 4HNE, HO1 and Cuzn-SOD, increased mitochondrial biogenesis and increased apoptosis. These precede the major induction of 'inflammatory wave' at p30 shown previously, but occur earlier than frank RGC loss at p42. In general, complex 1 deficiency in retina triggers oxidative stress and mitochondrial respiratory dysfunction that causes death of the most sensitive cells, including bipolar cells and their synaptic contacts and amacrine cells in the early period, 20-24days. The early death of these cells is the likely precursor to the sharp rise in inflammatory molecules that occurs at day 30 and coincides with vision loss, and greatly precedes the death of RGCs that occurs at p42. These data suggest that metabolic antioxidant support of the most sensitive cells in the retina, or anti-inflammatory suppression of the consequences of their death, are both rational strategies for mitochondrial blinding disease.


Subject(s)
Electron Transport Complex I/deficiency , Mitochondria/metabolism , Retinal Bipolar Cells/metabolism , Retinal Ganglion Cells/metabolism , Amacrine Cells/metabolism , Amacrine Cells/pathology , Animals , Apoptosis/physiology , Atrophy , Dendrites/metabolism , Dendrites/pathology , Disease Models, Animal , Disease Progression , Electron Transport Complex I/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Optic Atrophy, Hereditary, Leber/metabolism , Optic Atrophy, Hereditary, Leber/pathology , Organelle Biogenesis , Oxidative Stress/physiology , Post-Synaptic Density/metabolism , Post-Synaptic Density/pathology , Retinal Bipolar Cells/pathology , Retinal Ganglion Cells/pathology
7.
Hum Mol Genet ; 24(10): 2848-60, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25652399

ABSTRACT

Mitochondrial complex I (NADH dehydrogenase) is a major contributor to neuronal energetics, and mutations in complex I lead to vision loss. Functional, neuroanatomical and transcriptional consequences of complex I deficiency were investigated in retinas of the Ndufs4 knockout mouse. Whole-eye ERGs and multielectrode arrays confirmed a major retinal ganglion cell functional loss at P32, and retinal ganglion cell loss at P42. RNAseq demonstrated a mild and then sharp increase in innate immune and inflammatory retinal transcripts at P22 and P33, respectively, which were confirmed with QRT-PCR. Intraperitoneal injection of the inflammogen lipopolysaccharide further reduced retinal ganglion cell function in Ndufs4 KO, supporting the connection between inflammatory activation and functional loss. Complex I deficiency in the retina clearly caused innate immune and inflammatory markers to increase coincident with loss of vision, and RGC functional loss. How complex I incites inflammation and functional loss is not clear, but could be the result of misfolded complex I generating a 'non-self' response, and induction of innate immune response transcripts was observed before functional loss at P22, including ß-2 microglobulin and Cx3cr1, and during vision loss at P31 (B2m, Tlr 2, 3, 4, C1qa, Cx3cr1 and Fas). These data support the hypothesis that mitochondrial complex I dysfunction in the retina triggers an innate immune and inflammatory response that results in loss of retinal ganglion cell function and death, as in Leber's hereditary Optic Neuropathy and suggests novel therapeutic routes to counter mitochondrial defects that contribute to vision loss.


Subject(s)
Electron Transport Complex I/deficiency , Mitochondrial Diseases/physiopathology , Retina/physiopathology , Retinal Ganglion Cells/physiology , Animals , Cell Death , Electron Transport Complex I/genetics , Electron Transport Complex I/immunology , Female , Gene Knockout Techniques , Immunity, Innate/genetics , Inflammation/genetics , Male , Mice , Mice, Knockout , Mitochondrial Diseases/genetics , Mitochondrial Diseases/immunology , Retina/immunology
8.
Brain Res ; 1594: 82-91, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25446449

ABSTRACT

Parkinson׳s disease (PD) is the second most common neurodegenerative disorder in the developed world, and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN) of midbrain. Mitochondrial complex I dysfunction has been implicated in PD pathophysiology, yet the molecular mechanism by which complex I defects may cause DA neurodegeneration remain unclear. Using Ndufs4 mouse model of mitochondrial complex I deficiency, we observed a remarkable ubiquitin protein increase in SN of Ndufs4-/- (KO) mice. By contrast, neurofilaments were significantly decreased in SN of KO mice. Furthermore, mass spectrometry and co-immunoprecipitation (Co-IP) analysis indicated an increase in ubiquitinated neurofilaments in midbrain of KO mice, whereas 20S proteasome activities were decreased, which could potentially explain the buildup of ubiquitin protein. Collectively, these data suggest that mitochondrial complex I defects cause proteasome inhibition, a consequent increase in ubiquitinated neurofilaments and other proteins, and decrease the expression of neurofilaments that could be relevant to the mechanism of DA neuronal death in PD.


Subject(s)
Electron Transport Complex I/metabolism , Intermediate Filaments/metabolism , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Ubiquitin/metabolism , Animals , Blotting, Western , Disease Models, Animal , Electron Transport Complex I/deficiency , Immunohistochemistry , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Knockout , Polymerase Chain Reaction
9.
Cardiovasc Res ; 103(2): 258-67, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24835278

ABSTRACT

AIMS: MicroRNAs (miRNAs) play critical roles during the development of the cardiovascular system. Blocking miRNA biosynthesis in embryonic hearts through a conditional gene inactivation approach led to differential cardiac defects depending on the Cre drivers used in different studies. The goal of this study is to reveal the cardiogenic pathway that is regulated by the miRNA mechanism at midgestation, a stage that has not been evaluated in previous publications. METHODS AND RESULTS: We specifically inactivated Dicer1, which is essential for generation of functional mature miRNAs, in the myocardium by crossing cTnt-Cre mice with Dicer1(loxP) mice. cTnt-Cre efficiently inactivates target genes in cardiomyocytes at midgestation. All mutants died between E14.5 and E16.5 with severe myocardial wall defects, including reduced cell proliferation, increased cell death, and spongy myocardial wall. Expression of TGFß type I receptor (Tgfbr1), which encodes the Type I receptor of TGFß ligands, was up-regulated in mutant hearts. As expected, TGFß activity was increased in Dicer1-inactivated hearts. Our further molecular analysis suggested that Tgfbr1 is a direct target of three miRNAs. Reducing TGFß activities using a pharmacological inhibitor on in vitro cultured hearts, or through an in vivo genetic approach, partially rescued the cardiac defects caused by Dicer1 inactivation. CONCLUSIONS: We show for the first time that TGFß signalling is directly regulated by the miRNA mechanism during myocardial wall morphogenesis. Increased TGFß activity plays a major role in the cardiac defects caused by myocardial deletion of Dicer1. Thus, miRNA-mediated regulation of TGFß signalling is indispensable for normal cardiogenesis.


Subject(s)
DEAD-box RNA Helicases/metabolism , Heart/embryology , MicroRNAs/metabolism , Myocardium/metabolism , Organogenesis/physiology , Ribonuclease III/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Myocytes, Cardiac/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology
10.
Hum Mol Genet ; 21(23): 5147-58, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22949510

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder in the developed world, and is characterized by the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Somatic mitochondrial DNA (mtDNA) deletions reach their highest concentration with age in the SN in humans, and may contribute to PD; yet whether mtDNA deletions cause DA neuron degeneration remains unclear. Inherited mutations of Twinkle helicase involved in mtDNA replication causes a dominant increase in mtDNA deletions in humans. We constructed a mouse model expressing mutant Twinkle in DA neurons. Mutant mice had an increase in age-related mtDNA deletions, reduction of DA neuron number in SN at 17-22 months and displayed abnormalities in rota-rod behavior. Functional analysis of midbrain indicated a slight reduction in mitochondrial state II respiration in mutants, but no decrease in maximal respiration. Also, Parkin expression was significantly decreased in DA neurons in the SN of 22-month-old mutant mice, and in PC12 cells after 48 h transfection of mutant Twinkle. Both confocal imaging and coimmunoprecipitation indicated interaction of Twinkle with Parkin in the mitochondria. Parkin overexpression rescued the reduction of proteasome activity caused by mutant Twinkle in PC12 cells. In addition, the autophagy marker LC3 was increased in the SN of 22-month transgenics, and this increase was similarly mutant Twinkle-dependent in PC12 cells. Collectively, our data demonstrate that mammalian Twinkle is important for mitochondrial integrity in DA neurons and provide a novel mouse model in which increased mtDNA deletions may lead to DA neuron degeneration and parkinsonism.


Subject(s)
DNA Helicases/genetics , DNA, Mitochondrial/genetics , Dopaminergic Neurons/metabolism , Mitochondrial Proteins/genetics , Mutation , Parkinson Disease/genetics , Animals , Autophagy/genetics , Behavior, Animal , Cell Line , Cell Respiration/genetics , DNA Helicases/metabolism , Dopaminergic Neurons/pathology , Gene Expression , Gene Order , Gene Targeting , Humans , Mice , Mice, Transgenic , Mitochondrial Proteins/metabolism , Parkinson Disease/metabolism , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Substantia Nigra/metabolism , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
11.
Dev Biol ; 349(2): 250-60, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21070765

ABSTRACT

The canonical Wnt/ß-catenin signaling pathway has implications in early facial development; yet, its function and signaling mechanism remain poorly understood. We report here that the frontonasal and upper jaw primordia cannot be formed after conditional ablation of ß-catenin with Foxg1-Cre mice in the facial ectoderm and the adjacent telencephalic neuroepithelium. Gene expression of several cell-survival and patterning factors, including Fgf8, Fgf3, and Fgf17, is dramatically diminished in the anterior neural ridge (ANR, a rostral signaling center) and/or the adjacent frontonasal ectoderm of the ß-catenin conditional mutant mice. In addition, Shh expression is diminished in the ventral telencephalon of the mutants, while Tcfap2a expression is less affected in the facial primordia. Apoptosis occurs robustly in the rostral head tissues following inactivation of Fgf signaling in the conditional mutants. Consequently, the upper jaw, nasal, ocular and telencephalic structures are absent, but the tongue and mandible are relatively developed in the conditional mutants at birth. Using molecular biological approaches, we demonstrate that the Fgf8 gene is transcriptionally targeted by Wnt/ß-catenin signaling during early facial and forebrain development. Furthermore, we show that conditional gain-of-function of ß-catenin signaling causes drastic upregulation of Fgf8 mRNA in the ANR and the entire facial ectoderm, which also arrests facial and forebrain development. Taken together, our results suggest that canonical Wnt/ß-catenin signaling is required for early development of the mammalian face and related head structures, which mainly or partly acts through the initiation and modulation of balanced Fgf signaling activity.


Subject(s)
Facial Bones/embryology , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Ablation Techniques , Animals , Apoptosis/physiology , Chromatin Immunoprecipitation , DNA Primers/genetics , Ectoderm/metabolism , Fluorescent Antibody Technique , Forkhead Transcription Factors/genetics , Hedgehog Proteins/metabolism , In Situ Hybridization , In Situ Nick-End Labeling , Luciferases , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Telencephalon/cytology , Telencephalon/metabolism , beta Catenin/genetics
12.
Dev Dyn ; 239(1): 200-10, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19705442

ABSTRACT

The role of a key Wnt coreceptor Lrp6 during heart development remains unclear. Here we show that ablation of Lrp6 in mice causes conotruncal anomalies including double-outlet right ventricle (DORV), outflow tract (OFT) cushion hypoplasia, and ventricular septal defect (VSD). Cardiac neural crest cells are specifically lost in the dorsal neural tube and caudal pharyngeal arches of the mutant embryos. We also demonstrate that Lrp6 is required for proliferation and survival of cardiac progenitors and for the expression of Isl1 in the secondary heart field. Other known cardiogenic regulators such as Msx1, Msx2, and Fgf8 are also significantly diminished in the mutant pharyngeal arches and/or OFT. Unexpectedly, the myocardium differentiation factors Mef2c and Myocardin are upregulated in the mutant OFT. Our results indicate that Lrp6 is essential for cardiac neural crest and OFT development upstream of multiple important cardiogenic genes in different cardiac lineage cells during early cardiogenesis.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Heart Defects, Congenital/genetics , Heart/embryology , LDL-Receptor Related Proteins/metabolism , Neural Crest/physiology , Signal Transduction/physiology , Animals , Branchial Region/cytology , Branchial Region/embryology , Bromodeoxyuridine , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , LDL-Receptor Related Proteins/genetics , LIM-Homeodomain Proteins , Low Density Lipoprotein Receptor-Related Protein-6 , MEF2 Transcription Factors , Mice , Mice, Mutant Strains , Myogenic Regulatory Factors/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors
13.
Dev Dyn ; 239(1): 318-26, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19653321

ABSTRACT

Lrp6 is a key coreceptor in the canonical Wnt pathway that is widely involved in tissue/organ morphogenesis. We generated a loxP-floxed Lrp6 mouse line. Crossing with a general Cre deleter, we obtained the Lrp6-floxdel mice, in which the loxP-floxed exon 2 of Lrp6 gene has been deleted ubiquitously. The homozygotes of Lrp6-floxdel mice reproduced typical defects as seen in the conventional Lrp6-deficient mice, such as defects in eye, limb, and neural tube, and die around birth. We also found new phenotypes including cleft palate and agenesis of external genitalia in the Lrp6-floxdel mice. In addition, the Lrp6-deficient embryos are known to be defective in other systems and internal organs including the heart and brain. Thus, by selectively crossing with a lineage-specific or inducible Cre mouse line, the Lrp6 conditional gene-targeting mice will allow us to model specific types of birth defects for mechanism and prevention studies.


Subject(s)
Congenital Abnormalities/genetics , Disease Models, Animal , LDL-Receptor Related Proteins/deficiency , Animals , Blotting, Northern , Crosses, Genetic , DNA Primers/genetics , Electroporation , Exons/genetics , Gene Targeting , Immunoblotting , LDL-Receptor Related Proteins/genetics , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Microinjections , Reverse Transcriptase Polymerase Chain Reaction
14.
Development ; 136(18): 3161-71, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19700620

ABSTRACT

Neither the mechanisms that govern lip morphogenesis nor the cause of cleft lip are well understood. We report that genetic inactivation of Lrp6, a co-receptor of the Wnt/beta-catenin signaling pathway, leads to cleft lip with cleft palate. The activity of a Wnt signaling reporter is blocked in the orofacial primordia by Lrp6 deletion in mice. The morphological dynamic that is required for normal lip formation and fusion is disrupted in these mutants. The expression of the homeobox genes Msx1 and Msx2 is dramatically reduced in the mutants, which prevents the outgrowth of orofacial primordia, especially in the fusion site. We further demonstrate that Msx1 and Msx2 (but not their potential regulator Bmp4) are the downstream targets of the Wnt/beta-catenin signaling pathway during lip formation and fusion. By contrast, a ;fusion-resistant' gene, Raldh3 (also known as Aldh1a3), that encodes a retinoic acid-synthesizing enzyme is ectopically expressed in the upper lip primordia of Lrp6-deficient embryos, indicating a region-specific role of the Wnt/beta-catenin signaling pathway in repressing retinoic acid signaling. Thus, the Lrp6-mediated Wnt signaling pathway is required for lip development by orchestrating two distinctively different morphogenetic movements.


Subject(s)
LDL-Receptor Related Proteins/metabolism , Lip/embryology , Morphogenesis/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Apoptosis/physiology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Cell Proliferation , Cleft Lip/metabolism , Cleft Lip/pathology , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Lip/anatomy & histology , Lip/metabolism , Low Density Lipoprotein Receptor-Related Protein-6 , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , Retinal Dehydrogenase , Wnt Proteins/genetics
15.
Dev Dyn ; 237(12): 3681-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18985738

ABSTRACT

Coloboma, an ocular birth defect seen in humans and other species, is caused by incomplete closure of the optic fissure. Here, we demonstrate that genetic deletion of Lrp6, a bottleneck coreceptor in the canonical Wnt signaling pathway, results in ocular coloboma and neuroretinal patterning defects in mice. The expression of ventral neuroretinal patterning gene Vax2 was conserved but with dorsally shifted expression domains; however, the dorsal neuroretinal patterning gene Tbx5 was lost in the Lrp6-mutant eyes at embryonic day 10.5. Both Bmp4 and phosphorylated Smad 1/5/8 were also significantly attenuated in the dorsal neuroretina. In addition, the retinoic acid synthesizing enzymes Raldh1 and Raldh3 were significantly changed in the mutant eyes. Our findings suggest that defective retinal patterning causes coloboma in the Lrp6-deficient mice, and that canonical Wnt signaling plays a primary role in dorsal neuroretinal patterning and related morphogenetic movements by regulation of both Bmp and retinoic acid signaling pathways.


Subject(s)
Body Patterning , Coloboma/embryology , Coloboma/metabolism , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Neurons/metabolism , Animals , Bone Morphogenetic Protein 4/metabolism , Coloboma/genetics , Coloboma/pathology , Down-Regulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Low Density Lipoprotein Receptor-Related Protein-6 , Mice , Mice, Knockout , Mutation/genetics , Signal Transduction , Tretinoin/metabolism , Wnt Proteins/metabolism
16.
Dev Dyn ; 237(11): 3157-68, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18816448

ABSTRACT

Wnt reporter TOPgal mice carry a beta-galactosidase (betagal) gene under the control of the Wnt/beta-catenin signaling responsive elements. We found that the intensely immunolabeled betagal+ cells were co-immunolabeled with Nestin and formed a tangentially oriented single-cell layer in the "connecting or docking zone" where the olfactory sensory axons attached to the brain surface during mid-gestation. During early postnatal development, betagal+ cells were located in the inner olfactory nerve layer (ONLi) and co-labeled with olfactory ensheathing cell (OEC) markers S100beta and NPY but not with lineage-specific markers for neurons, oligodendrocytes, astrocytes, and microglia, demonstrating that the TOPgal marked a subpopulation of OECs. By confocal microscopy, we found that TOPgal activated processes extended along the developing glomerulus and formed multiple tunnel-like structures that ensheathe and bridge olfactory sensory axonal bundles from ONLi to the glomerulus, which may play a key role in glomerulus formation and convergent sorting of the peripheral olfactory axons.


Subject(s)
Axons/metabolism , Olfactory Nerve/cytology , Olfactory Nerve/embryology , Response Elements/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Intermediate Filament Proteins/biosynthesis , Mice , Mice, Transgenic , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Nerve Tissue Proteins/biosynthesis , Nestin , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , S100 Calcium Binding Protein beta Subunit , S100 Proteins/biosynthesis , S100 Proteins/genetics , Wnt Proteins/genetics , beta Catenin/genetics
17.
Circ Res ; 101(3): 277-85, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17585069

ABSTRACT

Congenital heart diseases are the most commonly observed human birth defects and are the leading cause of infant morbidity and mortality. Accumulating evidence indicates that transforming growth factor-beta/bone morphogenetic protein signaling pathways play critical roles during cardiogenesis. Smad4 encodes the only common Smad protein in mammals, which is a critical nuclear mediator of transforming growth factor-beta/bone morphogenetic protein signaling. The aim of this work was to investigate the roles of Smad4 during heart development. To overcome the early embryonic lethality of Smad4(-/-) mice, we specifically disrupted Smad4 in the myocardium using a Cre/loxP system. We show that myocardial-specific inactivation of Smad4 caused heart failure and embryonic lethality at midgestation. Histological analysis revealed that mutant mice displayed a hypocellular myocardial wall defect, which is likely the primary cause for heart failure. Both decreased cell proliferation and increased apoptosis contributed to the myocardial wall defect in mutant mice. Data presented in this article contradict a previous report showing that Smad4 is dispensable for heart development. Our further molecular characterization showed that expression of Nmyc and its downstream targets, including cyclin D1, cyclin D2, and Id2, were downregulated in mutant embryos. Reporter analysis indicated that the transcriptional activity of the 351-bp Nmyc promoter can be positively regulated by bone morphogenetic protein stimulation and negatively regulated by transforming growth factor-beta stimulation. Chromatin immunoprecipitation analysis revealed that the Nmyc promoter can form a complex with Smad4, suggesting that Nmyc is a direct downstream target of Smad4. In conclusion, this study provides the first mouse model showing that Smad4 plays essential roles during cardiogenesis.


Subject(s)
Fetal Heart/growth & development , Gene Expression Regulation, Developmental , Heart/embryology , Myocardium/metabolism , Smad4 Protein/physiology , Amino Acid Sequence , Animals , Apoptosis , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Cell Division , Female , Fetal Diseases/etiology , Genes, Lethal , Genes, myc , Heart Failure/embryology , Heart Failure/etiology , Heart Failure/pathology , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutagenesis, Site-Directed , Myocardium/pathology , Myocytes, Cardiac/pathology , NIH 3T3 Cells , Organogenesis , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/physiology , Sequence Homology, Amino Acid , Smad4 Protein/deficiency , Smad4 Protein/genetics , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
18.
Dev Biol ; 301(1): 276-86, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16959237

ABSTRACT

Accumulated evidence has suggested that BMP pathways play critical roles during mammalian cardiogenesis and impairment of BMP signaling may contribute to human congenital heart diseases (CHDs), which are the leading cause of infant morbidity and mortality. Alk3 encodes a BMP specific type I receptor expressed in mouse embryonic hearts. To reveal functions of Alk3 during atrioventricular (AV) cushion morphogenesis and to overcome the early lethality of Alk3(-/-) embryos, we applied a Cre/loxp approach to specifically inactivate Alk3 in the endothelium/endocardium. Our studies showed that endocardial depletion of Alk3 severely impairs epithelium-mesenchymal-transformation (EMT) in the atrioventricular canal (AVC) region; the number of mesenchymal cells formed in Tie1-Cre;Alk3(loxp/loxp) embryos was reduced to only approximately 20% of the normal level from both in vivo section studies and in vitro explant assays. We showed, for the first time, that in addition to its functions on mesenchyme formation, Alk3 is also required for the normal growth/survival of AV cushion mesenchymal cells. Functions of Alk3 are accomplished through regulating expression/activation/subcellular localization of multiple downstream genes including Smads and cell-cycle regulators. Taken together, our study supports the notion that Alk3-mediated BMP signaling in AV endocardial/mesenchymal cells plays a central role during cushion morphogenesis.


Subject(s)
Atrioventricular Node/embryology , Bone Morphogenetic Protein Receptors, Type I/physiology , Heart/embryology , Animals , Atrioventricular Node/cytology , Bone Morphogenetic Protein Receptors, Type I/genetics , Cell Proliferation , Cell Survival , Female , Fluorescent Antibody Technique , Genes, Lethal , Male , Mice , Morphogenesis
19.
Tumour Biol ; 26(6): 306-12, 2005.
Article in English | MEDLINE | ID: mdl-16254460

ABSTRACT

Neck lymphatic metastasis represents the single most important clinical prognostic factor in nasopharyngeal carcinoma (NPC), but underlying genetic mechanisms remain ill defined. In this study 23 samples of primary tumor (PT) and 9 of neck lymph node metastasis (NLNM) obtained from NPC patients were analyzed by comparative genomic hybridization (CGH) coupled with tissue microdissection and degenerate oligonucleotide primer-polymerase chain reaction (DOP-PCR). A similar pattern of chromosomal abnormalities was seen in PT and NLNM, the common aberrations were gains on 5p, 12p, 12q and 18p and deletions on 1p, 3p, 9q, 14q, 17p and 16q. However, NLNMs, but not PTs, also exhibited frequent losses on 9p, 16p, 17q, 20q, 21p, 21q and 22q and gains on 8q and 8p. The most frequent unique aberration in NLNMs was loss on 16p, observed in 100% (9/9) NLNMs tested, as well as loss of 20q, observed in 77.8% of tumors tested. For the first time, we report that a gain on 8p and a loss at 20q is common to NLNMs. The analysis furthermore suggests that specific alterations, e.g. losses of 9p, 16p, 7q, 20q, 21p, 21q, 22q and gains on 8q and 8p are associated with NLNM of NPC, and that these alterations may be involved in the onset and/or progression of a metastatic phenotype.


Subject(s)
Chromosome Aberrations , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/secondary , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Humans
20.
Tumour Biol ; 26(3): 158-64, 2005.
Article in English | MEDLINE | ID: mdl-15970651

ABSTRACT

OBJECTIVE: The genetic mechanisms underlying the development and progression of lung squamous cell carcinoma (SCC), the major subtype of non-small cell lung cancer, are still unknown. To better understand this disease, we studied the association between genetic alterations and the progression of lung SCC. METHODS: Chromosomal aberrations in 39 samples of lung SCC, including 21 nonmetastatic and 18 metastatic carcinomas, were characterized by comparative genomic hybridization and statistically correlated to clinical staging and metastatic ability. RESULTS: The average gains and losses per patient were significantly higher in the advanced-stage lung SCC and metastatic SCC group compared to the early-stage lung SCC and nonmetastatic SCC group. Gains of 2p, 20p and losses of 2q, 4q, 5q, 9q, 13p, 18q correlated with advanced-stage lung SCC. Losses on 2q, 4q, 6p, 16p, 16q, 18q, 20q, 21q and gains on 2p, 7p, 7q, 20p were more frequent in the metastatic SCC group, which was significantly different from the nonmetastatic SCC group. Gains on 2p, 20p and losses on 2q, 4q, 18q were not only associated with an advanced clinical stage but also with metastases of lung SCC. CONCLUSIONS: The results suggest that several chromosomal aberrations (e.g. gains on 2p, 20p and losses on 2q, 4q, 18q) may contribute to the progression of lung SCC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Chromosome Aberrations , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Adult , Aged , Disease Progression , Female , Humans , Male , Middle Aged , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL
...