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










Publication year range
1.
J Biol Chem ; 296: 100496, 2021.
Article in English | MEDLINE | ID: mdl-33667543

ABSTRACT

Bone morphogenetic proteins (BMPs) secreted by a variety of cell types are known to play essential roles in cell differentiation and matrix formation in the bone, cartilage, muscle, blood vessel, and neuronal tissue. BMPs activate intracellular effectors via C-terminal phosphorylation of Smad1, Smad5, and Smad9, which relay the signaling by forming a complex with Smad4 and translocate to the nucleus for transcriptional activation. Smad6 inhibits BMP signaling through diverse mechanisms operative at the membrane, cytosolic, and nuclear levels. However, the mechanistic underpinnings of Smad6 functional diversity remain unclear. Here, using a biochemical approach and cell differentiation systems, we report a cytosolic mechanism of action for Smad6 that requires arginine methylation at arginine 81 (R81) and functions through association with Smad1 and interference with the formation of Smad1-Smad4 complexes. By mutating the methylated arginine residue, R81, and by silencing the expression of protein arginine methyltransferase 1, we show that protein arginine methyltransferase 1 catalyzes R81 methylation of Smad6 upon BMP treatment, R81 methylation subsequently facilitates Smad6 interaction with the phosphorylated active Smad1, and R81 methylation facilitates Smad6-mediated interruption of Smad1-Smad4 complex formation and nuclear translocation. Furthermore, Smad6 WT but not the methylation-deficient R81A mutant inhibited BMP-responsive transcription, attenuated BMP-mediated osteogenic differentiation, and antagonized BMP-mediated inhibition of cell invasion. Taken together, our results suggest that R81 methylation plays an essential role in Smad6-mediated inhibition of BMP responses.


Subject(s)
Arginine/metabolism , Bone Morphogenetic Proteins/metabolism , Osteogenesis/physiology , Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Smad1 Protein/metabolism , Smad4 Protein/metabolism , Smad6 Protein/metabolism , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Cell Line , Humans , Methylation , Smad1 Protein/antagonists & inhibitors , Smad4 Protein/antagonists & inhibitors , Smad6 Protein/chemistry
2.
Biochem Cell Biol ; 99(5): 578-586, 2021 10.
Article in English | MEDLINE | ID: mdl-33646885

ABSTRACT

Duloxetine, a selective serotonin-norepinephrine reuptake inhibitor, is currently recommended for the treatment of chronic painful disorders such as fibromyalgia, chronic musculoskeletal pain, and diabetic peripheral neuropathy. We previously demonstrated that bone morphogenetic protein-4 (BMP-4) stimulates osteoprotegerin (OPG) production in osteoblast-like MC3T3-E1 cells, and that p70 S6 kinase positively regulates OPG synthesis. The present study aimed to investigate the effect of duloxetine on BMP-4-stimulated OPG synthesis in these cells. Duloxetine dose-dependently suppressed OPG release stimulated by BMP-4. Fluvoxamine, a selective serotonin reuptake inhibitor (SSRI), reduced BMP-4-stimulated OPG release, whereas a selective and specific norepinephrine reuptake inhibitor, reboxetine, failed to affect OPG release. In addition, another SSRI sertraline also inhibited BMP-4-stimulated OPG release. On the other hand, siRNA of SMAD1 reduced the OPG release stimulated by BMP-4, indicating the involvement of the SMAD1/5/8 pathway in OPG release. Rapamycin inhibited BMP-4-stimulated p70 S6 kinase phosphorylation, and compound C suppressed the SMAD1/5/8 phosphorylation stimulated by BMP-4. Duloxetine did not affect BMP-4-induced phosphorylation of p70 S6 kinase but suppressed SMAD1/5/8 phosphorylation. Both fluvoxamine and sertraline also inhibited BMP-4-elicited phosphorylation of SMAD1/5/8. These results strongly suggest that duloxetine suppresses BMP-4-stimulated OPG release via inhibition of the Smad1/5/8 signaling pathway in osteoblasts.


Subject(s)
Bone Morphogenetic Protein 4/antagonists & inhibitors , Duloxetine Hydrochloride/pharmacology , Osteoblasts/drug effects , Osteoprotegerin/antagonists & inhibitors , 3T3 Cells , Animals , Bone Morphogenetic Protein 4/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Osteoblasts/metabolism , Osteoprotegerin/metabolism , Signal Transduction/drug effects , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/metabolism , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/metabolism , Smad8 Protein/antagonists & inhibitors , Smad8 Protein/metabolism
3.
Eur Rev Med Pharmacol Sci ; 24(10): 5691-5696, 2020 05.
Article in English | MEDLINE | ID: mdl-32495904

ABSTRACT

OBJECTIVE: To elucidate the role of Prunella vulgaris L (PVL) in protecting glucocorticoids (GC)-induced osteogenesis inhibition, thereafter, protecting the deterioration of osteoporosis (OP). MATERIALS AND METHODS: Cell Counting Kit-8 (CCK-8) assay was conducted to assess the influence of PVL treatment on MSCs viability. Osteogenesis in MSCs was induced by Dexamethasone (DEX) stimulation. Regulatory effects of PVL on osteogenesis-related gene expressions, ALP activity, and mineralization ability in DEX-induced MSCs were determined. At last, protein levels of p-Smad1/5/9 and total-Smad1/5/9 influenced by DEX and PVL were measured by Western blot. RESULTS: PVL treatment did not pose a time- or dose-dependent influence on MSCs viability. DEX induction in MSCs downregulated ALP, RUNX2, Bglap, and Osterix. ALP activity and mineralization in DEX-induced MSCs were suppressed. Downregulated osteogenesis-related genes decreased ALP activity and mineralization in MSCs undergoing DEX stimulation were partially reversed by PVL treatment. Moreover, the downregulated p-Smad1/5/9 level in DEX-induced MSCs was elevated by PVL treatment, while total-Smad1/5/9 was not affected. CONCLUSIONS: PVL alleviated GC-induced suppression in MSCs osteogenesis by activating the Smad pathway, thereafter, protecting the deterioration of OP.


Subject(s)
Dexamethasone/antagonists & inhibitors , Glucocorticoids/antagonists & inhibitors , Mesenchymal Stem Cells/drug effects , Protective Agents/pharmacology , Prunella/chemistry , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Humans , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Protective Agents/chemistry , Smad1 Protein/antagonists & inhibitors , Smad5 Protein/antagonists & inhibitors , Smad8 Protein/antagonists & inhibitors
4.
Pak J Pharm Sci ; 33(1(Special)): 403-408, 2020 Jan.
Article in English | MEDLINE | ID: mdl-32173634

ABSTRACT

Inflammatory response that occur post-ischemia is a serious problem in the treatment of ischemic brain disease. MicroRNA-155 is a brain-specific or brain-enriched miRNA, which mediates inflammatory reactions in cerebral ischemic tissue by regulating inflammatory signal and the expression level of SOCS1. The present study was aimed to assess the effect of GuaLou GuiZhi Decoction (GLGZD) on miR-155 expression in activated microglia following inflammation and further explore the role of GLGZD on expression of the inflammation-related gene. BV2 cells were used to simulated by LPS to make the inflammatory model. Expression level of miR-155 was detected by Real-Time PCR. BV2 cells after simulated by LPS were then transfected with miR-155 mimic and its negative controls. Cytokines release were measured by corresponding purchased ELISA kits, respectively. Then target protein expression of miR-155 were detected by western blotting assay. After miRNA over expression transfections, expressions of inflammation-related factors, SOCS-1 and SAMD in BV2 cells after activation were measured by Western blot assay. Results showed that in BV2 cells after simulated by LPS, miR-155 was upregulated. The elevated miR-155 expression enhanced the inflammatory cytokine release. miR-155 directly target and negatively regulated SOCS-1 and SMAD-1 expression. Over expression of SOCS-1 and SMAD reduced inflammatory action that was enhanced by miR-155 mimic transfection. miR-155 was positively related with activation of NF-Ï°B signal pathways via SOCS-1 and SMAD. In conclusion, GuaLou GuiZhi Decoction (GLGZD) might exert its anti-inflammatory action by inhibiting the expression of miR-155, indicating that miR-155 may be used as a treatment target in clinical treatment with GuaLou GuiZhi Decoction (GLGZD) in ischemic brain.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Lipopolysaccharides/pharmacology , MicroRNAs/physiology , Microglia/drug effects , Animals , Brain Ischemia/drug therapy , Cells, Cultured , Cytokines/biosynthesis , Mice , MicroRNAs/antagonists & inhibitors , Microglia/physiology , Smad1 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling 1 Protein/antagonists & inhibitors
5.
Mol Med Rep ; 21(1): 61-68, 2020 01.
Article in English | MEDLINE | ID: mdl-31939626

ABSTRACT

Considering the increased resistance to antibiotics in the clinic and the ideal antibacterial properties of KR­12, the effects of KR­12­a6, an important analogue of KR­12, on the osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) were investigated. Osteogenic differentiation­associated experiments were conducted in hBMSCs, and KR­12­a6 was used as an additional stimulating factor during osteogenic induction. Quantitative analysis of alkaline phosphatase (ALP) and alizarin red staining, and reverse transcription­quantitative PCR analysis of the expression of osteogenesis­associated genes were performed to determine the effects of KR­12­a6 on the osteogenic differentiation of hBMSCs. LDN­212854 was selected to selectively suppress BMP/SMAD signaling. Western blotting was performed to investigate the underlying mechanisms. The intensity of ALP and alizarin red staining gradually increased with increasing KR­12­a6 concentrations. KR­12­a6 induced the strongest staining at 40 µg/ml, whereas 60 µg/ml and 80 µg/ml concentrations did not further increase the intensity of staining. The mRNA expression levels of RUNX2 and ALP increased in a dose­dependent manner as early as 3 days post­KR­12­a6 treatment. The mRNA expression of COL1A1, BSP and BMP2 exhibited significant upregulation from day 7 post­KR­12­a6 treatment. In contrast, the mRNA levels of OSX, OCN and OPN were enhanced dramatically at day 14 following KR­12­a6 stimulation. Additionally, KR­12­a6 significantly promoted the phosphorylation of Smad1/5. Furthermore, LDN­212854 suppressed the activation of Smad1/5 and inhibited the upregulation of several osteogenic differentiation­associated genes in KR­12­a6­treated hBMSCs. KR­12­a6 promoted the osteogenic differentiation of hBMSCs via BMP/SMAD signaling.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Imidazoles/pharmacology , Nylons/pharmacology , Osteogenesis/drug effects , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Adult , Alkaline Phosphatase/metabolism , Bone Morphogenetic Protein 2/genetics , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Imidazoles/pharmacokinetics , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Nylons/pharmacokinetics , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteopontin/genetics , Peptide Fragments/genetics , Signal Transduction/genetics , Smad1 Protein/antagonists & inhibitors , Smad5 Protein/antagonists & inhibitors , Sp7 Transcription Factor/genetics , Sp7 Transcription Factor/metabolism
6.
Toxicol Appl Pharmacol ; 355: 127-137, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30008374

ABSTRACT

Fibrosis is a pathophysiological state characterized by the excessive formation/deposition of fibrous extracellular matrix. Transforming growth factor-beta (TGF-ß) is a central profibrotic mediator, and targeting TGF-ß is a promising strategy in the development of drugs for the treatment of fibrosis. Therefore, the effect of LY2109761, a small molecule inhibitor against TGF-ß with targets beyond TGF-ß signaling, on fibrogenesis was elucidated in vitro (HepG2 cells and LX-2 cells) and ex vivo (human and rat precision-cut liver slices). Our results displayed an anti-fibrotic effect of LY2109761, as it markedly down-regulated gene and protein expression of collagen type 1, as well as gene expression of the inhibitor of metalloproteinases 1. This effect on fibrosis markers was partially mediated by targeting TGF-ß signaling, seeing that LY2109761 inhibited TGF-ß1 gene expression and SMAD2 protein phosphorylation. Interestingly, particularly at a high concentration, LY2109761 decreased SMAD1 protein phosphorylation and gene expression of the inhibitor of DNA binding 1, which appeared to be TGF-ß-independent effects. In conclusion, LY2109761 exhibited preclinical anti-fibrotic effects via both TGF-ß-dependent and -independent pathways. These results illustrate that small molecule inhibitors directed against TGF-ß could possibly influence numerous signaling pathways and thereby mitigate fibrogenesis.


Subject(s)
Fibrosis/drug therapy , Pyrazoles/pharmacology , Pyrroles/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Cell Line , Collagen Type I/antagonists & inhibitors , Collagen Type I/biosynthesis , Down-Regulation , Gene Expression/drug effects , Humans , In Vitro Techniques , Liver/drug effects , Liver/metabolism , Male , Phosphorylation , Rats , Rats, Wistar , Smad1 Protein/antagonists & inhibitors , Smad2 Protein/antagonists & inhibitors , Tissue Inhibitor of Metalloproteinase-2/antagonists & inhibitors
7.
Mol Hum Reprod ; 23(3): 155-165, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27986931

ABSTRACT

STUDY QUESTION: Does bone morphogenetic protein 2 (BMP2) regulate connexin43 (Cx43) and modulate cell-cell communication in luteinized human granulosa cells? SUMMARY ANSWER: BMP2 decreases gap junction intercellular communication (GJIC) of luteinized human granulosa cells by down-regulating Cx43 expression through an activin receptor-like kinase (ALK)2/ALK3-mediated Sma- and Mad-related protein (SMAD)-dependent signaling pathway. WHAT IS KNOWN ALREADY: BMP2 and its putative receptors are highly expressed in the human corpus luteum and are involved in the process of luteolysis. Cx43-coupled gap junctions play a critical role in the development and maintenance of corpus luteum. STUDY DESIGN DURATION: This is a laboratory study conducted over a 1-year period. At least three independent experiments with three replicates were conducted and the experimental samples were compared with the appropriate vehicle controls for all of the inhibition-approach, concentration-dependent or time-course studies. PARTICIPANTS/MATERIALS, SETTING, METHODS: SVOG cell line (immortalized human granulosa-lutein cells derived from in vitro fertilization patients in an academic research center) was used as the study model. The changes of Cx43 expression and levels of phosphorylated SMAD1/5/8 protein were evaluated after exposure to recombinant human BMP2. Real-time quantitative PCR and Western blot analysis were used to examine the specific mRNA and protein levels, respectively. The BMP/TGF-ß type I receptor inhibitors (Dorsomorphin, DMH-1 and SB431542) and target depletion small interfering RNAs (ALK2, ALK3, ALK6 and SMAD4) were used to investigate the underlying molecular mechanisms. A scrape loading and dye transfer assay was used to evaluate the GJIC between the SVOG cells. MAIN RESULTS AND THE ROLE OF CHANCE: Treatment with BMP2 down-regulated the expression of Cx43 and decreased the GJIC activity, whereas it increased the phosphorylated SMAD1/5/8 protein in SVOG cells (P < 0.05). These biological effects were abolished by pre-treatment with the BMP type I receptor inhibitors, Dorsomorphin and DMH-1 (P < 0.05), but not SB431542. Additionally, the individual or concomitant small interfering RNA-mediated knockdown of ALK2 and ALK3, but not ALK6 attenuated the BMP2-induced increases in phosphorylated SMAD1/5/8 and down-regulation of Cx43 expression (P < 0.05). The knockdown of SMAD4 completely abolished the BMP2-induced down-regulation of Cx43 expression (P < 0.05). LIMITATIONS REASONS FOR CAUTION: This experimental study was conducted in an in vitro cell culture system, and may not reflect a realistic intra-ovarian environment. WIDER IMPLICATIONS OF THE FINDINGS: Our results suggested that BMP2 may be involved in the local modulation of cell-cell communication in the luteal phase. This study also represents the first comprehensive research of molecular mechanisms of BMP2 in the down-regulation Cx43 in luteinized human granulosa cells. Such data may provide valuable insights into ovarian physiology and benefit the development of potential therapeutic methods for patients suffering from luteal insufficiency. LARGE SCALE DATA: N/A. STUDY FUNDING AND COMPETING INTEREST(s): This research was supported by an operating grant from the China-Canadian Joint Health Research Initiative Grants Program to P.C.K. Leung and J.Z. Sheng. The authors declare no competing interest with the contents of this article.


Subject(s)
Activin Receptors, Type I/genetics , Bone Morphogenetic Protein 2/pharmacology , Cell Communication/drug effects , Connexin 43/genetics , Luteal Cells/drug effects , Activin Receptors, Type I/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Line, Transformed , Connexin 43/antagonists & inhibitors , Connexin 43/metabolism , Female , Gap Junctions/drug effects , Gap Junctions/metabolism , Gene Expression Regulation , Humans , Luteal Cells/cytology , Luteal Cells/metabolism , Phosphorylation/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinolines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/genetics , Smad5 Protein/metabolism , Smad8 Protein/antagonists & inhibitors , Smad8 Protein/genetics , Smad8 Protein/metabolism
8.
FEBS J ; 283(9): 1689-700, 2016 05.
Article in English | MEDLINE | ID: mdl-26929154

ABSTRACT

Intervertebral discs (IVDs) provide stability and flexibility to the spinal column; however, IVDs, and in particular the nucleus pulposus (NP), undergo a degenerative process characterized by changes in the disc extracellular matrix (ECM), decreased cell viability, and reduced synthesis of proteoglycan and type II collagen. Here, we investigated the efficacy and feasibility of stem cell therapy using bone marrow mesenchymal stem cells (BMSCs) over-expressing bone morphogenetic protein 7 (BMP7) to promote ECM remodeling of degenerated IVDs. Lentivirus-mediated BMP7 over-expression induced differentiation of BMSCs into an NP phenotype, as indicated by expression of the NP markers collagen type II, aggrecan, SOX9 and keratins 8 and 19, increased the content of glycosaminoglycan, and up-regulated ß-1,3-glucuronosyl transferase 1, a regulator of chondroitin sulfate synthesis in NP cells. These effects were suppressed by Smad1 silencing, indicating that the effect of BMP7 on ECM remodeling was mediated by the Smad pathway. In vivo analysis in a rabbit model of disc degeneration showed that implantation of BMSCs over-expressing BMP7 promoted cell differentiation and proliferation in the NP, as well as their own survival, and these effects were mediated by the Smad pathway. The results of the present study indicate the beneficial effects of BMP7 on restoring ECM homeostasis in NP cells, and suggest potential strategies for improving cell therapy for the treatment of disc diseases.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Intervertebral Disc Degeneration/therapy , Lentivirus/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 7/metabolism , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type II/genetics , Collagen Type II/metabolism , Disease Models, Animal , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Glycosaminoglycans/metabolism , Humans , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Keratin-19/genetics , Keratin-19/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Lentivirus/metabolism , Mesenchymal Stem Cells/cytology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rabbits , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/genetics , Smad1 Protein/metabolism
9.
Dev Cell ; 34(2): 229-41, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26190145

ABSTRACT

FGF, BMP, and WNT balance embryonic nephron progenitor cell (NPC) renewal and differentiation. By modulating these pathways, we have created an in vitro niche in which NPCs from embryonic kidneys or derived from human embryonic stem cells can be propagated. NPC cultures expanded up to one billion-fold in this environment can be induced to form tubules expressing nephron differentiation markers. Single-cell culture reveals phenotypic variability within the early CITED1-expressing NPC compartment, indicating that it is a mixture of cells with varying progenitor potential. Furthermore, we find that the developmental age of NPCs does not correlate with propagation capacity, indicating that cessation of nephrogenesis is related to factors other than an intrinsic clock. This in vitro nephron progenitor niche will have important applications for expansion of cells for engraftment and will facilitate investigation of mechanisms that determine the balance between renewal and differentiation in these cells.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fibroblast Growth Factors/metabolism , Nephrons/embryology , Nuclear Proteins/biosynthesis , Trans-Activators/biosynthesis , Wnt Proteins/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Differentiation , Cell Proliferation , Cells, Cultured , Embryonic Stem Cells/cytology , Enzyme Activation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Mice, Inbred ICR , Mice, Transgenic , Nephrons/cytology , Nuclear Proteins/genetics , Organogenesis , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/metabolism , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/metabolism , Trans-Activators/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
10.
Mol Med Rep ; 12(2): 2189-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25891964

ABSTRACT

The current study investigated the role of exogenous cytochrome c in sepsis-induced myocardial dysfunction (SIMD) using a mouse model and aimed to elucidate its effect on transforming growth factor-ß1 (TGF-ß1) expression during this process. A total of 75 male Kunming mice were randomly divided into the following five group: Normal (N, n=15); sham-operation (SHAM, n=15); sepsis (CLP, n=15); normal saline (NS, n=15); and cytochrome c (Cytc, n=15). Animals were sacrificed at 0, 6 or 12 h and the samples were analyzed using transmission electron microscopy, histopathological examination, reverse transcription-quantitative polymerase chain reaction, ELISA, protein analysis by western blotting. The SIMD model was developed and a significant downregulation of TGF-ß1 gene expression, in addition to a reduction in the plasma and protein levels of TGF-ß1 as well as the protein levels of TGF-ß1-activated SMAD 1/5/8 were observed in the CLP group. The data from the current study indicate that using exogenous cytochrome c as a therapeutic strategy for SIMD is feasible, and may function via the downregulation of TGF-ß1 expression through the SMAD 1/5/8 signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cardiomyopathies/drug therapy , Cytochromes c/pharmacology , Sepsis/drug therapy , Transforming Growth Factor beta1/antagonists & inhibitors , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Disease Models, Animal , Gene Expression Regulation , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Myocardium/ultrastructure , Sepsis/complications , Sepsis/genetics , Sepsis/pathology , Signal Transduction , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/blood , Smad1 Protein/genetics , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/blood , Smad5 Protein/genetics , Smad8 Protein/antagonists & inhibitors , Smad8 Protein/blood , Smad8 Protein/genetics , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/genetics
11.
Blood ; 124(3): 393-402, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24894772

ABSTRACT

Bone morphogenetic protein (BMP) signaling regulates early hematopoietic development, proceeding from mesoderm patterning through the progressive commitment and differentiation of progenitor cells. The BMP pathway signals largely through receptor-mediated activation of Mothers Against Decapentaplegic homolog (SMAD) proteins, although alternate pathways are modulated through various components of mitogen-activated protein kinase (MAPK) signaling. Using a conditional, short hairpin RNA (shRNA)-based knockdown system in the context of differentiating embryonic stem cells (ESCs), we demonstrated previously that Smad1 promotes hemangioblast specification, but then subsequently restricts primitive progenitor potential. Here we show that co-knockdown of Smad5 restores normal progenitor potential of Smad1-depleted cells, suggesting opposing functions for Smad1 and Smad5. This balance was confirmed by cotargeting Smad1/5 with a specific chemical antagonist, LDN193189 (LDN). However, we discovered that LDN treatment after hemangioblast commitment enhanced primitive myeloid potential. Moreover, inhibition with LDN (but not SMAD depletion) increased expression of Delta-like ligands Dll1 and Dll3 and NOTCH activity; abrogation of NOTCH activity restored LDN-enhanced myeloid potential back to normal, corresponding with expression levels of the myeloid master regulator, C/EBPα. LDN but not SMAD activity was also associated with activation of the p38MAPK pathway, and blocking this pathway was sufficient to enhance myelopoiesis. Therefore, NOTCH and p38MAPK pathways balance primitive myeloid progenitor output downstream of the BMP pathway.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Myelopoiesis/physiology , Receptors, Notch/metabolism , Smad Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Morphogenetic Protein Receptors/antagonists & inhibitors , Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Calcium-Binding Proteins , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Knockdown Techniques , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/metabolism , Myelopoiesis/drug effects , Myelopoiesis/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/antagonists & inhibitors , Smad5 Protein/genetics , Smad5 Protein/metabolism
12.
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
13.
Stem Cells ; 27(8): 1741-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19544434

ABSTRACT

Inhibition of bone morphogenetic protein (BMP) signaling is required for vertebrate neural induction, and fibroblast growth factors (FGFs) may affect neural induction through phosphorylation at the linker region of Smad1, thus regulating BMP signaling. Here we show that human embryonic stem cells efficiently convert to neuroepithelial cells in the absence of BMP antagonists, or even when exposed to high concentrations of exogenous BMP4. Molecular and functional analyses revealed multiple levels of endogenous BMP signaling inhibition that may account for the efficient neural differentiation. Blocking FGF signaling inhibited neural induction, but did not alter the phosphorylation of the linker region of Smad1, suggesting that FGF enhances human neural specification independently of BMP signaling.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Proteins/antagonists & inhibitors , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Fibroblast Growth Factors/metabolism , Neurons/cytology , Neurons/drug effects , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/metabolism , Eye Proteins/biosynthesis , Fibroblast Growth Factors/antagonists & inhibitors , Fluorescent Antibody Technique , Homeodomain Proteins/biosynthesis , Humans , Immunohistochemistry , Neurons/metabolism , Octamer Transcription Factor-3/biosynthesis , PAX6 Transcription Factor , Paired Box Transcription Factors/biosynthesis , Phosphorylation , Repressor Proteins/biosynthesis , Signal Transduction/drug effects , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/metabolism
14.
Curr Opin Genet Dev ; 18(4): 304-10, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18590818

ABSTRACT

The intensity of the BMP signal is determined by cell surface receptors that phosphorylate Smad1/5/8 at the C-terminus. In addition to this BMP-activated phosphorylation, recent studies have shown that sequential phosphorylations by MAPK and GSK3 kinases can negatively regulate the activity of the pSmad1Cter signal. These phosphorylations in the linker region cause Smad1 to be transported to the centrosomal region, polyubiquitinylated and degraded by the proteasomal machinery. In Xenopus embryos, Wnt signals, which regulate GSK3, induce ectoderm to adopt an epidermal fate, and this Wnt effect requires an active BMP-Smad1/5/8 signaling pathway. These findings have profound implications for understanding how dorsal-ventral and anterior-posterior patterning are seamlessly integrated in the early embryonic morphogenetic field.


Subject(s)
Body Patterning/physiology , Smad1 Protein/physiology , Smad5 Protein/physiology , Smad8 Protein/physiology , Amino Acid Sequence , Animals , Body Patterning/genetics , Embryo, Nonmammalian , Humans , Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/physiology , Models, Biological , Molecular Sequence Data , Phosphorylation , Signal Transduction , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad8 Protein/genetics , Xenopus/embryology , Xenopus/genetics
15.
Cancer Res ; 67(6): 2747-56, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17363596

ABSTRACT

Mullerian-inhibiting substance (MIS), a transforming growth factor-beta family member, activates the nuclear factor-kappaB (NF-kappaB) pathway and induces the expression of B-cell translocation gene 2 (BTG2), IFN regulatory factor-1 (IRF-1), and the chemokine Gro-beta. Inhibiting NF-kappaB activation with a phosphorylation-deficient IkappaBalpha mutant abrogated MIS-mediated induction of all three genes. Expression of dominant-negative Smad1, in which serines at the COOH-terminal SSVS motif are converted to alanines, suppressed MIS-induced Smad1 phosphorylation and impaired MIS-stimulated Gro-beta promoter-driven reporter expression and Gro-beta mRNA. Suppressing Smad1 expression using small interfering RNA also mitigated MIS-induced Gro-beta mRNA, suggesting that regulation of Gro-beta expression by MIS was dependent on activation of NF-kappaB as well as Smad1. However, induction of IRF-1 and BTG2 mRNAs by MIS was independent of Smad1 activation. Characterization of kappaB-binding sequences within Gro-beta, BTG2, and IRF-1 promoters showed that MIS stimulated binding of p50 and p65 subunits to all three sites, whereas phosphorylated Smad1 (phospho-Smad1) protein was detectable only in the NF-kappaB complex bound to the kappaB site of the Gro-beta promoter. Consistent with these observations, chromatin immunoprecipitation assays showed recruitment of both phospho-Smad1 and p65 to the Gro-beta promoter in vivo, whereas p65, but not phospho-Smad1, was recruited to the BTG2 promoter. These results show a novel interaction between MIS-stimulated Smad1 and NF-kappaB signaling in which enhancement of NF-kappaB DNA binding and gene expression by phospho-Smad1 is dependent on the sequence of the kappaB consensus site within the promoter.


Subject(s)
Breast Neoplasms/metabolism , Chemokines, CXC/biosynthesis , Glycoproteins/pharmacology , NF-kappa B/metabolism , Smad1 Protein/metabolism , Testicular Hormones/pharmacology , Anti-Mullerian Hormone , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Chemokine CXCL2 , Chemokines, CXC/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Immediate-Early Proteins/biosynthesis , Immediate-Early Proteins/genetics , Interferon Regulatory Factor-1/biosynthesis , Interferon Regulatory Factor-1/genetics , NF-kappa B/antagonists & inhibitors , Phosphorylation , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Smad1 Protein/antagonists & inhibitors , Smad1 Protein/biosynthesis , Smad1 Protein/genetics , Transcription Factor RelA/metabolism , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...