Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 116: 105382, 2021 11.
Article in English | MEDLINE | ID: mdl-34598087

ABSTRACT

The bone morphogenetic protein-2 (BMP-2) is an essential regulator of bone formation and remodeling, which has also been implicated in the pathogenesis of osteoarthritis and its closely related chondrocyte senescence. The BMP-2 uses a conformational wrist epitope and a linear knuckle epitope to interact with type-I (BMPR-I) and type-II (BMPR-II) receptors, respectively. Previously, the knuckle epitope has been intensely studied, but the wrist epitope still remains largely unexplored due to its discontinuous nature. In the present work, the intermolecular interaction of BMP-2 with BMPR-I was investigated systematically at structural, energetic and dynamic levels. Three discrete hotspots that represent the key BMPR-I recognition sites of BMP-2 were identified; they are spatially dispersed over the two monomers of BMP-2 dimer and totally account for 83.5 % binding potency of BMP-2 to BMPR-I (hotspot 1: residues 49-70 in monomer 1; hotspot 2: residues 24-31 in monomer 2; hotspot 3: residues 88-107 in monomer 2). Therefore, we defined the three discrete hotspot sites as the core region of wrist epitope; their contribution to the binding increases in the order: hotspot 2 < hotspot 3 < hotspot 1. We demonstrated that the primary hotspot 1 site has a native U-shaped conformation in the full-length BMP-2 protein context, but it cannot maintain in the native conformation when split from the context to obtain a free hotspot-1 peptide, thus largely impairing its binding potency to BMPR-I. We further employed disulfide-bonded cyclization and head-to-tail cyclization to constrain the peptide conformation, and found that only the former can effectively constrain the peptide into native conformation, thus considerably improving its binding affinity to BMPR-I, whereas the latter totally disorders the native conformation, thus rendering the peptide as a full nonbinder of BMPR-I.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Chondrocytes/drug effects , Drug Design , Peptides/pharmacology , Bone Morphogenetic Protein 2/metabolism , Chondrocytes/metabolism , Dose-Response Relationship, Drug , Epitopes/drug effects , Humans , Molecular Structure , Osteogenesis/drug effects , Peptides/chemical synthesis , Peptides/chemistry , Structure-Activity Relationship
2.
Neurotherapeutics ; 18(3): 1798-1814, 2021 07.
Article in English | MEDLINE | ID: mdl-34159538

ABSTRACT

Oligodendrocyte precursor cells (OPCs) are present in demyelinated lesions of multiple sclerosis (MS) patients. However, their differentiation into functional oligodendrocytes is insufficient, and most lesions evolve into nonfunctional astroglial scars. Blockade of bone morphogenetic protein (BMP) signaling induces differentiation of OPCs into myelin-producing oligodendrocytes. We studied the effect of specific blockade of BMP-2/4 signaling, by intravenous (IV) treatment with anti-BMP-2/4 neutralizing mAb in both the inflammatory model of relapsing experimental autoimmune encephalomyelitis (R-EAE) and the cuprizone-toxic model of demyelination in mice. Administration of anti-BMP-2/4 to R-EAE-induced mice, on day 9 post-immunization (p.i.), ameliorated R-EAE signs, diminished the expression of phospho-SMAD1/5/8, primarily within the astrocytic lineage, increased the numbers of de novo immature and mature oligodendrocytes, and reduced the numbers of newly generated astrocytes within the spinal cord as early as day 18 p.i. This effect was accompanied with elevated remyelination, manifested by increased density of remyelinating axons (0.8 < g-ratios < 1), and reduced fully demyelinated and demyelinating axons, in the anti-BMP-2/4-treated R-EAE mice, studied by electron microscopy. No significant immunosuppressive effect was observed in the CNS and in the periphery, during the peak of the first attack, or at the end of the experiment. Moreover, IV treatment with anti-BMP-2/4 mAb in the cuprizone-challenged mice augmented the numbers of mature oligodendrocytes and remyelination in the corpus callosum during the recovery phase of the disease. Based on our findings, the specific blockade of BMP-2/4 has a therapeutic potential in demyelinating disorders such as MS, by inducing early oligodendrogenesis-mediated remyelination in the affected tissue.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 4/antagonists & inhibitors , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Oligodendroglia/drug effects , Remyelination/drug effects , Administration, Intravenous , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Male , Mice , Mice, Inbred C57BL , Oligodendroglia/physiology , Remyelination/physiology
3.
Drug Des Devel Ther ; 15: 1717-1724, 2021.
Article in English | MEDLINE | ID: mdl-33935494

ABSTRACT

BACKGROUND: Osteonecrosis of the femoral head (ONFH) seriously affects the quality of life and labor ability of patients. It is urgent and vital to find the methods for necrosis clinical treatment. OBJECTIVE: This study aims to study the potential protective effects of Alendronate in the early stage of femur head necrosis. METHODS: Ten clinal ONFH tissue samples were employed. H&E staining was employed for the observation of the pathological characteristics of ONFH. The rat model (n=12) was established by the treatment of liquid nitrogen and then treated with Alendronate. The protein expression of BMP2, EIF2AK3, EIF2A and ATF4 were detected via Western blotting and IHC. RESULTS: Fibrin and necrotizing granulation tissue were observed in ONFH tissues with lymphocytes and plasma cells infiltrating in the necrotic area, exhibiting the inflammatory muscle with abnormal shape and color. In the Model group, the BMP2 and ATF4 were mainly distributed in the cell boundaries. The relative protein expression of BMP2, EIF2AK3, EIF2A, ATF4 was decreased in the Model group, compared to the NC group, which was partially recovered by the Alendronate application. CONCLUSION: Alendronate application partially reversed the suppression of expression of BMP2, EIF2AK3, EIF2A, ATF4 caused by liquid nitrogen. Alendronate could be a promising strategy of curing ONFH via targeting BMP2/EIF2AK3/EIF2A/ATF4 pathway.


Subject(s)
Activating Transcription Factor 4/metabolism , Alendronate/pharmacology , Bone Morphogenetic Protein 2/metabolism , Femur Head Necrosis/drug therapy , Up-Regulation/drug effects , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Female , Femur Head Necrosis/metabolism , Femur Head Necrosis/pathology , Humans , Male , Middle Aged , Nitrogen/pharmacology , Osteonecrosis/drug therapy , Osteonecrosis/metabolism , Osteonecrosis/pathology , Rats , Rats, Sprague-Dawley , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics
4.
Protein J ; 39(6): 703-710, 2020 12.
Article in English | MEDLINE | ID: mdl-33130958

ABSTRACT

Bone morphogenic protein-2 (BMP-2) is the most documented member of BMP family and plays a crucial role in bone formation and growth. In this study, we systematically analyze and compare the complex crystal structures and interaction properties of BMP-2 with its cognate receptors BMPR-I/BMPR-II and with its natural antagonist crossveinless-2 (CV-2) using an integrated in silico-in vitro strategy. It is found that the antagonist-binding site is not fully overlapped with the two receptor-binding sites on BMP-2 surface; the antagonist can competitively disrupt BMP-2-BMPR-II interaction using a blocking-out-of-site manner, but has no substantial influence on BMP-2-BMPR-I interaction. Here, the antagonist-binding site is assigned as a new functional epitope armpit to differ from the traditional conformational epitope wrist and linear epitope knuckle at receptor-binding sites. Structural analysis reveals that the armpit comprises three sequentially discontinuous, structurally vicinal peptide segments, separately corresponding to a loop region and two ß-strands crawling on the protein surface. The three segments cannot work independently when splitting from the protein context, but can restore binding capability to CV-2 if they are connected to a single peptide. A systematic combination of different-length polyglycine linkers between these segments obtains a series of designed single peptides, from which several peptides that can potently interact with the armpit-recognition site of CV-2 with high affinity and specificity are identified using energetic analysis and fluorescence assay; they are expected to target BMP-2-CV-2 interaction in a self-inhibitory manner.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/chemistry , Carrier Proteins/chemistry , Computer Simulation , Epitopes/chemistry , Binding Sites , Bone Morphogenetic Protein Receptors, Type I/chemistry , Bone Morphogenetic Protein Receptors, Type II/chemistry , Humans
5.
J Comput Chem ; 41(30): 2544-2561, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32935366

ABSTRACT

In Glioblastoma (GBM) brain tumors, both Gremlin-1 and Noggin are reported to bind to BMP and inhibit BMP-signaling, thereby allowing the cell to maintain tumorous morphology. Enlisting the interfacial residues important for protein-protein complex formation between BMPs (BMP-2 and BMP-7) and antagonists (Gremlin-1 and Noggin), we analyzed the structural basis of their interactions. We found possible key mutations that destabilize these complexes, which may prevent GBM development. It was also observed that when the interfacial residues were either mutated to histidine or tryptophan, it led to higher destabilization energy values. Besides, our study of the Noggin interactive model of BMP-2 suggested preferential binding at binding site II over binding site I. In the case of Gremlin-1 and BMPs, our research, along with few previous studies, indicates a close-ended cis-trans interactive model.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 7/antagonists & inhibitors , Carrier Proteins/chemistry , Glioblastoma/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Binding Sites , Carrier Proteins/metabolism , Histidine/chemistry , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Thermodynamics , Tryptophan/chemistry
6.
Cells ; 9(4)2020 04 10.
Article in English | MEDLINE | ID: mdl-32290085

ABSTRACT

Osteoarthritis (OA) is a chronic inflammatory and progressive joint disease that results in cartilage degradation and subchondral bone remodeling. The proinflammatory cytokine interleukin 1 beta (IL-1ß) is abundantly expressed in OA and plays a crucial role in cartilage remodeling, although its role in the activity of chondrocytes in cartilage and subchondral remodeling remains unclear. In this study, stimulating chondrogenic ATDC5 cells with IL-1ß increased the levels of bone morphogenetic protein 2 (BMP-2), promoted articular cartilage degradation, and enhanced structural remodeling. Immunohistochemistry staining and microcomputed tomography imaging of the subchondral trabecular bone region in the experimental OA rat model revealed that the OA disease promotes levels of IL-1ß, BMP-2, and matrix metalloproteinase 13 (MMP-13) expression in the articular cartilage and enhances subchondral bone remodeling. The intra-articular injection of Noggin protein (a BMP-2 inhibitor) attenuated subchondral bone remodeling and disease progression in OA rats. We also found that IL-1ß increased BMP-2 expression by activating the mitogen-activated protein kinase (MEK), extracellular signal-regulated kinase (ERK), and specificity protein 1 (Sp1) signaling pathways. We conclude that IL-1ß promotes BMP-2 expression in chondrocytes via the MEK/ERK/Sp1 signaling pathways. The administration of Noggin protein reduces the expression of IL-1ß and BMP-2, which prevents cartilage degeneration and OA development.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Carrier Proteins/metabolism , Interleukin-1beta/antagonists & inhibitors , Osteoarthritis/metabolism , Animals , Bone Morphogenetic Protein 2/biosynthesis , Bone Morphogenetic Protein 2/metabolism , Bone Remodeling , Carrier Proteins/genetics , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Line , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Male , Mice , Osteoarthritis/genetics , Osteoarthritis/pathology , Rats , Rats, Sprague-Dawley , Transcriptome , Transfection
7.
Nutr Cancer ; 72(5): 873-883, 2020.
Article in English | MEDLINE | ID: mdl-31409173

ABSTRACT

Microcalcification seems to be an assurance signature for the prediction of breast cancer malignancy. However, neither systematic study for deciphering the molecular mechanism of mammary microcalcification has yet been conducted, nor a mechanistic study has been performed to find out its prevention. Thus, this study firstly aimed at determining if malignant breast tissues/metastatic breast cancer cells exhibit elevated intrinsic osteoblast-like potential responsible for driving the pathological microcalcification in breast tumors. Here, tumor sample analysis showed higher levels of various osteogenic genes (e.g., Runx2, osterix), and increased ALP activity and calcification in malignant breast tissues when compared to benign tissues, indicating the existence of elevated osteoblast-like potential in malignant breast tissues as compared to benign tissues. Similarly, cell culture study found that metastatic MDA-MB-231 cells acquired a higher osteoblast-like potential as compared to less metastatic breast cancer MCF-7 cells. It was also noticed that osteoinducer bone morphogenetic protein 2 (BMP-2) increased osteoblast-like differentiation and calcification potential in breast cancer cells. Moreover, omega-3 fatty acid docosahexaenoic acid (DHA) showed an inhibitory effect on BMP-2 induced osteoblast-like potential presumably by abrogating BMP signaling. Thus, this study for the first time unraveled that DHA may mitigate microcalcification by blocking osteoblast-like potential of breast cancer cells.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Breast Neoplasms/drug therapy , Docosahexaenoic Acids/pharmacology , Osteoblasts/drug effects , Bone Morphogenetic Protein 2/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcinosis/drug therapy , Calcinosis/metabolism , Calcinosis/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Humans , Osteoblasts/metabolism , Osteoblasts/pathology , Signal Transduction
8.
Stem Cell Res Ther ; 10(1): 237, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31387619

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common tumors globally, with varying prevalence based on endemic risk factors. Bone morphogenetic protein (BMP) exhibits a broad spectrum of biological activities in various tissues including angiogenesis. Here, this study aimed to investigate the mechanism of BMP2 in HCC by mediating the mitogen-activated protein kinase (MAPK)/p38 signaling pathway. METHODS: BMP2 expression was quantified in HCC and adjacent tissues. BMP2 gain- and loss-of-function experiments were conducted by infection with lentivirus over-expressing BMP2 or expressing shRNA against BMP2. The angiogenesis was evaluated with HepG2 cells co-cultured with ECV304 cells. SB-239063 was applied to inhibit the activation of the MAPK/p38 signaling pathway so as to identify the significance of this pathway in HCC progression. Finally, in vivo experiments were conducted to identify the role of BMP2 and the MAPK/p38 signaling pathway in tumor growth and angiogenesis. RESULTS: BMP2 was highly expressed in HCC. Over-expression of BMP2 was found to accelerate cell proliferation, migration, invasion, microvascular density, and angiogenesis and decrease cell apoptosis in vitro and in vivo. BMP2 silencing exhibited inhibitory effects on HCC cell invasion and angiogenesis. The co-culture system illustrated that HepG2 cells secreted BMP2 in ECV304, and silenced BMP2 in HepG2 cells resulted in the inactivation of the MAPK/p38 signaling pathway, thus suppressing cancer progression, tumor growth, and angiogenesis in HCC. CONCLUSION: Taken together, the key findings of this study propose that silencing of BMP2 inhibits angiogenesis and tumor growth in HCC, highlighting BMP2 silencing as a potential strategy for the treatment of HCC.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Carcinoma, Hepatocellular/pathology , Endothelial Cells/metabolism , Liver Neoplasms/pathology , Signal Transduction , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Female , Hep G2 Cells , Humans , Imidazoles/pharmacology , Liver Neoplasms/blood supply , Liver Neoplasms/metabolism , Male , Mice , Mice, Nude , Middle Aged , Neovascularization, Pathologic , Pyrimidines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Cell Death Dis ; 10(8): 551, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320612

ABSTRACT

Retinoic acid (RA), an active derivative of vitamin A, is critical for the neural system development. During the neural development, the RA/RA receptor (RAR) pathway suppresses BMP signaling-mediated proliferation and differentiation of neural progenitor cells. However, how the stability of RAR is regulated during neural system development and how BMP pathway genes expression in neural tissue from human fetuses affected with neural tube defects (NTDs) remain elusive. Here, we report that FBXO30 acts as an E3 ubiquitin ligase and targets RARγ for ubiquitination and proteasomal degradation. In this way, FBXO30 positively regulates BMP signaling in mammalian cells. Moreover, RA treatment leads to suppression of BMP signaling by reducing the level of FBXO30 in mammalian cells and in mouse embryos with NTDs. In samples from human NTDs with high levels of retinol, downregulation of BMP target genes was observed, along with aberrant FBXO30 levels. Collectively, our results demonstrate that RARγ levels are controlled by FBXO30-mediated ubiquitination and that FBXO30 is a key regulator of BMP signaling. Furthermore, we suggest a novel mechanism by which high-retinol levels affect the level of FBXO30, which antagonizes BMP signaling during early stage development.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , F-Box Proteins/metabolism , Neural Tube Defects/metabolism , Receptors, Retinoic Acid/metabolism , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/pharmacology , Disease Models, Animal , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , F-Box Proteins/genetics , Female , Fetus/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , HEK293 Cells , Humans , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Neural Tube Defects/embryology , Neural Tube Defects/genetics , Protein Binding , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Signal Transduction/genetics , Tretinoin/pharmacology , Ubiquitination/drug effects , Ubiquitination/genetics , Retinoic Acid Receptor gamma
10.
Cell Death Dis ; 10(5): 350, 2019 04 25.
Article in English | MEDLINE | ID: mdl-31024000

ABSTRACT

The mechanism of pathological osteogenesis in Ankylosing spondylitis (AS) is largely unknown. Our previous studies demonstrated that the imbalance between BMP-2 and Noggin secretion induces abnormal osteogenic differentiation of marrow-derived mesenchymal stem cells (MSCs) from AS patients in a two-dimensional culture environment. In this study, HA/ß-TCP scaffolds were further used as a three-dimensional (3D) biomimetic culture system to mimic the bone microenvironment in vivo to determine the abnormal osteogenic differentiation of AS-MSCs. We demonstrated that when cultured in HA/ß-TCP scaffolds, AS-MSCs had a stronger osteogenic differentiation capacity than that of MSCs from healthy donors (HD-MSCs) in vitro and in vivo. This dysfunction resulted from BMP2 overexpression in AS-MSCs, which excessively activated the Smad1/5/8 and ERK signalling pathways and finally led to enhanced osteogenic differentiation. Both the signalling pathway inhibitors and siRNAs inhibiting BMP2 expression could rectify the enhanced osteogenic differentiation of AS-MSCs. Furthermore, BMP2 expression in ossifying entheses was significantly higher in AS patients. In summary, our study demonstrated that AS-MSCs possess enhanced osteogenic differentiation in HA/ß-TCP scaffolds as a 3D biomimetic microenvironment because of BMP2 overexpression, but not Noggin. These results provide insights into the mechanism of pathological osteogenesis, which can aid in the development of niche-targeting medications for AS.


Subject(s)
Cell Differentiation , Culture Techniques/methods , Mesenchymal Stem Cells/metabolism , Osteogenesis , Spondylitis, Ankylosing/pathology , Biomimetic Materials/chemistry , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Calcium Phosphates/chemistry , Cell Proliferation , Cells, Cultured , Durapatite/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mesenchymal Stem Cells/cytology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Spondylitis, Ankylosing/metabolism , Tissue Scaffolds/chemistry
11.
FASEB J ; 33(6): 7213-7224, 2019 06.
Article in English | MEDLINE | ID: mdl-30865837

ABSTRACT

MicroRNAs (miRNAs) crucially modulate fundamental biologic processes such as angiogenesis. In the present study, we focused on the molecular function of miRNA-370-3p (miR-370) in regulating the angiogenic activity of endothelial cells (ECs). Transfection with miR-370 mimic (miR-370m) significantly inhibited the sprouting of human dermal microvascular EC (HDMEC) and HUVEC spheroids and mouse aortic rings, whereas miR-370 inhibitor (miR-370i) promoted sprout formation. Additional in vitro assays demonstrated the pleiotropic inhibitory effects of miR-370m on HDMEC proliferation, migration, and tube formation. Moreover, Matrigel plugs containing miR-370m-transfected HDMECs exhibited a reduced microvessel density after implantation into CD1 nude mice when compared with controls. In contrast, miR-370i exerted proangiogenic effects. Mechanistic analyses revealed that miR-370 directly targets smoothened (SMO) and down-regulates bone morphogenetic protein (BMP)-2 expression in HDMECs. Accordingly, inhibition of SMO by cyclopamine reversed miR-370i-induced HDMEC proliferation and migration. In addition, BMP-2 treatment counteracted miR-370m-suppressed tube formation of HDMECs, whereas blockade of BMP-2 with neutralizing antibody significantly inhibited miR-370i-induced tube formation. Taken together, these novel findings indicate that miR-370 is a potent inhibitor of angiogenesis, which directly targets SMO and BMP-2.-Gu, Y., Becker, V., Zhao, Y., Menger, M. D., Laschke, M. W. miR-370 inhibits the angiogenic activity of endothelial cells by targeting smoothened (SMO) and bone morphogenetic protein (BMP)-2.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Endothelial Cells/metabolism , MicroRNAs/physiology , Neovascularization, Physiologic/genetics , Smoothened Receptor/physiology , Animals , Aorta , Bone Morphogenetic Protein 2/antagonists & inhibitors , Capillaries/cytology , Cell Division , Cell Movement , Cells, Cultured , Endothelial Cells/transplantation , Fibroblasts , Human Umbilical Vein Endothelial Cells , Humans , Keratinocytes , Male , Mice , Mice, Nude , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects , Organ Culture Techniques , Osteoblasts , RNA Interference , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Skin/blood supply , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/genetics , Spheroids, Cellular , Transfection , Veratrum Alkaloids/pharmacology
12.
Muscle Nerve ; 59(4): 501-508, 2019 04.
Article in English | MEDLINE | ID: mdl-30623463

ABSTRACT

INTRODUCTION: Muscle precursor cells (MPC) are integral to the maintenance of skeletal muscle and have recently been implicated in playing a role in bone repair. The primary objective of this study was to understand better the role of oxidative stress during the osteogenic differentiation of MPCs. METHODS: Muscle precursor cells were treated with various combinations of ascorbic acid (AA), bone morphogenetic protein (BMP)-2, and either a superoxide dismutase analog (4-hydroxy-TEMPO [TEMPOL]) or polyethyleneglycol-conjugated catalase. Muscle precursor cell proliferation and differentiation were determined, and alkaline phosphatase activity was measured as an index of osteogenic differentiation. RESULTS: After treatment with 200 µM AA, superoxide was increased 1.5-fold, whereas AA in combination with 100 ng/ml BMP-2 did not increase alkaline phosphatase (ALP) activity. When cells were treated with TEMPOL in combination with 100 ng/ml BMP-2 and 200 µM AA, ALP activity significantly increased. DISCUSSION: These data suggest that increasing oxidative stress with AA induces sublethal oxidative stress that prevents BMP-2-induced osteogenic differentiation of MPCs. Muscle Nerve 59:501-508, 2019.


Subject(s)
Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation/drug effects , Muscle, Skeletal/drug effects , Myoblasts/drug effects , Osteogenesis/drug effects , Alkaline Phosphatase/analysis , Alkaline Phosphatase/metabolism , Animals , Catalase/pharmacology , Cyclic N-Oxides/pharmacology , Male , Mesenchymal Stem Cells , Oxidative Stress , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Spin Labels
13.
J Periodontal Res ; 54(3): 300-309, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30511378

ABSTRACT

OBJECTIVES: The inhibitory action of the superficial gingival connective tissues may limit the regenerative potential of alveolar bone in periodontal therapy or dental implant applications. The aims of this study were to investigate the hypothesis that gingival fibroblasts (GF) can inhibit bone morphogenetic protein (BMP)-induced osteoblastic differentiation, to determine their expression of BMP inhibitors, and finally to determine whether reduction of these inhibitors can relieve suppression of osteoblastic differentiation. METHODS: Gingival fibroblasts were co-cultured either directly or indirectly with calvarial osteoblasts to assess alkaline phosphatase inhibitory activity, a marker of osteoblastic differentiation. To test total BMP-inhibitory activity of rat GF, conditioned media (GFCM) were collected from cultures. ROS 17/2.8 osteoblastic cells were stimulated with BMP2, together with GFCM. Inhibitor expression was tested using RT-qPCR, Western blotting and in situ hybridization. Removal of inhibitors was carried out using immunoprecipitation beads. RESULTS: Co-culture experiments showed GF-secreted factors that inhibit BMP-stimulated ALP activity. 10 ng/ml BMP2 increased alkaline phosphatase expression in ROS cells by 41%. GFCM blocked BMP activity which was equivalent to the activity of 100 ng/ml Noggin, a well-described BMP inhibitor. Cultured gingival fibroblasts constitutively expressed BMP antagonist genes from the same subfamily, Grem1, Grem2 and Nbl1 and the Wnt inhibitor Sfrp1. Gremlin1 (6.7 × reference gene expression) had highest levels of basal expression. ISH analysis showed Gremlin1 expression was restricted to the inner half of the gingival lamina propria and the PDL. Removal of Gremlin1 protein from GFCM eliminated the inhibitory effect of GFCM on ALP activity in ROS cells. Subsequent addition of recombinant Gremlin1 restored the inhibitory activity. CONCLUSIONS: Factors secreted by gingival fibroblasts inhibit BMP-induced bone formation and a range of BMP inhibitors are constitutively expressed in gingival connective tissues. These inhibitors, particularly Gremlin1, may limit coronal alveolar bone regenerative potential during oral and periodontal surgery.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Cell Differentiation , Fibroblasts/physiology , Gingiva/cytology , Osteoblasts/physiology , Osteogenesis , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Alveolar Process/physiology , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Regeneration/genetics , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques , Cytokines , Fibroblasts/metabolism , Male , Nerve Tissue Proteins/metabolism , Osteogenesis/drug effects , Proteins/metabolism , Rats, Wistar
14.
Drug Des Devel Ther ; 12: 3419-3430, 2018.
Article in English | MEDLINE | ID: mdl-30349195

ABSTRACT

INTRODUCTION: Alcoholism can lead to low mineral density, compromised regenerative bone capacity and delayed osteointegration of dental implants. This may be partially attributed to the inhibitive effect of all-trans retinoic acid (ATRA), a metabolite of alcohol, on osteoblastogenesis. Our previous studies demonstrated that heterodimeric bone morphogenetic protein 2/7 (BMP2/7) was a more potent BMP than homodimeric BMP2 or BMP7, and could antagonize the inhibitive effect of ATRA to rescue osteoblastogenesis. MATERIALS AND METHODS: In this study, we compared the effectiveness of BMP2/7, BMP2 and BMP7 in restoring osteoblastogenesis of murine preosteoblasts upon inhibition with 1 µM ATRA, and we further analyzed the potential mechanisms. We measured the following parameters: cell viability, ALP, OCN, mineralization, the expression of osteogenic differentiation marker genes (Collagen I, ALP and OCN) and the expression of BMP signaling key genes (Dlx5, Runx2, Osterix and Smad1). RESULTS: BMP2/7 treatment alone induced significantly higher osteoblastogenesis compared to BMP2 and BMP7. When cells were treated by ATRA, BMP2/7 was superior only in rescuing cell viability and ALP activity, compared to BMP2 or BMP7. However, BMP2/7 was not superior to BMP2 or BMP7 in restoring OCN expression and extracellular mineralized nodules, or in rescuing expression of two key osteogenic genes, Dlx5 and Runx2. Irrespective of their dimeric types or potency, the selected BMPs could antagonize the inhibitory effect of ATRA on osteoblastogenesis. CONCLUSION: The presence of ATRA, BMP2/7 still induced significantly higher cell viability and early differentiation than the homodimers. However, ATRA significantly attenuated the advantages of BMP2/7 in inducing late and final osteoblastogenic differentiation over the homodimers.


Subject(s)
Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 7/antagonists & inhibitors , Osteoblasts/drug effects , Osteogenesis/drug effects , Tretinoin/pharmacology , 3T3 Cells , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 7/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Mice , Osteoblasts/cytology , Protein Multimerization , Structure-Activity Relationship
15.
Cell Physiol Biochem ; 48(6): 2399-2408, 2018.
Article in English | MEDLINE | ID: mdl-30121651

ABSTRACT

BACKGROUND/AIMS: High mobility group box 1 (Hmgb1) is associated with a variety of physiological processes including embryonic development, cell proliferation and differentiation, but little information is available regarding its biological role in decidualization. METHODS: In situ hybridization, real-time PCR, RNA interference, gene overexpression and MTS assay were used to analyze the spatiotemporal expression of Hmgb1 in mouse uterus during the pre-implantation period, and explore its function and regulatory mechanisms during uterine decidualization. RESULTS: Hmgb1 mRNA was obviously observed in uterine epithelium on day 2 and 3 of pregnancy, but its expression was scarcely detected on day 4 of pregnancy. With the onset of embryo implantation, abundant Hmgb1 expression was noted in the subluminal stromal cells around the implanting blastocyst at implantation sites. Meanwhile, the accumulation of Hmgb1 mRNA was visualized in the decidual cells. Hmgb1 advanced the proliferation of uterine stromal cells and induced the expression of prolactin family 8, subfamily a, member 2 (Prl8a2), a reliable differentiation marker for decidualization. In uterine stromal cells, cAMP analogue 8-Br-cAMP up-regulated the expression of Hmgb1, but the up-regulation was abrogated by protein kinase A (PKA) inhibitor H89. Silencing of Hmgb1 by specific siRNA impeded the induction of 8-Br-cAMP on Prl8a2. Further analysis evidenced that Hmgb1 was a critical mediator of Kruppel-like factor 5 (Klf5) function in stromal differentiation. Knockdown of bone morphogenetic protein 2 (Bmp2) prevented the up-regulation of Prl8a2 elicited by Hmgb1 overexpression, whereas addition of exogenous recombinant Bmp2 protein (rBmp2) reversed the repression of Hmgb1 siRNA on Prl8a2 expression. CONCLUSION: Hmgb1 may play an important role during mouse uterine decidualization.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , HMGB1 Protein/metabolism , Kruppel-Like Transcription Factors/metabolism , Prolactin/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Cell Proliferation/drug effects , Cells, Cultured , Embryo Implantation , Female , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Isoquinolines/pharmacology , Kruppel-Like Transcription Factors/genetics , Mice , Pregnancy , Prolactin/genetics , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Stromal Cells/cytology , Stromal Cells/metabolism , Sulfonamides/pharmacology , Up-Regulation/drug effects , Uterus/cytology
16.
Article in English | MEDLINE | ID: mdl-29977865

ABSTRACT

Palmitoylation has been recently identified as an important post-translational rheostat for controlling protein function in eukaryotes. However, the molecular machinery underlying palmitoylation remains unclear in the neglected tropical parasite, Leishmania donovani. Herein, we have identified a catalog of 20 novel palmitoyl acyltransferases (PATs) and characterized the promastigote-specific PAT (LdPAT4) containing the canonical Asp-His-His-Cys (DHHC) domain. Immunofluorescence analysis using in-house generated LdPAT4-specific antibody demonstrated distinct expression of LdPAT4 in the flagellar pocket of promastigotes. Using metabolic labeling-coupled click chemistry method, the functionality of this recombinant enzyme could be authenticated in E. coli strain expressing LdPAT4-DHHC domain. This was evident by the cellular uptake of palmitic acid analogs, which could be successfully inhibited by 2-BMP, a PAT-specific inhibitor. Using CSS-Palm based in-silico proteomic analysis, we could predict up to 23 palmitoylated sites per protein in the promastigotes, and further identify distinctive palmitoylated protein clusters involved in microtubule assembly, flagella motility and vesicular trafficking. To highlight, proteins such as Flagellar Member proteins (FLAM1, FLAM5), Intraflagellar Transport proteins (IFT88), and flagellar motor assembly proteins including the Dynein family were found to be enriched. Furthermore, analysis of global palmitoylation in promastigotes using Acyl-biotin exchange purification identified a set of S-palmitoylated proteins overlapping with the in-silico proteomics data. The attenuation of palmitoylation using 2-BMP demonstrated several phenotypic alterations in the promastigotes including distorted morphology, reduced motility (flagellar loss or slow flagellar beating), and inefficient invasion of promastigotes to host macrophages. These analyses confirm the essential role of palmitoylation in promastigotes. In summary, the findings suggest that LdPAT4 acts as a functional acyltransferase that can regulate palmitoylation of proteins involved in parasite motility and invasion, thus, can serve as a potential target for designing chemotherapeutics in Visceral Leishmaniasis.


Subject(s)
Acyltransferases/chemistry , Leishmania donovani/enzymology , Lipoylation/physiology , Acyltransferases/genetics , Acyltransferases/isolation & purification , Base Sequence , Bone Morphogenetic Protein 2/antagonists & inhibitors , Escherichia coli/genetics , Gene Expression , Gene Ontology , Genes, Protozoan , Humans , Leishmania donovani/genetics , Leishmania donovani/pathogenicity , Macrophages/parasitology , Molecular Docking Simulation , Protein Transport , Proteomics/methods , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins , Sequence Alignment , Transforming Growth Factor beta/antagonists & inhibitors
17.
Arterioscler Thromb Vasc Biol ; 38(9): 2148-2159, 2018 09.
Article in English | MEDLINE | ID: mdl-30026273

ABSTRACT

Objective- Calcific aortic valve disease is the most prevalent valvulopathy in Western countries. An unanticipated pathogenetic clue involving IFN (interferon) was disclosed by the finding of constitutive type I IFN activity associated with aortic valve calcification in children with the atypical Singleton-Merten syndrome. On this basis, the role of type I IFN on inflammation and calcification in human aortic valve interstitial cells (AVIC) was examined. Approach and Results- IFN-α was weakly proinflammatory but potentiated lipopolysaccharide-mediated activation of NF (nuclear factor)-κB and the ensuing induction of proinflammatory molecules in human AVIC. Stimulation with IFN-α and in combination with lipopolysaccharide promoted osteoblast-like differentiation characterized by increased osteoblastic gene expression, BMP (bone morphogenetic protein)-2 secretion, and ectopic phosphatase activity. Sex differences were observed. Likewise, IFN-α treatment of human AVICs in osteogenic medium resulted in increased formation of calcific nodules. Strikingly, IFN-α-mediated calcification was significantly higher in AVICs from males, and was blocked by tofacitinib, a JAK (Janus kinase) inhibitor, and by a BMP antagonist. A female-specific protective mechanism involving the activation of PI3K-Akt (protein kinase B) pathways and cell survival was disclosed. Females exhibited higher levels of BCL2 in valve cells and tissues and lower annexin V staining on cell stimulation. Conclusions- IFN-α acts as a proinflammatory and pro-osteogenic cytokine in AVICs, its effects being potentiated by lipopolysaccharide. Results also uncovered sex differences with lower responses in female AVICs and sex-specific mechanisms involving apoptosis. Data point to JAK/STAT (signal transducer and activator of transcription) system as a potential therapeutic target for calcific aortic valve disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Calcinosis/metabolism , Interferon Type I/drug effects , Interferon Type I/metabolism , Janus Kinase Inhibitors/pharmacology , Aortic Valve/cytology , Aortic Valve/metabolism , Aortic Valve Stenosis/pathology , Apoptosis , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/metabolism , Calcinosis/pathology , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Female , Humans , Lipopolysaccharides/pharmacology , Male , NF-kappa B/metabolism , Osteoblasts/physiology , Piperidines/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT Transcription Factors/metabolism , Sex Factors , Signal Transduction , Toll-Like Receptor 4/metabolism
18.
Acta Biomater ; 74: 222-235, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29778895

ABSTRACT

Induced pluripotent stem cells (iPSCs) are regarded as a new cell source for regenerative medicine. Recent advances in tissue engineering have brought to light the therapeutic application of induced pluripotent stem cells (iPSCs) in bone defect repair. However, a safe and efficient way to differentiate iPSCs into osteogenic lineage remains to be a major challenge. Here we describe an approach using anti-BMP2 antibodies (Abs) to mediate osteogenic differentiation of iPSC-derived mesenchymal stromal cells (iMSCs). We first proved that 3G7 (an anti-BMP2 Ab) not only bound to BMP2, but also allowed the bound BMP2 to engage the BMP2 receptors on iMSCs. Subcutaneous implantation sites loaded with iMSCs + 3G7 group showed significant bone formation and vascularization in mice while those sites with exogenous BMP2 exhibited dystrophic calcification and significantly lower vascularization. Our in vitro study demonstrated that the anti-BMP2 Ab/BMP2 immune complex were capable of dictating the acquisition of osteogenic phenotype of iMSCs and subsequent mineralization. The study provided the first evidence of antibody-mediated differentiation of iMSCs and osseous regeneration in vivo. This novel strategy takes full advantage of the endogenous bioactive molecules for osseous regeneration and its potential therapeutic application is promising. STATEMENT OF SIGNIFICANCE: Induced pluripotent stem cells (iPSCs) and its derived cells hold significant promise for the treatment of bone defects. In present study, we carried out the concept of antibody-mediated bone regeneration into the iPSC research for the first time. We demonstrated that anti-BMP2 Ab/BMP2 immune complex was capable of promoting osteogenic differentiation of iPSC-derived MSCs (iMSCs), likely through the classical BMP2/Smad1/Runx2 pathway. Subcutaneous co-delivery of iMSCs and anti-BMP2 Abs resulted in significant bone formation and vascularization. These findings suggested antibody mediated osteogenic differentiation may be a favorable approach for iPSC-based bone tissue engineering.


Subject(s)
Antibodies/pharmacology , Bone Morphogenetic Protein 2/antagonists & inhibitors , Calcification, Physiologic/drug effects , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Regeneration/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Mice
19.
Exp Neurol ; 299(Pt B): 289-298, 2018 01.
Article in English | MEDLINE | ID: mdl-29055717

ABSTRACT

Plexiform neurofibromas (PNs), which may be present at birth in up to half of children with type 1 neurofibromatosis (NF1), can cause serious loss of function, such as quadriparesis, and can undergo malignant transformation. Surgery is the first line treatment although the invasive nature of these tumors often prevents complete resection. Recent clinical trials have shown promising success for some drugs, notably selumetinib, an inhibitor of MAP kinase kinase (MEK). We have developed three-dimensional (3D) cell culture models of immortalized cells from NF1 PNs and of control Schwann cells (SCs) that we believe mimic more closely the in vivo condition than conventional two-dimensional (2D) cell culture. Our goal is to facilitate pre-clinical identification of potential targeted therapeutics for these tumors. Three drugs, selumetinib (a MEK inhibitor), picropodophyllin (an IGF-1R inhibitor) and LDN-193189 (a BMP2 inhibitor) were tested with dose-response design in both 2D and 3D cultures for their abilities to block net cell growth. Cell lines grown in 3D conditions showed varying degrees of resistance to the inhibitory actions of all three drugs. For example, control SCs became resistant to growth inhibition by selumetinib in 3D culture. LDN-193189 was the most effective drug in 3D cultures, with only slightly reduced potency compared to the 2D cultures. Characterization of these models also demonstrated increased proteolysis of collagen IV in the matrix by the PN driver cells as compared to wild-type SCs. The proteolytic capacity of the PN cells in the model may be a clinically significant property that can be used for testing the ability of drugs to inhibit their invasive phenotype.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques , Drug Screening Assays, Antitumor/methods , Neurofibroma, Plexiform/pathology , Benzimidazoles/pharmacology , Bone Morphogenetic Protein 2/antagonists & inhibitors , Cells, Cultured , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Genes, Neurofibromatosis 1 , Genes, Reporter , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , Molecular Targeted Therapy , Neurofibroma, Plexiform/drug therapy , Neurofibroma, Plexiform/genetics , Neurofibromatosis 1/pathology , Phenotype , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Schwann Cells/cytology , Transduction, Genetic , Tumor Cells, Cultured , Red Fluorescent Protein
20.
Int J Mol Med ; 41(1): 51-60, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115380

ABSTRACT

The present study aimed to assess the effects of therapy with adiponectin (APN) gene-modified adipose-derived stem cells (ADSCs) on pulmonary arterial hypertension (PAH) in rats and the underlying cellular and molecular mechanisms. ADSCs were successfully isolated from the rats and characterized. ADSCs were effectively infected with the green fluorescent protein (GFP)-empty (ADSCs-V) or the APN-GFP (ADSCs-APN) lentivirus and the APN expression was evaluated by ELISA. Sprague-Dawley rats were administered monocrotaline (MCT) to develop PAH. The rats were treated with MCT, ADSCs, ADSCs-V and ADSCs-APN. Then ADSCs-APN in the lung were investigated by confocal laser scanning microscopy and western blot analysis. Engrafted ADSCs in the lung were located around the vessels. Mean pulmonary arterial pressure (mPAP) and the right ventricular hypertrophy index (RVHI) in the ADSCs-APN-treated mice were significantly decreased as compared with the ADSCs and ADSCs-V treatments. Pulmonary vascular remodeling was assessed. Right ventricular (RV) function was evaluated by echocardiography. We found that pulmonary vascular remodeling and the parameters of RV function were extensively improved after ADSCs-APN treatment when compared with ADSCs and ADSCs-V treatment. Pulmonary artery smooth muscle cells (PASMCs) were isolated from the PAH rats. The antiproliferative effect of APN on PASMCs was assayed by Cell Counting Kit-8. The influence of APN and specific inhibitors on the levels of bone morphogenetic protein (BMP), adenosine monophosphate activated protein kinase (AMPK), and small mothers against decapentaplegia (Smad) pathways was detected by western blot analysis. We found that APN suppressed the proliferation of PASMCs isolated from the PAH rats by regulating the AMPK/BMP/Smad pathway. This effect was weakened by addition of the AMPK inhibitor (compound C) and BMP2 inhibitor (noggin). Therefore, combination treatment with ADSCs and APN effectively attenuated PAH in rats by inhibiting PASMC proliferation and regulating the AMPK/BMP/Smad pathway.


Subject(s)
Adiponectin/administration & dosage , Cell Proliferation/genetics , Hypertension, Pulmonary/therapy , Myocytes, Smooth Muscle/drug effects , AMP-Activated Protein Kinase Kinases , Adipocytes/transplantation , Adiponectin/genetics , Adipose Tissue/cytology , Adipose Tissue/transplantation , Animals , Bone Morphogenetic Protein 2/antagonists & inhibitors , Bone Morphogenetic Protein 2/genetics , Cell Proliferation/drug effects , Genetic Therapy , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Lentivirus/genetics , Myocytes, Smooth Muscle/metabolism , Protein Kinases/drug effects , Protein Kinases/genetics , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...