Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Development ; 147(10)2020 05 21.
Article in English | MEDLINE | ID: mdl-32345746

ABSTRACT

Synapses exhibit an astonishing degree of adaptive plasticity in healthy and disease states. We have investigated whether synapses also adjust to life stages imposed by novel developmental programs for which they were never molded by evolution. Under conditions in which Drosophila larvae are terminally arrested, we have characterized synaptic growth, structure and function at the neuromuscular junction (NMJ). Although wild-type larvae transition to pupae after 5 days, arrested third instar (ATI) larvae persist for 35 days, during which time NMJs exhibit extensive overgrowth in muscle size, presynaptic release sites and postsynaptic glutamate receptors. Remarkably, despite this exuberant growth, stable neurotransmission is maintained throughout the ATI lifespan through a potent homeostatic reduction in presynaptic neurotransmitter release. Arrest of the larval stage in stathmin mutants also reveals a degree of progressive instability and neurodegeneration that was not apparent during the typical larval period. Hence, an adaptive form of presynaptic depression stabilizes neurotransmission during an extended developmental period of unconstrained synaptic growth. More generally, the ATI manipulation provides a powerful system for studying neurodegeneration and plasticity across prolonged developmental timescales.


Subject(s)
Drosophila/growth & development , Drosophila/genetics , Larva/growth & development , Larva/genetics , Long-Term Synaptic Depression/genetics , Nerve Degeneration/genetics , Neuromuscular Junction/growth & development , Animals , Axons/pathology , Drosophila Proteins/genetics , Female , Homeostasis/genetics , Male , Mutation , Neuromuscular Junction/metabolism , RNA Interference , Smad Proteins, Receptor-Regulated/genetics , Stathmin/genetics , Synapses/metabolism , Synaptic Transmission/genetics
2.
Mol Cell ; 53(6): 893-903, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24613342

ABSTRACT

Ubiquitination of transcription activators has been reported to regulate transcription via both proteolytic and nonproteolytic routes, yet the function of the ubiquitin (Ub) signal in the nonproteolytic process is poorly understood. By use of the heterologous transcription activator LexA-VP16 in Saccharomyces cerevisiae, we show that monoubiquitin fusion of the activator prevents stable interactions between the activator and DNA, leading to transcription inhibition without activator degradation. We identify the AAA(+) ATPase Cdc48 and its cofactors as the Ub receptor responsible for extracting the monoubiquitinated activator from DNA. Our results suggest that deubiquitination of the activator is critical for transcription activation. These findings with LexA-VP16 extend in both yeast and mammalian cells to native transcription activators Met4 and R-Smads, respectively, that are known to be oligo-ubiquitinated. The results illustrate a role for Ub and Cdc48 in transcriptional regulation and gene expression that is independent of proteolysis.


Subject(s)
Adenosine Triphosphatases/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Cell Cycle Proteins/genetics , Nuclear Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/genetics , Smad Proteins, Receptor-Regulated/genetics , Transcriptional Activation , Ubiquitin/genetics , Adenosine Triphosphatases/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Binding Sites , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA/genetics , DNA/metabolism , Humans , Nuclear Proteins/metabolism , Protein Binding , Proteolysis , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Transcription, Genetic , Ubiquitin/metabolism , Ubiquitination , Valosin Containing Protein
3.
Evol Dev ; 21(1): 44-55, 2019 01.
Article in English | MEDLINE | ID: mdl-30588766

ABSTRACT

Members of the phylum Arthropoda, comprising over 80% of total animal species, have evolved regenerative abilities, but little is known about the molecular mechanisms mediating this process. Transforming growth factor ß (TGF-ß) signaling mediates a diverse set of essential processes in animals and is a good candidate pathway for regulation of regeneration in arthropods. In this study we investigated the role of activin signaling, a TGF-ß superfamily pathway, in limb regeneration in the crayfish. We identified and cloned a downstream transcription factor in the activin pathway, Smox, and characterized its function with regard to other elements of the activin signaling pathway. Gene knockdown of Smox by RNAi induced regeneration of complete but smaller pereopods after autotomy. This indicates that activin signaling via Smox functions in regulation of pereopod growth and size. The expression levels of both Smox and the activin receptor babo were closely correlated with molting. The expression level of Smox increased when babo was knocked down by RNAi, indicating that Smox and babo transcription are linked. Our study suggests that the Babo-Smox system in activin signaling is conserved in decapods, and supports an evolutionary conservation of this aspect of molecular signaling during regeneration between protostomes and deuterostomes.


Subject(s)
Astacoidea/physiology , Smad Proteins, Receptor-Regulated/metabolism , Animals , Cloning, Molecular , Extremities/physiology , Gene Knockdown Techniques , Regeneration , Smad Proteins, Receptor-Regulated/chemistry , Smad Proteins, Receptor-Regulated/genetics
4.
Apoptosis ; 24(11-12): 972-989, 2019 12.
Article in English | MEDLINE | ID: mdl-31641960

ABSTRACT

Developmentally regulated programmed cell death (PCD) is one of the key cellular events for precise controlling of neuronal population during postembryonic development of the central nervous system. Previously we have shown that a group of corazonin-producing peptidergic neurons (vCrz) undergo apoptosis in response to ecdysone signaling via ecdysone receptor (EcR)-B isoforms and Ultraspiracle during early phase of metamorphosis. Further utilizing genetic, transgenic, and mosaic analyses, we have found that TGF-ß signaling mediated by a glia-produced ligand, Myoglianin, type-I receptor Baboon (particularly Babo-A isoform) and dSmad2, is also required autonomously for PCD of the vCrz neurons. Our studies show that TGF-ß signaling is not acting epistatically to EcR or vice versa. We also show that ectopic expression of a constitutively active phosphomimetic form of dSmad2 (dSmad2PM) is capable of inducing premature death of vCrz neurons in larva but not other larval neurons. Intriguingly, the dSmad2PM-mediated killing is completely suppressed by coexpression of a dominant-negative form of EcR (EcRDN), suggesting that EcR function is required for the proapoptotic dSmad2PM function. Based on these data, we suggest that TGF-ß and ecdysone signaling pathways act cooperatively to induce vCrz neuronal PCD. We propose that this type of two-factor authentication is a key developmental strategy to ensure the timely PCD of specific larval neurons during metamorphosis.


Subject(s)
Activin Receptors/metabolism , Apoptosis , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Metamorphosis, Biological/genetics , Neurons/metabolism , Receptors, Steroid/metabolism , Activin Receptors/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Animals, Genetically Modified , Apoptosis/physiology , Central Nervous System/cytology , Central Nervous System/growth & development , Central Nervous System/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/metabolism , Ecdysone/physiology , Gene Expression Regulation, Developmental/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Larva/cytology , Larva/metabolism , Metamorphosis, Biological/physiology , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Isoforms/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Steroid/genetics , Signal Transduction/genetics , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/physiology
5.
Genes Dev ; 25(15): 1654-61, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21828274

ABSTRACT

Nodal signaling, mediated through SMAD transcription factors, is necessary for pluripotency maintenance and endoderm commitment. We identified a new motif, termed SMAD complex-associated (SCA), that is bound by SMAD2/3/4 and FOXH1 in human embryonic stem cells (hESCs) and derived endoderm. We demonstrate that two basic helix-loop-helix (bHLH) proteins-HEB and E2A-bind the SCA motif at regions overlapping SMAD2/3 and FOXH1. Furthermore, we show that HEB and E2A associate with SMAD2/3 and FOXH1, suggesting they form a complex at critical target regions. This association is biologically important, as E2A is critical for mesendoderm specification, gastrulation, and Nodal signal transduction in Xenopus tropicalis embryos. Taken together, E proteins are novel Nodal signaling cofactors that associate with SMAD2/3 and FOXH1 and are necessary for mesendoderm differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Smad Proteins, Receptor-Regulated/metabolism , Amino Acid Motifs , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Chromatin Immunoprecipitation , Embryonic Stem Cells , Endoderm/metabolism , Forkhead Transcription Factors/genetics , Gastrulation/genetics , Gene Expression Regulation, Developmental/genetics , High-Throughput Nucleotide Sequencing , Humans , Left-Right Determination Factors/metabolism , Protein Binding , Signal Transduction , Smad Proteins, Receptor-Regulated/chemistry , Smad Proteins, Receptor-Regulated/genetics , Xenopus/embryology
6.
Arch Toxicol ; 92(9): 2935-2946, 2018 09.
Article in English | MEDLINE | ID: mdl-30097701

ABSTRACT

MicroRNA (miRNA)-mediated gene regulation contributes to liver pathophysiology, including hepatic stellate cell (HSC) activation and fibrosis progression. Here, we investigated the role of miR-942 in human liver fibrosis. The expression of miR-942, HSC activation markers, transforming growth factor-beta pseudoreceptor BMP and activin membrane-bound inhibitor (BAMBI), as well as collagen deposition, were investigated in 100 liver specimens from patients with varying degree of hepatitis B virus (HBV)-related fibrosis. Human primary HSCs and the immortalized cell line (LX2 cells) were used for functional studies. We found that miR-942 expression was upregulated in activated HSCs and correlated inversely with BAMBI expression in liver fibrosis progression. Transforming growth factor beta (TGF-ß) and lipopolyssacharide (LPS), two major drivers of liver fibrosis and inflammation, induce miR-942 expression in HSCs via Smad2/3 respective NF-κB/p50 binding to the miR-942 promoter. Mechanistically, the induced miR-942 degrades BAMBI mRNA in HSCs, thereby sensitizing the cells for fibrogenic TGF-ß signaling and also partly mediates LPS-induced proinflammatory HSC fate. In conclusion, the TGF-ß and LPS-induced miR-942 mediates HSC activation through downregulation of BAMBI in human liver fibrosis. Our study provides new insights on the molecular mechanism of HSC activation and fibrosis.


Subject(s)
Hepatic Stellate Cells/pathology , Liver Cirrhosis/pathology , Membrane Proteins/genetics , MicroRNAs/metabolism , Cells, Cultured , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Lipopolysaccharides/pharmacology , Liver Cirrhosis/genetics , Membrane Proteins/metabolism , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta1/pharmacology
7.
Dev Biol ; 417(1): 104-13, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27452629

ABSTRACT

In contrast with Drosophila melanogaster, practically nothing is known about the involvement of the TGF-ß signaling pathway in the metamorphosis of hemimetabolan insects. To partially fill this gap, we have studied the role of Smad factors in the metamorphosis of the German cockroach, Blattella germanica. In D. melanogaster, Mad is the canonical R-Smad of the BMP branch of the TGF-ß signaling pathway, Smox is the canonical R-Smad of the TGF-ß/Activin branch and Medea participates in both branches. In insects, metamorphosis is regulated by the MEKRE93 pathway, which starts with juvenile hormone (JH), whose signal is transduced by Methoprene-tolerant (Met), which stimulates the expression of Krüppel homolog 1 (Kr-h1) that acts to repress E93, the metamorphosis trigger. In B. germanica, metamorphosis is determined at the beginning of the sixth (final) nymphal instar (N6), when JH production ceases, the expression of Kr-h1 declines, and the transcription of E93 begins to increase. The RNAi of Mad, Smox and Medea in N6 of B. germanica reveals that the BMP branch of the TGF-ß signaling pathway regulates adult ecdysis and wing extension, mainly through regulating the expression of bursicon, whereas the TGF-ß/Activin branch contributes to increasing E93 and decreasing Kr-h1 at the beginning of N6, crucial for triggering adult morphogenesis, as well as to regulating the imaginal molt timing.


Subject(s)
Cockroaches/embryology , Drosophila melanogaster/embryology , Metamorphosis, Biological/physiology , Molting/physiology , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Activins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Juvenile Hormones/metabolism , Kruppel-Like Transcription Factors/metabolism , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Reproduction ; 153(2): 221-231, 2017 02.
Article in English | MEDLINE | ID: mdl-27879343

ABSTRACT

The molecular mechanisms involved in regulating the development of small, gonadotrophin-independent follicles are poorly understood; however, many studies have highlighted an essential role for TGFB ligands. Canonical TGFB signalling is dependent upon intracellular SMAD proteins that regulate transcription. STRAP has been identified in other tissues as an inhibitor of the TGFB-SMAD signalling pathway. Therefore, in this study we aimed to determine the expression and role of STRAP in the context of early follicle development. Using qPCR, Strap, Smad3 and Smad7 revealed similar expression profiles in immature ovaries from mice aged 4-16 days containing different populations of early growing follicles. STRAP and SMAD2/3 proteins co-localised in granulosa cells of small follicles using immunofluorescence. Using an established culture model, neonatal mouse ovary fragments with a high density of small non-growing follicles were used to examine the effects of Strap knockdown using siRNA and STRAP protein inhibition by immuno-neutralisation. Both interventions caused a reduction in the proportion of small, non-growing follicles and an increase in the proportion and size of growing follicles in comparison to untreated controls, suggesting inhibition of STRAP facilitates follicle activation. Recombinant STRAP protein had no effect on small, non-growing follicles, but increased the mean oocyte size of growing follicles in the neonatal ovary model and also promoted the growth of isolated preantral follicles in vitro Overall findings indicate STRAP is expressed in the mouse ovary and is capable of regulating development of small follicles in a stage-dependent manner.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Ovarian Follicle/growth & development , Ovary/growth & development , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Animals , Female , Gene Knockdown Techniques , Granulosa Cells/chemistry , Mice , Mice, Inbred C57BL , Oocytes/drug effects , Oocytes/growth & development , Ovary/metabolism , RNA-Binding Proteins , Recombinant Proteins/pharmacology , Signal Transduction/physiology , Smad Proteins, Receptor-Regulated/genetics , Smad2 Protein/analysis , Smad3 Protein/analysis , Smad3 Protein/genetics , Transcriptome , Transforming Growth Factor beta/metabolism
9.
Arterioscler Thromb Vasc Biol ; 36(3): 553-60, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26769046

ABSTRACT

OBJECTIVE: Bone morphogenic protein 4 (BMP4) is an important mediator of endothelial dysfunction in cardio-metabolic diseases, whereas platelet-derived growth factors (PDGFs) are major angiogenic and proinflammatory mediator, although the functional link between these 2 factors is unknown. The present study investigated whether PDGF mediates BMP4-induced endothelial dysfunction in diabetes mellitus. APPROACH AND RESULTS: We generated Ad-Bmp4 to overexpress Bmp4 and Ad-Pdgfa-shRNA to knockdown Pdgfa in mice through tail intravenous injection. SMAD4-shRNA lentivirus, SMAD1-shRNA, and SMAD5 shRNA adenovirus were used for knockdown in human and mouse endothelial cells. We found that PDGF-AA impaired endothelium-dependent vasodilation in aortas and mesenteric resistance arteries. BMP4 upregulated PDGF-AA in human and mouse endothelial cells, which was abolished by BMP4 antagonist noggin or knockdown of SMAD1/5 or SMAD4. BMP4-impared relaxation in mouse aorta was also ameliorated by PDGF-AA neutralizing antibody. Tail injection of Ad-Pdgfa-shRNA ameliorates endothelial dysfunction induced by Bmp4 overexpression (Ad-Bmp4) in vivo. Serum PDGF-AA was elevated in both diabetic patients and diabetic db/db mice compared with nondiabetic controls. Pdgfa-shRNA or Bmp4-shRNA adenovirus reduced serum PDGF-AA concentration in db/db mice. PDGF-AA neutralizing antibody or tail injection with Pdgfa-shRNA adenovirus improved endothelial function in aortas and mesenteric resistance arteries from db/db mice. The effect of PDGF-AA on endothelial function in mouse aorta was also inhibited by Ad-Pdgfra-shRNA to inhibit PDGFRα. CONCLUSIONS: The present study provides novel evidences to show that PDGF-AA impairs endothelium-dependent vasodilation and PDGF-AA mediates BMP4-induced adverse effect on endothelial cell function through SMAD1/5- and SMAD4-dependent mechanisms. Inhibition of PGDF-AA ameliorates vascular dysfunction in diabetic mice.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Diabetes Mellitus/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Platelet-Derived Growth Factor/metabolism , Smad Proteins, Receptor-Regulated/metabolism , Vasodilation , Adult , Aged , Animals , Antibodies, Neutralizing/pharmacology , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/pharmacology , Case-Control Studies , Cells, Cultured , Diabetes Mellitus/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/pharmacology , RNA Interference , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Smad Proteins, Receptor-Regulated/genetics , Time Factors , Tissue Culture Techniques , Transfection , Up-Regulation , Vasodilation/drug effects , Vasodilator Agents/pharmacology
10.
Hum Mol Genet ; 23(8): 2145-56, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24293546

ABSTRACT

Haploinsufficiency for CHD7, an ATP-dependent nucleosome remodeling factor, is the leading cause of CHARGE syndrome. While congenital heart defects (CHDs) are major clinical features of CHARGE syndrome, affecting >75% of patients, it remains unclear whether CHD7 can directly regulate cardiogenic genes in embryos. Our complementary yeast two-hybrid and biochemical assays reveal that CHD7 is a novel interaction partner of canonical BMP signaling pathway nuclear mediators, SMAD1/5/8, in the embryonic heart. Moreover, CHD7 associates in a BMP-dependent manner with the enhancers of a critical cardiac transcription factor, Nkx2.5, that contain functional SMAD1-binding elements. Both the active epigenetic signature of Nkx2.5 regulatory elements and its proper expression in cardiomyocytes require CHD7. Finally, inactivation of Chd7 in mice impairs multiple BMP signaling-regulated cardiogenic processes. Our results thus support the model that CHD7 is recruited by SMAD1/5/8 to the enhancers of BMP-targeted cardiogenic genes to epigenetically regulate their expression. Impaired BMP activities in embryonic hearts may thus have a major contribution to CHDs in CHARGE syndrome.


Subject(s)
Bone Morphogenetic Protein 1/metabolism , DNA-Binding Proteins/physiology , Embryo, Mammalian/metabolism , Epigenomics , Heart/embryology , Homeodomain Proteins/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transcription Factors/genetics , Animals , Blotting, Western , Bone Morphogenetic Protein 1/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Immunoenzyme Techniques , Immunoprecipitation , Mice , Organogenesis/physiology , Protein Binding , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Regulatory Elements, Transcriptional , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Smad Proteins, Receptor-Regulated/genetics , Transcription Factors/metabolism , Transcription, Genetic , Two-Hybrid System Techniques
11.
Arterioscler Thromb Vasc Biol ; 35(12): 2626-37, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26471266

ABSTRACT

OBJECTIVE: Bone morphogenetic protein-9 (BMP9)/activin-like kinase-1 and delta-like 4 (DLL4)/Notch promote endothelial quiescence, and we aim to understand mechanistic interactions between the 2 pathways. We identify new targets that contribute to endothelial quiescence and test whether loss of Dll4(+/-) in adult vasculature alters BMP signaling. APPROACH AND RESULTS: Human endothelial cells respond synergistically to BMP9 and DLL4 stimulation, showing complete quiescence and induction of HEY1 and HEY2. Canonical BMP9 signaling via activin-like kinase-1-Smad1/5/9 was disrupted by inhibition of Notch signaling, even in the absence of exogenous DLL4. Similarly, DLL4 activity was suppressed when the basal activin-like kinase-1-Smad1/5/9 pathway was inhibited, showing that these pathways are interdependent. BMP9/DLL4 required induction of P27(KIP1) for quiescence, although multiple factors are involved. To understand these mechanisms, we used proteomics data to identify upregulation of thrombospondin-1, which contributes to the quiescence phenotype. To test whether Dll4 regulates BMP/Smad pathways and endothelial cell phenotype in vivo, we characterized the vasculature of Dll4(+/-) mice, analyzing endothelial cells in the lung, heart, and aorta. Together with changes in endothelial structure and vascular morphogenesis, we found that loss of Dll4 was associated with a significant upregulation of pSmad1/5/9 signaling in lung endothelial cells. Because steady-state endothelial cell proliferation rates were not different in the Dll4(+/-) mice, we propose that the upregulation of pSmad1/5/9 signaling compensates to maintain endothelial cell quiescence in these mice. CONCLUSIONS: DLL4/Notch and BMP9/activin-like kinase-1 signaling rely on each other's pathways for full activity. This represents an important mechanism of cross talk that enhances endothelial quiescence and sensitively coordinates cellular responsiveness to soluble and cell-tethered ligands.


Subject(s)
Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Endothelial Cells/metabolism , Growth Differentiation Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptor, Notch1/metabolism , Thrombospondin 1/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Adaptor Proteins, Signal Transducing , Animals , Aorta/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cells, Cultured , Coronary Vessels/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Genotype , Growth Differentiation Factor 2 , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lung/blood supply , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA Interference , Receptor, Notch1/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Thrombospondin 1/genetics , Transfection
12.
J Biol Chem ; 289(28): 19571-84, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24841199

ABSTRACT

Nucleotide-binding domain and leucine-rich repeat containing PYD-3 (NLRP3) is a pattern recognition receptor that is implicated in the pathogenesis of inflammation and chronic diseases. Although much is known regarding the NLRP3 inflammasome that regulates proinflammatory cytokine production in innate immune cells, the role of NLRP3 in non-professional immune cells is unclear. Here we report that NLRP3 is expressed in cardiac fibroblasts and increased during TGFß stimulation. NLRP3-deficient cardiac fibroblasts displayed impaired differentiation and R-Smad activation in response to TGFß. Only the central nucleotide binding domain of NLRP3 was required to augment R-Smad signaling because the N-terminal Pyrin or C-terminal leucine-rich repeat domains were dispensable. Interestingly, NLRP3 regulation of myofibroblast differentiation proceeded independently from the inflammasome, IL-1ß/IL-18, or caspase 1. Instead, mitochondrially localized NLRP3 potentiated reactive oxygen species to augment R-Smad activation. In vivo, NLRP3-deficient mice were protected against angiotensin II-induced cardiac fibrosis with preserved cardiac architecture and reduced collagen 1. Together, these results support a distinct role for NLRP3 in non-professional immune cells independent from the inflammasome to regulate differential aspects of wound healing and chronic disease.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myofibroblasts/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Carrier Proteins/genetics , Collagen Type I/biosynthesis , Collagen Type I/genetics , Fibrosis , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Myocardium/pathology , Myofibroblasts/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Smad Proteins, Receptor-Regulated/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vasoconstrictor Agents/adverse effects , Vasoconstrictor Agents/pharmacology
13.
Development ; 139(2): 259-68, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22159578

ABSTRACT

BMP activity is essential for many steps of neural development, including the initial role in neural induction and the control of progenitor identities along the dorsal-ventral axis of the neural tube. Taking advantage of chick in ovo electroporation, we show a novel role for BMP7 at the time of neurogenesis initiation in the spinal cord. Using in vivo loss-of-function experiments, we show that BMP7 activity is required for the generation of three discrete subpopulations of dorsal interneurons: dI1-dI3-dI5. Analysis of the BMP7 mouse mutant shows the conservation of this activity in mammals. Furthermore, this BMP7 activity appears to be mediated by the canonical Smad pathway, as we demonstrate that Smad1 and Smad5 activities are similarly required for the generation of dI1-dI3-dI5. Moreover, we show that this role is independent of the patterned expression of progenitor proteins in the dorsal spinal cord, but depends on the BMP/Smad regulation of specific proneural proteins, thus narrowing this BMP7 activity to the time of neurogenesis. Together, these data establish a novel role for BMP7 in primary neurogenesis, the process by which a neural progenitor exits the cell cycle and enters the terminal differentiation pathway.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Interneurons/physiology , Neurogenesis/physiology , Signal Transduction/physiology , Smad Proteins, Receptor-Regulated/metabolism , Spinal Cord/embryology , Analysis of Variance , Animals , Chick Embryo , Immunohistochemistry , In Situ Hybridization , Interneurons/metabolism , Luciferases , Mice , Mutation/genetics , Neurogenesis/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Smad Proteins, Receptor-Regulated/genetics
14.
Mol Hum Reprod ; 20(5): 373-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24413384

ABSTRACT

In the ovary, connexin-coupled gap junctions in granulosa cells play crucial roles in follicular and oocyte development as well as in corpus luteum formation. Our previous work has shown that theca cell-derived bone morphogenetic protein (BMP)4 and BMP7 decrease gap junction intercellular communication (GJIC) activity via the down-regulation of connexin43 (Cx43) expression in immortalized human granulosa cells. However, the effects of oocyte-derived growth factors on Cx43 expression remain to be elucidated. The present study was designed to investigate the effects of oocyte-derived growth differentiation factor (GDF)9 and BMP15 on the expression of Cx43 in a human granulosa cell line, SVOG. We also examined the effect relative to GJIC activity and investigated the potential mechanisms of action. In SVOG cells, treatment with BMP15 but not GDF9 significantly decreased Cx43 mRNA and protein levels and GJIC activity. These suppressive effects, along with the induction of Smad1/5/8 phosphorylation, were attenuated by co-treatment with a BMP type I receptor inhibitor, dorsomorphin. Furthermore, knockdown of the central component of the transforming growth factor-ß superfamily signaling pathway, Smad4, using small interfering RNA reversed the suppressive effects of BMP15 on Cx43 expression and GJIC activity. The suppressive effects of BMP15 on Cx43 expression were further confirmed in primary human granulosa-lutein cells obtained from infertile patients undergoing an in vitro fertilization procedure. These findings suggest that oocyte-derived BMP15 decreases GJIC activity between human granulosa cells by down-regulating Cx43 expression, most likely via a Smad-dependent signaling pathway.


Subject(s)
Bone Morphogenetic Protein 15/metabolism , Connexin 43/metabolism , Gap Junctions/metabolism , Granulosa Cells/metabolism , Growth Differentiation Factor 9/metabolism , Oocytes/metabolism , Paracrine Communication , Cell Line , Connexin 43/genetics , Down-Regulation , Female , Gap Junctions/drug effects , Granulosa Cells/drug effects , Humans , Oocytes/drug effects , Paracrine Communication/drug effects , Phosphorylation , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Time Factors
15.
Dev Biol ; 367(2): 216-27, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22609550

ABSTRACT

Bone Morphogenetic Proteins (BMPs) have multiple activities in the developing spinal cord: they specify the identity of the dorsal-most neuronal populations and then direct the trajectories of dorsal interneuron (dI) 1 commissural axons. How are these activities decoded by dorsal neurons to result in different cellular outcomes? Our previous studies have shown that the diverse functions of the BMPs are mediated by the canonical family of BMP receptors and then regulated by specific inhibitory (I) Smads, which block the activity of a complex of Smad second messengers. However, the extent to which this complex translates the different activities of the BMPs in the spinal cord has remained unresolved. Here, we demonstrate that the receptor-activated (R) Smads, Smad1 and Smad5 play distinct roles mediating the abilities of the BMPs to direct cell fate specification and axon outgrowth. Smad1 and Smad5 occupy spatially distinct compartments within the spinal cord, with Smad5 primarily associated with neural progenitors and Smad1 with differentiated neurons. Consistent with this expression profile, loss of function experiments in mouse embryos reveal that Smad5 is required for the acquisition of dorsal spinal neuron identities whereas Smad1 is critical for the regulation of dI1 axon outgrowth. Thus the R-Smads, like the I-Smads, have discrete roles mediating BMP-dependent cellular processes during spinal interneuron development.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Smad Proteins, Receptor-Regulated/metabolism , Spinal Cord/embryology , Spinal Cord/metabolism , Animals , Avian Proteins/antagonists & inhibitors , Avian Proteins/genetics , Avian Proteins/metabolism , Axons/metabolism , Base Sequence , Chick Embryo , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Interneurons/cytology , Interneurons/metabolism , Mice , Mice, Mutant Strains , Mice, Transgenic , Models, Neurological , Neurogenesis , RNA, Small Interfering/genetics , Rats , Smad Proteins, Receptor-Regulated/antagonists & inhibitors , Smad Proteins, Receptor-Regulated/genetics , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/genetics , Smad5 Protein/metabolism , Spinal Cord/cytology
16.
Gene Ther ; 20(9): 922-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23594990

ABSTRACT

Nonhealing bone defects are difficult to treat. As the bone morphogenic protein and transforming growth factor beta pathways have been implicated in bone healing, we hypothesized that percutaneous Smad7 silencing would enhance signaling through both pathways and improve bone formation. Critical sized parietal trephine defects were created and animals received percutaneous injection of: agarose alone or agarose containing nonsense or Smad7 small interfering RNA (siRNA). At 12 weeks, SMADs1, 2, 3, 5, 7 and 8 levels were assessed. Smad1/5/8 osteogenic target, Dlx5, and SMAD2/3 angiogenic target, plasminogen activator inhibitor-1 (Pai1), transcription levels were measured. Noncanonical signaling through TGFß activated kinase-1 (Tak1) and target, runt-related transcription factor 2 (Runx2) and collagen1α1 (Col1α1), transcription were also measured. Micro-computed tomography and Gomori trichome staining were used to assess healing. Percutaneous injection of Smad7 siRNA significantly knocked down Smad7 mRNA (86.3 ± 2.5%) and protein levels (46.3 ± 3.1%). The SMAD7 knockdown resulted in a significant increase in receptor-regulated SMADs (R-SMAD) (Smad 1/5/8 and Smad2/3) nuclear translocation. R-SMAD nuclear translocation increased Dlx5 and Pai1 transcription. Additionally, noncanonical signaling through Tak1 increased Runx2 and Col1α1 target transcription. Compared with animals treated with agarose alone (33.9 ± 2.8% healing) and nonsense siRNA (31.5 ± 11.8% healing), animals treated Smad7 siRNA had significantly great (91.2 ± 3.8%) healing. Percutaneous Smad7 silencing increases signal transduction through canonical and noncanonical pathways resulting in significant bone formation. Minimally invasive gene therapies may prove effective in the treatment of nonhealing bone defects.


Subject(s)
Fractures, Bone/therapy , Genetic Therapy , Osteogenesis , Skull , Smad7 Protein/genetics , Smad7 Protein/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Models, Animal , Fractures, Bone/genetics , Fractures, Bone/metabolism , Gene Knockdown Techniques , Humans , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Signal Transduction , Skull/metabolism , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism
17.
Hum Mol Genet ; 20(5): 894-904, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21138941

ABSTRACT

Loss-of-function mutations in the genes encoding dystrophin and the associated membrane proteins, the sarcoglycans, produce muscular dystrophy and cardiomyopathy. The dystrophin complex provides stability to the plasma membrane of striated muscle during muscle contraction. Increased SMAD signaling due to activation of the transforming growth factor-ß (TGFß) pathway has been described in muscular dystrophy; however, it is not known whether this canonical TGFß signaling is pathogenic in the muscle itself. Drosophila deleted for the γ/δ-sarcoglycan gene (Sgcd) develop progressive muscle and heart dysfunction and serve as a model for the human disorder. We used dad-lacZ flies to demonstrate the signature of TGFß activation in response to exercise-induced injury in Sgcd null flies, finding that those muscle nuclei immediately adjacent to muscle injury demonstrate high-level TGFß signaling. To determine the pathogenic nature of this signaling, we found that partial reduction of the co-SMAD Medea, homologous to SMAD4, or the r-SMAD, Smox, corrected both heart and muscle dysfunction in Sgcd mutants. Reduction in the r-SMAD, MAD, restored muscle function but interestingly not heart function in Sgcd mutants, consistent with a role for activin but not bone morphogenic protein signaling in cardiac dysfunction. Mammalian sarcoglycan null muscle was also found to exhibit exercise-induced SMAD signaling. These data demonstrate that hyperactivation of SMAD signaling occurs in response to repetitive injury in muscle and heart. Reduction of this pathway is sufficient to restore cardiac and muscle function and is therefore a target for therapeutic reduction.


Subject(s)
Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila , Heart/physiopathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Smad Proteins, Receptor-Regulated/metabolism , Smad4 Protein/metabolism , Animals , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Female , Humans , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Myocardium/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta/metabolism
18.
Genome Res ; 20(1): 36-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19926752

ABSTRACT

Embryonic stem (ES) cells are under precise control of both intrinsic self-renewal gene regulatory network and extrinsic growth factor-triggered signaling cascades. How external signaling pathways connect to core self-renewal transcriptional circuits is largely unknown. To probe this, we chose BMP signaling, which is previously recognized as a master control for both self-renewal and lineage commitment of murine ES cells. Here, we mapped target gene promoter occupancy of SMAD1/5 and SMAD4 on a genome-wide scale and found that they associate with a large group of developmental regulators that are enriched for H3K27 trimethylation and H3K4 trimethylation bivalent marks and are repressed in the self-renewing state, whereas they are rapidly induced upon differentiation. Smad knockdown experiments further indicate that SMAD-mediated BMP signaling is largely required for differentiation-related processes rather than directly influencing self-renewal. Among the SMAD-associated genes, we further identified Dpysl2 (previously known as Crmp2) and the H3K27 demethylase Kdm6b (previously known as Jmjd3) as BMP4-modulated early neural differentiation regulators. Combined with computational analysis, our results suggest that SMAD-mediated BMP signaling balances self-renewal versus differentiation by modulating a set of developmental regulators.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Chromosome Mapping , Embryonic Stem Cells/cytology , Signal Transduction , Smad Proteins, Receptor-Regulated , Animals , Binding Sites , Bone Morphogenetic Proteins/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genome , Mice , Molecular Sequence Data , Neurons/cytology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Sequence Analysis, DNA , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism
19.
Development ; 137(3): 477-85, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20081192

ABSTRACT

The C. elegans daf-8 gene encodes an R-Smad that is expressed in a subset of head neurons, the intestine, gonadal distal tip cells and the excretory cell. We found that DAF-8, which inhibits the DAF-3 Co-Smad, is associated with DAF-3 and the DAF-14 Smad in vivo and in vitro. Overexpression of daf-8 conferred a dauer-defective phenotype and suppressed constitutive dauer formation in daf-8 and daf-14 mutants. In contrast to mammalian systems described thus far, active DAF-3 drives a feedback regulatory loop that represses transcription of daf-7 (a TGFbeta ligand) and daf-8 by directly binding to their regulatory regions. Hence, DAF-8 and DAF-3 are mutually antagonistic. The feedback repression may reinforce the developmental switch by allowing DAF-3 to freely activate dauer transcription in target tissues, unless sufficiently inhibited by DAF-8 and DAF-14. In the adult, DAF-8 downregulates lag-2 expression in the distal tip cells, thus promoting germ line meiosis. This function does not involve DAF-3, thereby avoiding the feedback loop that functions in the dauer switch.


Subject(s)
Feedback, Physiological , Smad Proteins/genetics , Transcription Factors/physiology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Gene Regulatory Networks , Mutation , Phenotype , Smad Proteins/physiology , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
20.
J Biol Chem ; 286(29): 26016-27, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21566131

ABSTRACT

ADAMTS5 has been implicated in the degradation of cartilage aggrecan in human osteoarthritis. Here, we describe a novel role for the enzyme in the regulation of TGFß1 signaling in dermal fibroblasts both in vivo and in vitro. Adamts5(-/-) mice, generated by deletion of exon 2, exhibit impaired contraction and dermal collagen deposition in an excisional wound healing model. This was accompanied by accumulation in the dermal layer of cell aggregates and fibroblastic cells surrounded by a pericellular matrix enriched in full-length aggrecan. Adamts5(-/-) wounds exhibit low expression (relative to wild type) of collagen type I and type III but show a persistently elevated expression of tgfbRII and alk1. Aggrecan deposition and impaired dermal repair in Adamts5(-/-) mice are both dependent on CD44, and Cd44(-/-)/Adamts5(-/-) mice display robust activation of TGFß receptor II and collagen type III expression and the dermal regeneration seen in WT mice. TGFß1 treatment of newborn fibroblasts from wild type mice results in Smad2/3 phosphorylation, whereas cells from Adamts5(-/-) mice phosphorylate Smad1/5/8. The altered TGFß1 response in the Adamts5(-/-) cells is dependent on the presence of aggrecan and expression of CD44, because Cd44(-/-)/Adamts5(-/-) cells respond like WT cells. We propose that ADAMTS5 deficiency in fibrous tissues results in a poor repair response due to the accumulation of aggrecan in the pericellular matrix of fibroblast progenitor cells, which prevents their transition to mature fibroblasts. Thus, the capacity of ADAMTS5 to modulate critical tissue repair signaling events suggests a unique role for this enzyme, which sets it apart from other members of the ADAMTS family of proteases.


Subject(s)
ADAM Proteins/deficiency , Aggrecans/metabolism , Dermis/physiopathology , Hyaluronan Receptors/metabolism , Sequence Deletion , Transforming Growth Factor beta1/metabolism , Wound Healing/genetics , ADAM Proteins/genetics , ADAMTS5 Protein , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activin Receptors, Type II , Aggrecans/genetics , Animals , Animals, Newborn , Cell Aggregation/drug effects , Dermis/drug effects , Dermis/metabolism , Dermis/pathology , Epithelioid Cells/drug effects , Epithelioid Cells/metabolism , Epithelioid Cells/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation/drug effects , Genotype , Humans , Male , Mice , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins, Receptor-Regulated/genetics , Smad Proteins, Receptor-Regulated/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL