Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Cell Tissue Res ; 396(1): 103-117, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38403744

ABSTRACT

The formation of the epiretinal fibrotic membrane by retinal pigment epithelial (RPE) cells is a primary pathological change for proliferative vitreoretinopathy (PVR). Bone morphogenetic protein 6 (BMP6) is an antifibrogenic factor in various cells. To date, it is still unknown whether BMP6 can interfere with the fibrogenesis of RPE cells during the progression of PVR. This work aimed to address the relationship between BMP6 and transforming growth factor-ß2 (TGF-ß2)-elicited fibrogenesis of RPE cells, an experimental model for studying PVR in vitro. The BMP6 level was down-regulated, while the TGF-ß2 level was up-regulated in the vitreous humor of PVR patients. The BMP6 level was down-regulated in human RPE cells challenged with TGF-ß2. The treatment of RPE cells with TGF-ß2 resulted in significant increases in proliferation, migration, epithelial-to-mesenchymal transition (EMT), and extracellular matrix (ECM) remodelling. These effects were found to be inhibited by the overexpression of BMP6 or exacerbated by the knockdown of BMP6. BMP6 overexpression reduced the phosphorylation of p38 and JNK in TGF-ß2-stimulated RPE cells, while BMP6 knockdown showed the opposite effects. The inhibition of p38 or JNK partially reversed the BMP6-silencing-induced promoting effects on TGF-ß2-elicited fibrogenesis in RPE cells. Taken together, BMP6 demonstrates the ability to counteract the proliferation, migration, EMT, and ECM remodelling of RPE cells induced by TGF-ß2. This is achieved through the regulation of the p38 and JNK MAPK pathways. These findings imply a potential connection between BMP6 and PVR, and highlight the potential application of BMP6 in therapeutic interventions for PVR.


Subject(s)
Vitreoretinopathy, Proliferative , Humans , Vitreoretinopathy, Proliferative/drug therapy , Vitreoretinopathy, Proliferative/metabolism , Vitreoretinopathy, Proliferative/pathology , Retinal Pigment Epithelium , Transforming Growth Factor beta2/pharmacology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/therapeutic use , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/therapeutic use , Epithelial-Mesenchymal Transition , Epithelial Cells/metabolism , Retinal Pigments/metabolism , Retinal Pigments/pharmacology , Retinal Pigments/therapeutic use , Cell Movement
2.
J Assist Reprod Genet ; 41(1): 31-48, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37930517

ABSTRACT

PURPOSE: To evaluate whether PTX3 is differentially expressed in the granulosa lutein cells derived from women with PCOS and whether BMP6 can regulate the expression of PTX3 in hGL cells. METHODS: The expression levels of BMP6 and PTX3 in granulosa lutein cells were evaluated by RT-qPCR. The correlation between the expression levels of BMP6 /PTX3 and oocyte quality indexes were analyzed using clinical samples. The cells were incubated with BMP6 at different concentrations and times to check the expression of PTX3 in KGN cells. TGF-ß type I inhibitors and small interfering RNA targeting ALK2/3/6,SMAD1/5/8 and SMAD4 were used to study the involvement of SMAD dependent pathways in KGN cells. RESULTS: The levels of BMP6 in hGL cells were negatively correlated with the corresponding oocyte maturation rate and high-quality embryo rate, whereas the levels of PTX3 were positively correlated with the corresponding oocyte maturation rate in PCOS. Additionally, the in vitro cell cultured results showed BMP6 significantly inhibited the expression of PTX3 in KGN cells. Furthermore, using a dual inhibition approach (kinase inhibitors and small interfering RNAs), we identified the ALK2/ALK3 type I receptors and BMPR2/ACVR2A type II receptors and the downstream SMAD1/SMAD5-SMAD4 signaling pathway were responsible for the BMP6-induced cellular activities in KGN cells. CONCLUSIONS: The suppressive effect of BMP6 on PTX3 was mediated by ALK2/ALK3 type I receptors and BMPR2/ACVR2A type II receptors in granulosa cells through the SMAD1/5-SMAD4 dependent signaling pathway in PCOS.Our findings provides new insights into the understanding of the pathogenesis of PCOS-related ovulatory disorders.


Subject(s)
C-Reactive Protein , Luteal Cells , Polycystic Ovary Syndrome , Serum Amyloid P-Component , Female , Humans , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein Receptors, Type II/genetics , Down-Regulation/genetics , Granulosa Cells/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism
3.
J Gene Med ; 23(3): e3311, 2021 03.
Article in English | MEDLINE | ID: mdl-33527563

ABSTRACT

BACKGROUND: Fragments of subcutaneous adipose tissue that have been genetically modified to express bone morphogenetic protein-2 (BMP-2) regenerate large segmental osseous lesions in rodents. Gene-activated adipose tissue can be implanted into osseous defects without prior cell extraction and cell culture. The present study aimed to explore whether the heterodimers BMP-2/6 or BMP-2/7 exceed the osteoinductive effect of BMP-2 on adipose tissue. METHODS: In an in vitro tissue culture system, freshly harvested rat subcutaneous adipose tissue was cultivated in the presence of either BMP-2 or BMP-2/6 or BMP-2/7 at a high (200 ng/ml) and low (50 ng/ml) concentration. Gene expression analysis as well as histological and immunohistochemical methods were applied to test for osteoinduction. RESULTS: A concentration of 200 ng/ml of homodimeric BMP-2 induced osteogenic differentiation most potently, showing more calcification and a higher expression level of bone markers than both concentrations of BMP-2/6 or -2/7. A concentration of 50 ng/ml of BMP-2 was a significantly stronger osteogenic inducer than both concentrations of BMP-2/6 and the low concentration of BMP-2/7. The most potent heterodimeric driver of osteoinduction was BMP-2/7 at a high concentration, demonstrating effects similar to those of BMP-2 at a low concentration. CONCLUSIONS: Homodimeric BMP-2 evoked osteoinduction within adipose tissue more potently and at a lower concentration than heterodimeric BMP-2/6 or BMP-2/7. This result agrees well with the fact that it might be easier to translate adipose grafts activated by homodimeric BMP-2 clinically. Preclinical in vivo gene transfer studies are necessary to confirm the results of the present study.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Bone Regeneration/drug effects , Osteogenesis/drug effects , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Animals , Biomarkers/metabolism , Gene Expression/drug effects , Humans , Male , Rats , Rats, Inbred F344 , Recombinant Proteins/pharmacology , Tissue Culture Techniques
4.
Biol Reprod ; 101(2): 445-456, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31210269

ABSTRACT

Connective tissue growth factor (also known as CTGF or CCN2) is a secreted matricellular protein that belongs to the CCN family. With wide-ranging biological activities and tissue expression patterns, CTGF plays a critical role in regulating various cellular functions. In the female reproductive system, CTGF is highly expressed in granulosa cells in growing ovarian follicles and is involved in the regulation of follicular development, ovulation, and luteal function. In the mammalian ovary, bone morphogenetic protein 6 (BMP6) is an important intraovarian modulator of follicular development. In this study, we demonstrated that BMP6 treatment significantly increased the expression of CTGF in both primary and immortalized human granulosa cells. Using both pharmacological inhibitors and Small interfering RNA-mediated knockdown approaches, we showed that ALK2 and ALK3 type I receptors are required for BMP6-induced cellular activities. Furthermore, this effect is most likely mediated by a Sma- and Mad-related protein (SMAD)-dependent pathway. Our studies provide novel insight into the molecular mechanisms by which an intraovarian growth factor affects the production of another factor via a paracrine effect in human granulosa cells.


Subject(s)
Bone Morphogenetic Protein 6/pharmacology , Connective Tissue Growth Factor/metabolism , Granulosa Cells/metabolism , Smad Proteins/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Connective Tissue Growth Factor/genetics , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Granulosa Cells/drug effects , Humans , Luteinization , Luteinizing Hormone , Signal Transduction , Smad Proteins/genetics
5.
Am J Hematol ; 94(11): 1227-1235, 2019 11.
Article in English | MEDLINE | ID: mdl-31400017

ABSTRACT

The erythroferrone (ERFE) is the erythroid regulator of hepatic iron metabolism by suppressing the expression of hepcidin. Congenital dyserythropoietic anemia type II (CDAII) is an inherited hyporegenerative anemia due to biallelic mutations in the SEC23B gene. Patients with CDAII exhibit marked clinical variability, even among individuals sharing the same pathogenic variants. The ERFE expression in CDAII is increased and related to abnormal erythropoiesis. We identified a recurrent low-frequency variant, A260S, in the ERFE gene in 12.5% of CDAII patients with a severe phenotype. We demonstrated that the ERFE-A260S variant leads to increased levels of ERFE, with subsequently marked impairment of iron regulation pathways at the hepatic level. Functional characterization of ERFE-A260S in the hepatic cell system demonstrated its modifier role in iron overload by impairing the BMP/SMAD pathway. We herein described for the first time an ERFE polymorphism as a genetic modifier variant. This was with a mild effect on disease expression, under a multifactorial-like model, in a condition of iron-loading anemia due to ineffective erythropoiesis.


Subject(s)
Anemia, Dyserythropoietic, Congenital/genetics , Bone Morphogenetic Proteins/physiology , Iron Overload/etiology , Liver/metabolism , Peptide Hormones/genetics , Signal Transduction/genetics , Smad Proteins/physiology , Adolescent , Adult , Anemia, Dyserythropoietic, Congenital/complications , Anemia, Dyserythropoietic, Congenital/metabolism , Blood Transfusion , Bone Morphogenetic Protein 6/pharmacology , Cell Line , Child , Erythropoiesis/genetics , Female , Genetic Association Studies , Hepcidins/biosynthesis , Hepcidins/blood , Hepcidins/genetics , Humans , Male , Peptide Hormones/blood , Peptide Hormones/pharmacology , Peptide Hormones/physiology , Recombinant Proteins/pharmacology , Severity of Illness Index , Smad Proteins/biosynthesis , Smad Proteins/genetics , Young Adult
6.
Nature ; 498(7455): 492-6, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23748444

ABSTRACT

Cerebral cavernous malformation (CCM) is a vascular dysplasia, mainly localized within the brain and affecting up to 0.5% of the human population. CCM lesions are formed by enlarged and irregular blood vessels that often result in cerebral haemorrhages. CCM is caused by loss-of-function mutations in one of three genes, namely CCM1 (also known as KRIT1), CCM2 (OSM) and CCM3 (PDCD10), and occurs in both sporadic and familial forms. Recent studies have investigated the cause of vascular dysplasia and fragility in CCM, but the in vivo functions of this ternary complex remain unclear. Postnatal deletion of any of the three Ccm genes in mouse endothelium results in a severe phenotype, characterized by multiple brain vascular malformations that are markedly similar to human CCM lesions. Endothelial-to-mesenchymal transition (EndMT) has been described in different pathologies, and it is defined as the acquisition of mesenchymal- and stem-cell-like characteristics by the endothelium. Here we show that endothelial-specific disruption of the Ccm1 gene in mice induces EndMT, which contributes to the development of vascular malformations. EndMT in CCM1-ablated endothelial cells is mediated by the upregulation of endogenous BMP6 that, in turn, activates the transforming growth factor-ß (TGF-ß) and bone morphogenetic protein (BMP) signalling pathway. Inhibitors of the TGF-ß and BMP pathway prevent EndMT both in vitro and in vivo and reduce the number and size of vascular lesions in CCM1-deficient mice. Thus, increased TGF-ß and BMP signalling, and the consequent EndMT of CCM1-null endothelial cells, are crucial events in the onset and progression of CCM disease. These studies offer novel therapeutic opportunities for this severe, and so far incurable, pathology.


Subject(s)
Disease Progression , Epithelial-Mesenchymal Transition , Hemangioma, Cavernous, Central Nervous System/pathology , Animals , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein 6/pharmacology , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Hemangioma, Cavernous, Central Nervous System/genetics , Humans , KRIT1 Protein , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Up-Regulation
7.
J Mater Sci Mater Med ; 31(1): 4, 2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31832785

ABSTRACT

In the present study, a chitosan-based, multifunctional and double-faced barrier membrane was developed for the periodontitis therapy. The porous surface of the membrane was coated with bone-like hydroxyapatite (HA) produced by microwave-assisted biomimetic method and enriched with bone morphogenetic factor 6 (BMP-6) to enhance the bioactivity of chitosan. This surface of the membrane was designed to be in contact with the hard tissue that was damaged due to periodontitis. Otherwise the nonporous surface of membrane, which is in contact with the inflammatory soft tissue, was coated with electrospun polycaprolactone (PCL) fibers to prevent the migration of epithelial cells to the defect area. PrestoBlue, Scanning Electron Microscope (SEM) and real-time PCR results demonstrated that while porous surface of the membrane was enhancing the proliferation and differentiation of MC3T3-E1 preosteoblasts, nonporous surface of membrane did not allow migration of epithelial Madine Darby Bovine Kidney (MDBK) cells. The barrier membrane developed here is biodegradable and can be easily manipulated, has osteogenic activity and inactivity for epithelial cells. Thus, by implanting this membrane to the damaged periodontal tissue, bone regeneration will take place and integrity of periodontal tissues will be preserved.


Subject(s)
Bone Morphogenetic Protein 6/pharmacology , Chitosan/chemistry , Membranes, Artificial , Nanostructures/chemistry , Animals , Biocompatible Materials , Blood Vessel Prosthesis , Cattle , Cell Line , Cell Survival , Humans , Mice , Nanostructures/ultrastructure , Osteoblasts , Surface Properties , Tissue Scaffolds
8.
J Cell Sci ; 129(1): 206-18, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26598555

ABSTRACT

Several vascular disorders, such as aberrant angiogenesis, atherosclerosis and pulmonary hypertension, have been linked to dysfunctional BMP signaling. Vascular hyperpermeability via distortion of endothelial cell adherens junctions is a common feature of these diseases, but the role of BMPs in this process has not been investigated. BMP signaling is initiated by binding of ligand to, and activation of, BMP type I (BMPRI) and type II (BMPRII) receptors. Internalization of VE-cadherin as well as c-Src kinase-dependent phosphorylation have been implicated in the loosening of cell-cell contacts, thereby modulating vascular permeability. Here we demonstrate that BMP6 induces hyperpermeabilization of human endothelial cells by inducing internalization and c-Src-dependent phosphorylation of VE-cadherin. Furthermore, we show BMP-dependent physical interaction of VE-cadherin with the BMP receptor ALK2 (BMPRI) and BMPRII, resulting in stabilization of the BMP receptor complex and, thereby, the support of BMP6-Smad signaling. Our results provide first insights into the molecular mechanism of BMP-induced vascular permeability, a hallmark of various vascular diseases, and provide the basis for further investigations of BMPs as regulators of vascular integrity, both under physiological and pathophysiological conditions.


Subject(s)
Antigens, CD/metabolism , Bone Morphogenetic Protein 6/pharmacology , Cadherins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Signal Transduction/drug effects , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Bone Morphogenetic Protein Receptors/metabolism , Capillary Permeability/drug effects , Cell Membrane Permeability/drug effects , Endocytosis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Models, Biological , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins pp60(c-src)/metabolism
9.
Exp Dermatol ; 27(11): 1287-1293, 2018 11.
Article in English | MEDLINE | ID: mdl-30230035

ABSTRACT

Hypoxia-inducible factor-1α (HIF-1α) has been reported to be up-regulated in psoriatic epidermis, resulting in increased proliferation and abnormal differentiation of human keratinocytes (KCs). However, the role of HIF-1α in psoriatic epidermis, which is mainly composed of KCs, is poorly understood. Here, we show that morphogenic protein 6 (BMP6) is down-regulated when HIF-1α is upregulated in patients with psoriasis skin lesions. HIF-1α overexpression in primary human KCs promoted proliferation and inhibited terminal differentiation. Furthermore, HIF1-α repressed the expression of BMP6 by binding directly to the hypoxia-response element (HRE) in the BMP6 promotor region, which shows that BMP6 is a novel target gene of HIF-1α. We also found that HIF-1α-mediated BMP6 suppression could alter the proliferation status by modulating the expression levels of cell cycle regulatory proteins and also affect the early differentiation of KCs. Therefore, we suggest that HIF-1α-dependent BMP6 suppression has a critical role in the induction of hyper-proliferation and abnormal differentiation in psoriatic KCs.


Subject(s)
Bone Morphogenetic Protein 6/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Psoriasis/genetics , Antigens, Neoplasm/metabolism , Bone Morphogenetic Protein 6/pharmacology , Carbonic Anhydrase IX/metabolism , Cell Cycle/genetics , Cell Differentiation/drug effects , Cell Hypoxia , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation , Glucose Transporter Type 1/metabolism , Humans , Keratinocytes/physiology , Primary Cell Culture , Promoter Regions, Genetic , Psoriasis/metabolism , Transfection
10.
Cell Tissue Res ; 364(1): 125-35, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26475719

ABSTRACT

We have previously shown that the combined delivery of mesenchymal stem cells (MSCs), vascular endothelial growth factor (VEGF) and bone morphogenetic protein 6 (BMP-6) induces significantly more bone formation than that induced by the delivery of any single factor or a combination of any two factors. We now determine whether the exogenous addition of VEGF and BMP-6 is sufficient for bone healing when MSCs are not provided. Poly(lactic-co-glycolic acid) (PLAGA) microsphere-based three-dimensional scaffolds (P) were fabricated by thermal sintering of PLAGA microspheres. The scaffolds were chemically cross-linked with 200 ng recombinant human VEGF (P(VEGF)) or BMP-6 (P(BMP-6)) or both (P(VEGF+BMP-6)) by the EDC-NHS-MES method. Release of the proteins from the scaffolds was detected for 21 days in vitro which confirmed their comparable potential to supply the proteins in vivo. The scaffolds were delivered to a critical-sized mandibular defect created in 32 Sprague Dawley rats. Significant bone regeneration was observed only in rats with P(VEGF+BMP-6) scaffolds at weeks 2, 8 and 12 as revealed by micro-computer tomography. Vascular ingrowth was higher in the P(VEGF+BMP-6) group as seen by microfil imaging than in other groups. Trichrome staining revealed that a soft callus formed in P(VEGF), P(BMP-6) and P(VEGF+BMP-6) but not in P. MSCs isolated from rat femurs displayed expression of the bone-specific marker osteocalcin when cultured with P(VEGF), P(BMP-6), or P(VEGF+BMP-6) but not with P. Robust mineralization and increased alkaline phosphatase gene expression were seen in rat MSCs when cultured on P(VEGF+BMP-6) but not on P, P(VEGF), or P(BMP-6). Thus, unlike the delivery of VEGF or BMP-6 alone, the combined delivery of VEGF and BMP-6 to the bone defect significantly enhanced bone repair through the enhancement of angiogenesis and the differentiation of endogenously recruited MSCs into the bone repair site.


Subject(s)
Bone Morphogenetic Protein 6 , Lactic Acid , Mandibular Diseases/therapy , Mesenchymal Stem Cells/metabolism , Polyglycolic Acid , Tissue Scaffolds/chemistry , Vascular Endothelial Growth Factor A , Animals , Bone Morphogenetic Protein 6/chemistry , Bone Morphogenetic Protein 6/pharmacology , Humans , Lactic Acid/chemistry , Lactic Acid/pharmacology , Mandible/metabolism , Mandible/pathology , Mandibular Diseases/pathology , Mesenchymal Stem Cells/pathology , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Inbred F344 , Vascular Endothelial Growth Factor A/chemistry , Vascular Endothelial Growth Factor A/pharmacology
11.
Reprod Domest Anim ; 51(1): 59-68, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26660854

ABSTRACT

BMP-6 has been found to be important to ovarian cells and oocyte, as well as to uterus. Thus, this study investigated the effect of bone morphogenetic protein (BMP-6) and recombinant follicle-stimulating hormone (rFSH) alone or in combination on the in vitro culture (IVC) of isolated caprine secondary follicles (Experiment 1) and the mRNA levels for BMP receptors/Smad signalling pathway (BMPR1A, BMPR2, SMAD1, SMAD4, SMAD5, SMAD6, SMAD7 and SMAD8) in vivo and in vitro using BMP-6 (Experiment 2). Secondary follicles were cultured in αMEM(+) alone (control medium) or supplemented with BMP-6 at 1 or 10 ng/ml and rFSH alone or the combination of both BMP-6 concentrations and rFSH. The results from Experiment 1 showed that the antrum formation rate was higher in the BMP-6 at 1 ng/ml (p < 0.05) than in MEM. In Experiment 2, the mRNA expression for BMPR2, SMAD1, SMAD5 and SMAD6 was detected in non-cultured control and after in vitro culture (MEM and 1 ng/ml BMP-6); while the expression of SMAD7 and SMAD8 mRNA was only detected after IVC, SMAD4 was only detected in the BMP-6 at 1 ng/ml treatment. In conclusion, the low BMP-6 concentration positively influenced antrum formation and ensured normal mRNA expression for BMP receptor and Smads after IVC of caprine secondary follicles.


Subject(s)
Bone Morphogenetic Protein 6/pharmacology , Goats/physiology , Ovarian Follicle/drug effects , Ovarian Follicle/physiology , Signal Transduction/drug effects , Animals , Bone Morphogenetic Protein Receptors/genetics , Female , Follicle Stimulating Hormone/pharmacology , RNA, Messenger/analysis , Recombinant Proteins/pharmacology , Tissue Culture Techniques/veterinary
13.
Cell Biol Int ; 39(4): 400-10, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25492426

ABSTRACT

Mesenchymal-epithelial transition (MET) is an inevitable process for cellular reprogramming. MET could be induced by suppressing epithelial-mesenchymal transition (EMT) signaling and activating an epithelial program within the cells. Aiming at MET, we investigated the potential of keratinocyte growth factor (KGF) and bone morphogenetic protein (BMP)-6 separately for the induction of MET in 3T3L1 mouse adipose cells and to trace the molecular events that probably upregulate during MET induction. KGF successfully induced MET through upregulation of epithelial related genes and transcript expression on 3T3L1 cells. In contrast, BMP-6 plays completely the reverse role through downregulation of all epithelial related genes and transcript expression. In KGF based treatment, seven genes (K8, K18, EpCAM, K5, K14, SMN1 and α-SMA) out of a total of eight genes were significantly (P < 0.05/P < 0.01) upregulated. Immunostaining and immunoblotting also revealed significant (P < 0.05/P < 0.01) expression of several epithelial-specific surface antigens and transcripts. Moreover, Ayoub Shaklar staining (specific to keratin) of KGF treated cells showed formation of keratin (reddish brown color) within cytoplasm of the cells, whereas control and BMP-6 treated cells did not. Conclusively, KGF was observed to have the potential to enhance MET and these clues could be used in future research into cellular reprogramming and regenerative medicine.


Subject(s)
Bone Morphogenetic Protein 6/pharmacology , Cellular Reprogramming/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibroblast Growth Factor 7/pharmacology , 3T3-L1 Cells , Animals , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cell Differentiation/drug effects , Down-Regulation/drug effects , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Fluorescent Antibody Technique , Keratins/genetics , Keratins/metabolism , Mice , Microscopy, Confocal , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Up-Regulation/drug effects
14.
J Cell Sci ; 125(Pt 18): 4306-19, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22641693

ABSTRACT

Bone morphogenetic proteins (BMPs) are a large family of multi-functional secreted signalling molecules. Previously BMP2/4 were shown to inhibit skin pigmentation by downregulating tyrosinase expression and activity in epidermal melanocytes. However, a possible role for other BMP family members and their antagonists in melanogenesis has not yet been explored. In this study we show that BMP4 and BMP6, from two different BMP subclasses, and their antagonists noggin and sclerostin were variably expressed in melanocytes and keratinocytes in human skin. We further examined their involvement in melanogenesis and melanin transfer using fully matched primary cultures of adult human melanocytes and keratinocytes. BMP6 markedly stimulated melanogenesis by upregulating tyrosinase expression and activity, and also stimulated the formation of filopodia and Myosin-X expression in melanocytes, which was associated with increased melanosome transfer from melanocytes to keratinocytes. BMP4, by contrast, inhibited melanin synthesis and transfer to below baseline levels. These findings were confirmed using siRNA knockdown of BMP receptors BMPR1A/1B or of Myosin-X, as well as by incubating cells with the antagonists noggin and sclerostin. While BMP6 was found to use the p38MAPK pathway to regulate melanogenesis in human melanocytes independently of the Smad pathway, p38MAPK, PI3-K and Smad pathways were all involved in BMP6-mediated melanin transfer. This suggests that pigment formation may be regulated independently of pigment transfer. These data reveal a complex involvement of regulation of different members of the BMP family, their antagonists and inhibitory Smads, in melanocytes behaviour.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Protein 6/pharmacology , Keratinocytes/metabolism , Melanocytes/metabolism , Pigmentation/drug effects , Skin/cytology , Adult , Aged , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/metabolism , Bone Morphogenetic Protein Receptors/metabolism , Coculture Techniques , Epidermis/drug effects , Epidermis/metabolism , Epidermis/radiation effects , Female , Gene Knockdown Techniques , Humans , Keratinocytes/drug effects , Keratinocytes/enzymology , Keratinocytes/radiation effects , Melanins/biosynthesis , Melanocytes/drug effects , Melanocytes/radiation effects , Melanocytes/ultrastructure , Middle Aged , Models, Biological , Monophenol Monooxygenase/metabolism , Myosins/metabolism , Pigmentation/radiation effects , Pseudopodia/drug effects , Pseudopodia/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Smad Proteins/metabolism , Ultraviolet Rays , Up-Regulation/drug effects , Up-Regulation/radiation effects , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Int Orthop ; 38(3): 635-47, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24352822

ABSTRACT

PURPOSE: The purpose of this study was to revise the clinical use of commercial BMP2 (Infuse) and BMP7 (Osigraft) based bone devices and explore the mechanism of action and efficacy of low BMP6 doses in a novel whole blood biocompatible device OSTEOGROW. METHODS: Complications from the clinical use of BMP2 and BMP7 have been systemically reviewed in light of their role in bone remodeling. BMP6 function has been assessed in Bmp6-/- mice by µCT and skeletal histology, and has also been examined in mesenchymal stem cells (MSC), hematopoietic stem cells (HSC) and osteoclasts. Safety and efficacy of OSTEOGROW have been assessed in rats and rabbits. RESULTS: Clinical use issues of BMP2 and BMP7 have been ascribed to the limited understanding of their role in bone remodeling at the time of device development for clinical trials. BMP2 and BMP7 in bone devices significantly promote bone resorption leading to osteolysis at the endosteal surfaces, while in parallel stimulating exuberant bone formation in surrounding tissues. Unbound BMP2 and BMP7 in bone devices precipitate on the bovine collagen and cause inflammation and swelling. OSTEOGROW required small amounts of BMP6, applied in a biocompatible blood coagulum carrier, for stimulating differentiation of MSCs and accelerated healing of critical size bone defects in animals, without bone resorption and inflammation. BMP6 decreased the number of osteoclasts derived from HSC, while BMP2 and BMP7 increased their number. CONCLUSIONS: Current issues and challenges with commercial bone devices may be resolved by using novel BMP6 biocompatible device OSTEOGROW, which will be clinically tested in metaphyseal bone fractures, compartments where BMP2 and BMP7 have not been effective.


Subject(s)
Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein 6/therapeutic use , Drug Delivery Systems , Fractures, Bone/drug therapy , Osteogenesis/drug effects , Wound Healing/drug effects , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/pharmacology , Biocompatible Materials/therapeutic use , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/therapeutic use , Bone Morphogenetic Protein 6/administration & dosage , Bone Morphogenetic Protein 7/pharmacology , Bone Morphogenetic Protein 7/therapeutic use , Dose-Response Relationship, Drug , Fractures, Bone/physiopathology , Mice , Mice, Knockout , Models, Animal , Osteogenesis/physiology , Rabbits , Rats , Wound Healing/physiology
16.
Growth Factors ; 31(5): 141-53, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23879371

ABSTRACT

In the present study bone morphogenetic protein (BMP)-6 alone or in synergy with BMP-7 and vascular endothelial growth factor (VEGF) were tested with human adipose stem cells (hASCs) seeded on cell culture plastic or 3D bioactive glass. Osteogenic medium (OM) was used as a positive control for osteogenic differentiation. The same growth factor groups were also tested combined with OM. None of the growth factor treatments could enhance the osteogenic differentiation of hASCs in 3D- or 2D-culture compared to control or OM. In 3D-culture OM promoted significantly total collagen production, whereas in 2D-culture OM induced high total ALP activity and mineralization compared to control and growth factors groups, but also high cell proliferation. In this study, hASCs did not respond to exogenously added growth although various parameters of the study set-up may have affected these findings contradictory to the previous literature.


Subject(s)
Adipocytes/cytology , Adult Stem Cells/cytology , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Osteogenesis/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Adipocytes/drug effects , Adult Stem Cells/drug effects , Biocompatible Materials/pharmacology , Cells, Cultured , Glass , Humans , Plastics/pharmacology , Tissue Scaffolds
17.
Biochem Biophys Res Commun ; 440(4): 786-91, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24134848

ABSTRACT

Chondrogenesis has been widely investigated in vitro using bone marrow-derived mesenchymal stromal cells (BM-MSCs) or primary chondrocytes. However, their use raises some issues partially circumvented by the availability of Adipose tissue-derived MSCs. Herein; we characterized the chondrogenic potential of human Multipotent Adipose-Derived Stem (hMADS) cells, and their potential use as pharmacological tool. hMADS cells are able to synthesize matrix proteins including COMP, Aggrecan and type II Collagen. Furthermore, hMADS cells express BMP receptors in a similar manner to BM-MSC, and BMP6 treatment of differentiated cells prevents expression of the hypertrophic marker type X Collagen. We tested whether IL-1ß and nicotine could impact chondrocyte differentiation. As expected, IL-1ß induced ADAMTS-4 gene expression and modulated negatively chondrogenesis while these effects were reverted in the presence of the IL-1 receptor antagonist. Nicotine, at concentrations similar to those observed in blood of smokers, exhibited a dose dependent increase of Aggrecan expression, suggesting an unexpected protective effect of the drug under these conditions. Therefore, hMADS cells represent a valuable tool for the analysis of in vitro chondrocyte differentiation and to screen for potentially interesting pharmacological drugs.


Subject(s)
Adipose Tissue/cytology , Chondrocytes/cytology , Chondrogenesis/physiology , Multipotent Stem Cells/cytology , ADAM Proteins/genetics , ADAMTS4 Protein , Aggrecans/biosynthesis , Bone Morphogenetic Protein 6/pharmacology , Bone Morphogenetic Protein Receptors/metabolism , Cell Separation , Chondrogenesis/genetics , Collagen Type X/metabolism , Gene Expression/drug effects , Humans , Interleukin-1beta/pharmacology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Nicotine/pharmacology , Procollagen N-Endopeptidase/genetics
18.
Blood ; 117(3): 997-1004, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21076043

ABSTRACT

Hepcidin is a major regulator of iron homeostasis, and its expression in liver is regulated by iron, inflammation, and erythropoietic activity with mechanisms that involve bone morphogenetic proteins (BMPs) binding their receptors and coreceptors. Here we show that exogenous heparin strongly inhibited hepcidin expression in hepatic HepG2 cells at pharmacologic concentrations, with a mechanism that probably involves bone morphogenetic protein 6 sequestering and the blocking of SMAD signaling. Treatment of mice with pharmacologic doses of heparin inhibited liver hepcidin mRNA expression and SMAD phosphorylation, reduced spleen iron concentration, and increased serum iron. Moreover, we observed a strong reduction of serum hepcidin in 5 patients treated with heparin to prevent deep vein thrombosis, which was accompanied by an increase of serum iron and a reduction of C-reactive protein levels. The data show an unrecognized role for heparin in regulating iron homeostasis and indicate novel approaches to the treatment of iron-restricted iron deficiency anemia.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Gene Expression Regulation/drug effects , Heparin/pharmacology , Smad Proteins/metabolism , Aged , Aged, 80 and over , Animals , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Bone Morphogenetic Protein 6/pharmacology , C-Reactive Protein/metabolism , Female , Fondaparinux , Hep G2 Cells , Heparin/analogs & derivatives , Heparin/therapeutic use , Heparin, Low-Molecular-Weight/pharmacology , Hepcidins , Humans , Interleukin-6/pharmacology , Iron/blood , Iron/metabolism , Mice , Phosphorylation/drug effects , Polysaccharides/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Spleen/drug effects , Spleen/metabolism , Venous Thrombosis/blood , Venous Thrombosis/drug therapy
19.
J Nutr ; 143(7): 1061-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23700338

ABSTRACT

Hepcidin, the pivotal regulator of iron metabolism, plays a critical role in multiple diseases including anemia of chronic disease and hemochromatosis. Recent studies have focused on identifying antagonists of hepcidin. We hypothesized that bioactive extracts from Chinese medicinal plants may be efficacious in the inhibition of expression of the hepcidin-encoding gene (HAMP) product, hepcidin. To test this, we measured the level of hepcidin expression in cultured cells treated with 16 different medicinal plant extracts, all of which are used to treat anemia-related disorders in traditional Chinese medicine. Among the extracts tested, that of Caulis Spatholobi (CS; also called Jixueteng, the stem of Spatholobus suberectus Dunn) showed the most potent inhibitory effect on HAMP expression in the Huh7 cell line and was therefore selected for further mechanistic study. In cells treated with 400 µg/mL of extract, phosphorylated mothers against decapentaplegic homolog proteins 1/5/8 levels were 80% less than those of controls (P < 0.001), and the inhibitory effect on interleukin-6-induced HAMP expression (65% inhibition) was weaker than the strong inhibition on bone morphogenetic protein 6-induced HAMP expression (97% inhibition). Seven-week-old C57BL/6 female mice were fed an AIN-76A diet containing 10.8% dried CS and then analyzed on d 0, 5, 10, or 15. On d 5, there was a 60% decrease in hepatic HAMP expression (P < 0.05), an 18% decrease in hepatic iron concentration, and a 100% increase in serum iron concentration (P < 0.05) compared with the d 0 group. In conclusion, we identify the extract of CS as a novel, potent HAMP expression inhibitor, which may be further modified and optimized to become a dietary supplement or a therapeutic option for the amelioration of hepcidin-overexpression-related diseases, including iron deficiency anemia.


Subject(s)
Antimicrobial Cationic Peptides/genetics , Fabaceae/chemistry , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/genetics , Animals , Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Bone Morphogenetic Protein 6/pharmacology , Cell Line, Tumor , Female , Hep G2 Cells , Hepcidins , Humans , Interleukin-6/metabolism , Iron/blood , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA
20.
Cells Tissues Organs ; 198(6): 438-47, 2013.
Article in English | MEDLINE | ID: mdl-24732882

ABSTRACT

The dental follicle (DF) plays an essential role in tooth eruption via regulation of bone resorption and bone formation. Bone morphogenetic protein-6 (BMP6) expression in the DF is coincident with bone growth in the tooth crypt. DF stem cells (DFSCs) have been shown to possess strong osteogenic capability. This study aims to determine the expression of BMP6 in DFSCs and to elucidate the role of BMP6 in the osteogenesis of DFSCs. DFSCs and their non-stem cell counterpart, DF cells (DFCs), were obtained from the DFs of rat pups. We showed that expression of BMP6 was significantly higher in the DFSCs than in the DFCs. DFSCs lost osteogenic capability during in vitro expansion, and DFSCs in late passages had reduced BMP6 expression as compared to early passages of DFSCs when they were subjected to osteogenic induction. Addition of exogenous human recombinant BMP6 (hrBMP6) to the osteogenic medium dramatically enhanced the osteogenesis of the late-passage DFSCs. Knockdown of BMP6 by short interfering RNA in the DFSCs in early passages resulted in a decrease in osteogenesis, which could be restored by addition of hrBMP6. We concluded that DFSCs need to express high levels of BMP6 to maintain their osteogenesis capability. Increased BMP6 expression seen in vivo in the DF may reflect the activation of DFSCs for osteogenic differentiation for bone growth during tooth eruption.


Subject(s)
Bone Morphogenetic Protein 6/biosynthesis , Cell Differentiation/drug effects , Dental Sac/metabolism , Osteogenesis/drug effects , Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/pharmacology , Cell Differentiation/genetics , Cells, Cultured , Dental Sac/cytology , Humans , Osteogenesis/genetics , RNA Interference , RNA, Small Interfering , Rats , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL