Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters











Publication year range
1.
Commun Biol ; 4(1): 1039, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489520

ABSTRACT

The cardiac valvular endothelial cells (VECs) are an ideal cell source that could be used for making the valve organoids. However, few studies have been focused on the derivation of this important cell type. Here we describe a two-step chemically defined xeno-free method for generating VEC-like cells from human pluripotent stem cells (hPSCs). HPSCs were specified to KDR+/ISL1+ multipotent cardiac progenitors (CPCs), followed by differentiation into valve endothelial-like cells (VELs) via an intermediate endocardial cushion cell (ECC) type. Mechanistically, administration of TGFb1 and BMP4 may specify VEC fate by activating the NOTCH/WNT signaling pathways and previously unidentified targets such as ATF3 and KLF family of transcription factors. When seeded onto the surface of the de-cellularized porcine aortic valve (DCV) matrix scaffolds, hPSC-derived VELs exhibit superior proliferative and clonogenic potential than the primary VECs and human aortic endothelial cells (HAEC). Our results show that hPSC-derived valvular cells could be efficiently generated from hPSCs, which might be used as seed cells for construction of valve organoids or next generation tissue engineered heart valves.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Endothelial Cells/cytology , Heart Valves/cytology , Pluripotent Stem Cells/cytology , Transforming Growth Factor beta1/administration & dosage , Animals , Humans , Sus scrofa
2.
Biol Open ; 10(9)2021 09 15.
Article in English | MEDLINE | ID: mdl-34435204

ABSTRACT

Human embryonic stem cells (hESCs) possess an immense potential to generate clinically relevant cell types and unveil mechanisms underlying early human development. However, using hESCs for discovery or translation requires accurately identifying differentiated cell types through comparison with their in vivo counterparts. Here, we set out to determine the identity of much debated BMP-treated hESCs by comparing their transcriptome to recently published single cell transcriptomic data from early human embryos ( Xiang et al., 2020). Our analyses reveal several discrepancies in the published human embryo dataset, including misclassification of putative amnion, intermediate and inner cell mass cells. These misclassifications primarily resulted from similarities in pseudogene expression, highlighting the need to carefully consider gene lists when making comparisons between cell types. In the absence of a relevant human dataset, we utilized the recently published single cell transcriptome of the early post implantation monkey embryo to discern the identity of BMP-treated hESCs. Our results suggest that BMP-treated hESCs are transcriptionally more similar to amnion cells than trophectoderm cells in the monkey embryo. Together with prior studies, this result indicates that hESCs possess a unique ability to form mature trophectoderm subtypes via an amnion-like transcriptional state. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Amnion/cytology , Embryo, Mammalian/cytology , Embryonic Development/physiology , Human Embryonic Stem Cells/cytology , Transcriptome/physiology , Animals , Bone Morphogenetic Protein 4/administration & dosage , Gene Expression Regulation, Developmental , Haplorhini , Humans
3.
Colloids Surf B Biointerfaces ; 194: 111167, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32531716

ABSTRACT

This work aimed to seek a sustained drug release system based on poloxamer-based thermoresponsive gel for sustained release drugs to inhibit corneal neovascularization (CNV) after eye operations. Thus, we designed and prepared a thermoresponsive gel with a phase transition temperature from 22 °C to 25 °C. When the concentrations of poloxamer (P) was 18% (w/w) and ε-Polylysine (EPL) was 0.5 mg/mL (P-18-EPL-05) in the thermoresponsive gel solution, the obtained thermoresponsive gel showed a suitable viscosity and strength in physiological condition. The viscosity, storage modulus G' and loss modulus G" of P-18-EPL-05 were 8 × 102 mPa.s, 1.17 × 104 Pa and 3.77 × 103 Pa, respectively. In-vitro release studies indicated that the drug release ratio of P-18-EPL-05 gel better than that of the poloxamer solution alone. The animal experiments indicated that the thermoresponsive gel loading bone morphogenetic protein 4 (BMP4) was better to inhibit CNV than the common solvent one. Overall, these results demonstrated that P-18-EPL-05 gel would be a promising platform as drug sustained systems for inhibiting CNV after eye injury in ophthalmic applications.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Corneal Neovascularization , Drug Delivery Systems , Animals , Corneal Neovascularization/drug therapy , Delayed-Action Preparations , Excipients , Gels , Poloxamer , Temperature , Viscosity
4.
Braz. arch. biol. technol ; 62: e19180699, 2019. graf
Article in English | LILACS | ID: biblio-1055375

ABSTRACT

Abstract Bone morphogenetic protein-4 (BMP4) is a member of the bone morphogenetic protein family which plays an important role in bone formation, inflammation and cardiac hypertrophy. The aim of this study was to investigate the underlying molecular mechanism that BMP4-induced cardiomyocyte hypertrophy. H9c2 cells were used to measure cell surface area and protein synthesis. Western blot was used to examine hypertrophic marker brain natriuretic peptide (BNP) protein expression and phosphorylation of ERK1/2. The results exhibited that cell surface area, protein synthesis and BNP protein expression were increased with BMP4 treatment. While PD98059 inhibited these effects of BMP4. In addition, BMP4 treatment increased phosphorylation of ERK1/2 in a time- and dose-dependent manner. PD98059 treatment decreased phosphorylation of ERK1/2 that was increased by BMP4. These results suggest that BMP4 induces cardiomyocyte hypertrophy through the activation of ERK1/2 cell signaling pathway.


Subject(s)
Cardiomegaly/chemically induced , Bone Morphogenetic Protein 4/administration & dosage , Blotting, Western/instrumentation , Mitogen-Activated Protein Kinase 3 , Wnt Signaling Pathway
5.
J Cell Physiol ; 233(6): 4907-4918, 2018 06.
Article in English | MEDLINE | ID: mdl-29215714

ABSTRACT

Photobiomodulation (PBM) therapy displays relevant properties for tissue healing and regeneration, which may be of interest for the tissue engineering field. Here, we show that PBM is able to improve cell survival and to interact with recombinant human Bone Morphogenetic Protein 4 (rhBMP4) to direct and accelerate odonto/osteogenic differentiation of dental derived mesenchymal stem cells (MSCs). MSCs were encapsulated in an injectable and thermo-responsive cell carrier (Pluronic® F-127) loaded with rhBMP4 and then photoactivated. PBM improved MSCs self-renewal and survival upon encapsulation in the Pluronic® F-127. In the presence of rhBMP4, cell odonto/osteogenic differentiation was premature and markedly improved in the photoactivated MSCs. An in vivo calvarial critical sized defect model demonstrated significant increase in bone formation after PBM treatment. Finally, a balance in the reactive oxygen species levels may be related to the favorable results of PBM and rhBMP4 association. PBM may act in synergism with rhBMP4 and is a promise candidate to direct and accelerate hard tissue bioengineering.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Drug Carriers , Low-Level Light Therapy/methods , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/radiation effects , Poloxamer/chemistry , Tissue Engineering/methods , Tissue Scaffolds , Adolescent , Adult , Animals , Bone Morphogenetic Protein 4/chemistry , Bone Regeneration , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Self Renewal/drug effects , Cell Self Renewal/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , Humans , Hydrogels , Injections , Lasers, Semiconductor , Low-Level Light Therapy/instrumentation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Mice, Nude , NF-kappa B/metabolism , Osteogenesis/drug effects , Osteogenesis/radiation effects , Parietal Bone/injuries , Parietal Bone/pathology , Parietal Bone/surgery , Reactive Oxygen Species/metabolism , Time Factors , Young Adult
6.
Stem Cell Reports ; 9(6): 1754-1764, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29173898

ABSTRACT

Human pluripotent stem cells (hPSCs) are widely used to study cardiovascular cell differentiation and function. Here, we induced differentiation of hPSCs (both embryonic and induced) to proepicardial/epicardial progenitor cells that cover the heart during development. Addition of retinoic acid (RA) and bone morphogenetic protein 4 (BMP4) promoted expression of the mesodermal marker PDGFRα, upregulated characteristic (pro)epicardial progenitor cell genes, and downregulated transcription of myocardial genes. We confirmed the (pro)epicardial-like properties of these cells using in vitro co-culture assays and in ovo grafting of hPSC-epicardial cells into chick embryos. Our data show that RA + BMP4-treated hPSCs differentiate into (pro)epicardial-like cells displaying functional properties (adhesion and spreading over the myocardium) of their in vivo counterpart. The results extend evidence that hPSCs are an excellent model to study (pro)epicardial differentiation into cardiovascular cells in human development and evaluate their potential for cardiac regeneration.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Heart/growth & development , Induced Pluripotent Stem Cells/cytology , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cardiovascular System/cytology , Cardiovascular System/growth & development , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Myocardium/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pericardium/cytology , Pericardium/growth & development , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stem Cells/cytology , Tretinoin/administration & dosage
7.
Theriogenology ; 97: 113-123, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28583594

ABSTRACT

Bone morphogenetic protein-4 (BMP-4) and FSH play important regulatory roles in follicular growth and steroidogenesis in vivo. The purpose of this study was to investigate the effects of BMP-4 and FSH on in vitro growth (IVG) and steroidogenesis of bovine oocyte-cumulus-granulosa complexes (OCGCs). We cultured OCGCs collected from early antral follicles (0.5-1 mm) in medium without BMP-4 and FSH for 4 days and investigated the appearance of OCGCs and their steroidogenesis. During the first 4 days of IVG, morphologically normal OCGCs produced more estradiol-17ß (E2), but less progesterone (P4). Morphologically normal OCGCs were subjected to an additional culture in medium supplemented with BMP-4 (0, 10, and 50 ng/mL) and FSH (0 and 0.5 ng/mL) until day 12. We examined the viability and steroidogenesis of OCGCs after 8 and 12 days of culture. Oocyte growth, characteristics of granulosa cells, and the maturational competence of oocytes were also investigated. On day 8, the viability of OCGCs cultured without FSH was higher in the 10 ng/mL BMP-4 group than in the 50 ng/mL BMP-4 group (P < 0.05). No significant difference was observed in the viability of groups cultured with FSH, regardless of the addition of BMP-4, and FSH improved the viability of 50 ng/mL BMP-4 group similar to 10 ng/mL BMP-4 group. The total number of granulosa cells was larger in 10 ng/mL BMP-4 group cultured with FSH than in 50 ng/mL BMP-4 group cultured with FSH on day 8 (P < 0.05). E2 production decreased from days 8-12, and P4 production increased throughout IVG culture, regardless of the addition of BMP-4 and FSH (P < 0.05). No significant differences in E2 production were observed between groups from days 4-8, regardless of whether BMP-4 was added without FSH; however, E2 production in the group cultured with 50 ng/mL BMP-4 was suppressed by FSH. BMP-4 suppressed E2 production from days 8-12, regardless of whether FSH was added. The group cultured with 10 ng/mL BMP-4 without FSH showed the lowest P4 production among all groups for all culture periods. OCGCs that produced mature oocytes tended to secrete more E2 and less P4 than OCGCs that produced immature oocytes. In conclusion, until day 8 of the IVG culture, P4 production by OCGCs was suppressed by the addition of 10 ng/mL BMP-4 in the absence of FSH, without inhibiting E2 production. These conditions appear to mimic growing follicles until day 8 and mimic degenerating follicles from days 8-12 of culture.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Estradiol/biosynthesis , Follicle Stimulating Hormone/pharmacology , Granulosa Cells/drug effects , Oocytes/drug effects , Progesterone/biosynthesis , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cattle , Cell Culture Techniques/veterinary , Culture Media , Female , Follicle Stimulating Hormone/administration & dosage , Granulosa Cells/physiology , Oocytes/growth & development
8.
PLoS One ; 12(4): e0175095, 2017.
Article in English | MEDLINE | ID: mdl-28380024

ABSTRACT

Controlled release drug delivery systems are well established as oral and implantable dosage forms. However, the controlled release paradigm can also be used to present complex soluble signals responsible for cellular organization during development. Endochondral ossification (EO), the developmental process of bone formation from a cartilage matrix is controlled by several soluble signals with distinct functions that vary in structure, molecular weight and stability. This makes delivering them from a single vehicle rather challenging. Herein, a gelatin-based delivery system suitable for the delivery of small molecules as well as recombinant human (rh) proteins (rhWNT3A, rhFGF2, rhVEGF, rhBMP4) is reported. The release behavior and biological activity of the released molecules was validated using analytical and biological assays, including cell reporter systems. The simplicity of fabrication of the gelatin device should foster its adaptation by the diverse scientific community interested in interrogating developmental processes, in vivo.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Drug Delivery Systems/methods , Fibroblast Growth Factor 2/administration & dosage , Gelatin/therapeutic use , Osteogenesis/drug effects , Vascular Endothelial Growth Factor A/administration & dosage , Wnt3A Protein/administration & dosage , Bone Morphogenetic Protein 4/pharmacology , Cell Line , Fibroblast Growth Factor 2/pharmacology , Humans , Vascular Endothelial Growth Factor A/pharmacology , Wnt3A Protein/pharmacology
9.
Differentiation ; 93: 1-14, 2017.
Article in English | MEDLINE | ID: mdl-27639333

ABSTRACT

While human induced pluripotent stem (hiPS) cells have potential use in regenerative medicine, there are no reports on odontoblastic differentiation of hiPS cells. In the current study, to examine integrin profiles and explore the early signaling cascade of odontoblastic differentiation in hiPS cells, we investigated the regulation of autophagy-related gene (Atg) and wingless/int1 (Wnt) signaling in gelatin scaffold (GS) combined with bone morphogenetic protein (BMP)-4 (GS/BMP-4)-mediated odontoblastic differentiation. Following GS/BMP-4 treatment, there was a dramatic loss of α3 and α6 integrins, and reciprocal strong induction of α1 integrin expression in the differentiated cells. GS/BMP-4 increased the mRNA and protein levels of Atg10, Lrp5/Fzd9 (an Atg10 receptor), and Wnt5 together with the amount of autophagosomes and autophagic fluxes. Treatment with siRNAs against Atg10 and Wnt5a individually suppressed the GS/BMP-4-induced increase in odontoblastic differentiation. The odontoblastic phenotype was inhibited by chloroquine, but increased after treatment with rapamycin (an autophagy enhancer). Taken together with our previous findings, we have replicated our results from the rodent system in a novel human system. We have revealed a unique sequential cascade involving Atg10, Wnt5a, α1 integrin, and matrix metalloproteinase-3 in GS/BMP-4-induced differentiation of hiPS cells into odontoblast-like cells at a relatively early stage.


Subject(s)
Autophagy-Related Proteins/genetics , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/drug effects , Integrin alpha Chains/genetics , Matrix Metalloproteinase 3/genetics , Vesicular Transport Proteins/genetics , Wnt-5a Protein/genetics , Bone Morphogenetic Protein 4/administration & dosage , Cell Differentiation/drug effects , Gelatin/administration & dosage , Gelatin/chemistry , Humans , Induced Pluripotent Stem Cells/cytology , Odontoblasts/drug effects , RNA, Small Interfering/administration & dosage , Regenerative Medicine , Signal Transduction/drug effects , Sirolimus/administration & dosage , Tissue Scaffolds
10.
Macromol Biosci ; 17(3)2017 03.
Article in English | MEDLINE | ID: mdl-27748553

ABSTRACT

Producing meiosis-competent germ cells (GCs) from embryonic stem cells (ESCs) is essential for developing advanced therapies for infertility. Here, a novel approach is presented for generation of GCs from ESCs. In this regard, microparticles (MPs) have been developed from alginate sulfate loaded with bone morphogenetic protein 4 (BMP4). The results here show that BMP4 release from alginate sulfate MPs is significantly retarded by the sulfated groups compared to neat alginate. Then, BMP4-laden MPs are incorporated within the aggregates during differentiation of GCs from ESCs. It is observed that BMP4-laden MPs increase GC differentiation from ESCs at least twofold compared to the conventional soluble delivery method. Interestingly, following meiosis induction, Dazl, an intrinsic factor that enables GCs to enter meiosis, and two essential meiosis genes (Stra8 and Smc1b) are upregulated significantly in MP-induced aggregates compared to aggregates, which are formed by the conventional method. Together, these data show that controlled delivery of BMP4 during ESC differentiation into GC establish meiosis-competent GCs which can serve as an attractive GC source for reproductive medicine.


Subject(s)
Bone Morphogenetic Protein 4/chemistry , Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Mesenchymal Stem Cells/drug effects , Alginates/administration & dosage , Alginates/chemistry , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cell-Derived Microparticles/chemistry , Embryonic Stem Cells/cytology , Germ Cells/drug effects , Germ Cells/growth & development , Glucuronic Acid/administration & dosage , Glucuronic Acid/chemistry , Hexuronic Acids/administration & dosage , Hexuronic Acids/chemistry , Meiosis/genetics , Mesenchymal Stem Cells/cytology , Mice
11.
Inflammation ; 40(2): 442-453, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27995357

ABSTRACT

The endothelium serves as a selective barrier and controls the exchange of nutrients, hormones, and leukocytes between blood and tissues. Molecular mechanisms contributing to the pathogenesis of endothelial barrier dysfunction remain incompletely understood. Accumulating evidence implicates bone morphogenetic protein (BMP)-modulator BMPER as a key regulator in endothelial biology. Herein, we analyze the impact of BMPER in the control of endothelial barrier function. To assess the role of BMPER in vascular barrier function in mice, we measured the leakage of Evans blue dye from blood into interstitial lung tissue. BMPER+/- mice exhibited a significantly higher degree of vascular leak compared with wild-type siblings. In accordance with our in vivo observation, siRNA-based BMPER knockdown in human umbilical endothelial cells increased endothelial permeability measured by FITC-dextran passage in transwell assays. Mechanistically, BMPER knockdown reduced the expression of VE-cadherin, a pivotal component of endothelial adherens junctions. Conversely, recombinant human BMPER protein upregulated VE-cadherin protein levels and improved endothelial barrier function in transwell assays. The effects of BMPER knockdown on VE-cadherin expression and endothelial permeability were induced by enhanced BMP activity. Supporting this notion, activation of BMP4-Smad-Id1 signaling reduced VE-cadherin levels and impaired endothelial barrier function in vitro. In vivo, Evans blue dye accumulation was higher in the lungs of BMP4-treated C57BL/6 mice compared to controls indicating that BMP4 increased vascular permeability. High levels of BMPER antagonized BMP4-Smad5-Id1 signaling and prevented BMP4-induced downregulation of VE-cadherin and endothelial leakage, suggesting that BMPER exerts anti-BMP effects and restores endothelial barrier function. Taken together, this data demonstrates that BMPER-modulated BMP pathway activity regulates VE-cadherin expression and vascular barrier function.


Subject(s)
Carrier Proteins/physiology , Endothelium, Vascular/physiology , Animals , Antigens, CD/metabolism , Bone Morphogenetic Protein 4/administration & dosage , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Proteins/metabolism , Cadherins/metabolism , Capillary Permeability/drug effects , Carrier Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , RNA, Small Interfering/pharmacology , Signal Transduction
12.
In Vitro Cell Dev Biol Anim ; 52(8): 885-93, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27496193

ABSTRACT

Human embryonic stem cells (hESCs) can differentiate into all somatic lineages including stratified squamous epithelia. Thus, efficient methods are required to direct hESC differentiation to obtain a pure subpopulation for tissue engineering. The study aimed to assess the effects of retinoic acid (RA), bone morphogenetic protein-4 (BMP4), and ascorbic acid (AA) on the differentiation of hESCs into keratinocyte progenitors in vitro. The first media contained AA and BMP4; the second contained RA, AA, and BMP4; the third was commercial-defined keratinocyte serum-free medium, which was used to differentiate H9 hESCs (direct approach) or embryoid bodies (EBs) (indirect approach) into keratinocyte progenitors. Real-time RT-PCR, immunofluorescence, and flow-cytometry were used to characterize the differentiated cells. Cells induced by AA + BMP4 + RA showed the typical epithelial morphology, while cells induced by AA + BMP4 showed multiple appearances. CK14 and p63 messenger RNA (mRNA) expressions in the AA + BMP4 + RA-treated cells were higher than those of the AA + BMP4-treated cells (CK14: 22.4-fold; p63: 84.7-fold). Epithelial marker CK18 mRNA expressions at 14 d of differentiation and keratinocyte marker CK14 and transcription factor p63 mRNA expressions at 35 d of differentiation were higher in cells differentiated from hESCs compared with those differentiated from EBs (CK18 10.51 ± 3.26 vs. 6.67 ± 1.28; CK14 9.27 ± 3.61 vs. 5.32 ± 1.86; p63 0.73 ± 0.06 vs. 0.44 ± 0.12, all P < 0.05) After hESC induction by AA+BMP4+RA, CK14 mRNA expression was upregulated after day 21, peaking by 35 d of differentiation. Combined RA, BMP4, and AA could effectively induce differentiation of hESCs into keratinocyte progenitors in vitro. These keratinocytes could be used for oral mucosa and skin tissue engineering.


Subject(s)
Cell Differentiation/drug effects , Human Embryonic Stem Cells/cytology , Keratinocytes/cytology , Tissue Engineering , Ascorbic Acid/administration & dosage , Bone Morphogenetic Protein 4/administration & dosage , Gene Expression Regulation, Developmental/drug effects , Human Embryonic Stem Cells/transplantation , Humans , Keratinocytes/drug effects , Mouth Mucosa/growth & development , RNA, Messenger/biosynthesis , Skin/cytology , Skin/growth & development , Tretinoin/administration & dosage
13.
Cancer Lett ; 375(2): 238-244, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26970275

ABSTRACT

Bone morphogenetic protein 4 (BMP4) is a key regulator of cell proliferation and differentiation. In breast cancer cells, BMP4 has been shown to reduce proliferation in vitro and interestingly, in some cases, also to induce migration and invasion. Here we investigated whether BMP4 influences breast cancer metastasis formation by using a xenograft mouse model. MDA-MB-231 breast cancer cells were injected intracardially into mice and metastasis formation was monitored using bioluminescence imaging. Mice treated with BMP4 developed metastases slightly earlier as compared to control animals but the overall number of metastases was similar in both groups (13 in the BMP4 group vs. 12 in controls). In BMP4-treated mice, bone metastases were more common (10 vs. 7) but adrenal gland metastases were less frequent (1 vs. 5) than in controls. Immunostaining revealed no differences in signaling activation, proliferation rate, blood vessel formation, EMT markers or the number of cancer-associated fibroblasts between the treatment groups. In conclusion, BMP4 caused a trend towards accelerated metastasis formation, especially in bone. More work is needed to uncover the long-term effects of BMP4 and the clinical relevance of these findings.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Bone Neoplasms/genetics , Breast Neoplasms/genetics , Recombinant Proteins/administration & dosage , Animals , Bone Morphogenetic Protein 4/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Metastasis , Recombinant Proteins/genetics , Xenograft Model Antitumor Assays
14.
Biomed Mater ; 11(2): 025011, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27007569

ABSTRACT

Ischemic stroke is often associated with loss of cortical neurons leading to various neurological deficits. A cell replacement based on stem cell transplantation to repair the damaged brain requires the generation of specific neuronal subtypes. Recently, induced pluripotent stem cells have been used to generate various subtypes of neurons in vitro for transplantation in stroke-damaged brains. However, whether these cells can be primed as neuronal precursors to become cortical projection neurons by means of biomaterials releasing differentiation factors is not known. Here, we report that microspheres of biodegradable poly(ester-amide) composed of adipic acid, L-phenyl-alanine and 1,4-butanediol, loaded with differentiation factors, can be used to fate human induced pluripotent stem cell-derived long-term expandable neuroepithelial-like stem cells to cortical projection neurons. The three factors, Wnt3A, BMP4 and cyclopamine, were released from loaded microspheres over at least one month following biphasic dynamic time course, promoting cortical differentiation of the cells in vitro. Microspheres did not evoke significant inflammatory response after transplantation into intact rodent brain. Our study shows the potential of biodegradable polymer microspheres to promote neuronal differentiation by continuous release of factors, thereby creating the appropriate microenvironment. This new strategy may improve the efficacy of stem cell-based therapeutic approaches.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Neurons/cytology , Absorbable Implants , Animals , Biocompatible Materials/chemistry , Bone Morphogenetic Protein 4/administration & dosage , Cell Differentiation/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Drug Delivery Systems , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/transplantation , Materials Testing , Microspheres , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/transplantation , Neurogenesis/drug effects , Neurons/drug effects , Polyesters/chemistry , Rats , Rats, Sprague-Dawley , Stroke/therapy , Veratrum Alkaloids/administration & dosage , Wnt3A Protein/administration & dosage
15.
Oncotarget ; 7(14): 17920-31, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26908439

ABSTRACT

Malignant gliomas patients have a poor survival rate, partially due to the inability in delivering therapeutic agents to the tumors, especially to the metastasis of human glioma stem cells (hGSCs). To explore whether the human neural stem cells (hNSCs) with an over-expression of BMP4 (hNSCs-BMP4) can trace and inhibit hGSCs, in this study, we examined the migration of hNSCs to hGSCs using transwell assay in vitro and performed the fluorescent tracer experiment in vivo. We examined the proliferation, differentiation, apoptosis and migration of hGSCs after co-culturing with hNSCs-BMP4 in vitro and tested the tropism and antitumor effects of hNSCs-BMP4 in the established brain xenograft models of hGSCs. We found that hNSCs-BMP4 could secrete BMP4 and trace hGSCs both in vitro and in vivo. When compared to the normal human astrocytes (NHAs) and hNSCs, hNSCs-BMP4 could significantly inhibit the invasive growth of hGSCs, promote their differentiation and apoptosis by activating Smad1/5/8 signaling, and prolong the survival time of the tumor-bearing nude mice. Collectively, this study suggested that hNSCs-BMP4 may help in developing therapeutic approaches for the treatment of human malignant gliomas.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glioblastoma/pathology , Glioblastoma/therapy , Neoplastic Stem Cells/pathology , Neural Stem Cells/pathology , Animals , Bone Morphogenetic Protein 4/metabolism , Brain Neoplasms/metabolism , Female , Glioblastoma/metabolism , Heterografts , Humans , Mice , Mice, Nude , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation
16.
Development ; 142(19): 3383-93, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26443636

ABSTRACT

The development of organs with an epithelial parenchyma relies on reciprocal mesenchymal-epithelial communication. Mouse corneal epithelium stratification is the consequence of a coordinated developmental process based on mesenchymal-epithelial interactions. The molecular mechanism underlying these interactions remains unclear. The Wnt/ß-catenin signaling pathway is involved in fundamental aspects of development through the regulation of various growth factors. Here, we show that conditional ablation of either ß-catenin (Ctnnb1(cKO)) or co-receptors Lrp5/6 (Lrp5/6(cKO)) in corneal stromal cells results in precocious stratification of the corneal epithelium. By contrast, ectopic expression of a murine Ctnnb1 gain-of-function mutant (Ctnnb1(cGOF)) retards corneal epithelium stratification. We also discovered that Bmp4 is upregulated in the absence of ß-catenin in keratocytes, which further triggers ERK1/2 (Mapk3/1) and Smad1/5 phosphorylation and enhances transcription factor p63 (Trp63) expression in mouse corneal basal epithelial cells and in a human corneal epithelial cell line (HTCE). Interestingly, mouse neonates given a subconjunctival BMP4 injection displayed a phenotype resembling that of Ctnnb1(cKO). Conditional ablation of Bmp4 eradicates the phenotype produced in Ctnnb1(cKO) mice. Furthermore, ChIP and promoter-luciferase assays show that ß-catenin binds to and suppresses Bmp4 promoter activity. These data support the concept that cross-talk between the Wnt/ß-catenin/Bmp4 axis (in the stromal mesenchyme) and Bmp4/p63 signaling (in the epithelium) plays a pivotal role in epithelial stratification during corneal morphogenesis.


Subject(s)
Bone Morphogenetic Protein 4/antagonists & inhibitors , Epithelium, Corneal/embryology , Gene Expression Regulation, Developmental/physiology , Morphogenesis/physiology , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Blotting, Western , Bone Morphogenetic Protein 4/administration & dosage , Chromatin Immunoprecipitation , Doxycycline , Fluorescence , Galactosides , Histological Techniques , Immunohistochemistry , Indoles , Low Density Lipoprotein Receptor-Related Protein-5/deficiency , Low Density Lipoprotein Receptor-Related Protein-6/deficiency , Luciferases , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Phosphoproteins/metabolism , Real-Time Polymerase Chain Reaction , Trans-Activators/metabolism
17.
Int J Cancer ; 137(4): 797-809, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25648303

ABSTRACT

The potential role of osteoblasts in bone and bone marrow (BM) metastases in neuroblastoma (NBL) remains unclear. In this study, we examined the effect of NBL cells on the osteoblastic differentiation of BM-derived mesenchymal stromal cells (BMMSC). We show that the presence of NBL cells enhanced the osteoblastic differentiation of BMMSC driven by bone morphogenetic protein (BMP)-4, in the absence of any effect on NBL cell proliferation. Expression profiles of BMMSC driven toward osteoblastic differentiation revealed an increase in vascular endothelial growth factor A (Vegfa) expression in the presence of NBL cells. We demonstrated that NBL cells increased BMMSC-derived VEGFA mRNA and protein and that this was enhanced by BMP-4. However, in similar conditions, neither the addition of an mVEGFA blocking antibody nor exogenous recombinant (r) mVEGFA affected osteoblastic differentiation. In contrast, siRNA- mediated knock-down of VEGFA in BMMSC prevented osteoblastic differentiation in BMP-4-treated cocultures, an effect that was not reversed in the presence of rmVEGFA. An analysis of murine bones injected with hNBL cells revealed an increase of mVEGFA producing cells near tumor cells concomitantly with an increase in Vegfa and Runx2 mRNA. This coincided with an increase in osteoclasts, in Rankl/Opg mRNA ratio and with the formation of osteolytic lesions. Thus NBL cells promote osteoblastogenesis in the BM by increasing VEGFA expression in BMMSC. Our study provides a new insight into the role of VEGFA in NBL metastases by pointing to the role of stroma-derived intracrine VEGFA in osteoblastogenesis.


Subject(s)
Cell Differentiation/genetics , Lymphocyte Activation/genetics , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cell Line , Cell Proliferation/genetics , Core Binding Factor Alpha 1 Subunit/biosynthesis , Gene Expression Regulation, Developmental , Humans , Mice , Neuroblastoma/metabolism , Osteoblasts/metabolism , Osteoprotegerin/biosynthesis , RNA, Messenger/biosynthesis , Vascular Endothelial Growth Factor A/genetics
18.
Stem Cell Rev Rep ; 11(3): 408-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25277766

ABSTRACT

Bone marrow-derived mesenchymal stem cells (MSC) can be differentiated into myocytes, as well as adipocytes, chondrocytes, and osteocytes in culture. Calcium channels mediate excitation-contraction coupling and are essential for the function of muscle. However, little is known about the expression of calcium channel subunits and calcium handling in stem cells. We examined whether the expression of calcium channel subunits in MSC is similar to that of skeletal muscle satellite cells and if their levels of expression are modified after treatment with bone morphogenetic protein-4 (BMP4). We found that during myogenic differentiation, MSC first express the α2δ1 subunit and the cardiac channel subunit Cav1.2. In contrast to the α2δ1 subunit levels, the Cav1.2 subunit decreases rapidly with time. The skeletal channel subunit Cav1.1 is detected at day 3 but its expression increases considerably, resembling more closely the expression of the subunits in satellite cells. Treatment of MSC with BMP4 caused a significant increase in expression of Cav1.2, a delay in expression of Cav1.1, and a reduction in the duration of calcium transients when extracellular calcium was removed. Calcium currents and transients followed a pattern related to the expression of the cardiac (Cav1.2) or skeletal (Cav1.1) α1subunits. These results indicate that differentiation of untreated MSC resembles differentiation of skeletal muscle and that BMP4 reduces skeletal muscle calcium channel expression and promotes the expression of cardiac calcium channels during myogenic differentiation.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Calcium Channels, L-Type/biosynthesis , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 4/administration & dosage , Calcium Channels, L-Type/genetics , Calcium Signaling , Chondrocytes/cytology , Chondrocytes/metabolism , Gene Expression Regulation, Developmental/drug effects , Mesenchymal Stem Cells/cytology , Mice , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Osteocytes/cytology , Osteocytes/metabolism
19.
J Chem Neuroanat ; 57-58: 54-61, 2014 May.
Article in English | MEDLINE | ID: mdl-24681169

ABSTRACT

Fractones are extracellular matrix structures that display a fractal ultrastructure and that are visualized as puncta after immunolabeling for laminin or heparan sulfate proteoglycans. In the adult brain, fractones are found throughout the subventricular zone (SVZ). The role of fractones is just emerging. We have recently shown that fractones sequester fibroblast growth factor-2 and bone morphogenetic protein-7 from the brain ventricles to regulate cell proliferation in the SVZ of the lateral ventricle, the primary neural stem cell niche and neurogenic zone in adulthood. Here, we have examined in vivo the effect of bone morphogenetic protein-4 (BMP-4) on cell proliferation in the SVZ and we have determined whether BMP-4 interacts with fractones to promote this effect. To examine BMP-4 effect on cell proliferation, BMP-4 was intracerebroventricularly injected, and bromodeoxyuridine immunolabeling was performed on frozen sections of the adult mouse brain. To identify the location of BMP-4 binding, biotinylated-BMP-4 was injected, and its binding localized post-mortem with streptavidin, Texas red conjugate. Injection of heparitinase-1 was used to desulfate fractones and determine whether the binding and the effect of BMP-4 on cell proliferation are heparan sulfate-dependent. BMP-4 inhibited cell proliferation in the SVZ neurogenic zone. Biotinylated-BMP-4 bound to fractones and some adjacent blood vessels. Co-injection of heparitinase-1 and biotinylated-BMP-4 resulted in the absence of signal for biotinylated-BMP-4, indicating that the binding was heparan sulfate dependent. Moreover, preventing the binding of BMP-4 to fractones by heparitinase-1 reinforced the inhibitory effect of BMP-4 on cell proliferation in the SVZ. These results show that BMP-4 inhibits cell proliferation in the SVZ neurogenic zone and that the binding of BMP-4 to fractone-associated heparan sulfates moderates this inhibitory effect. Together with our previous results, these data support the view that fractones capture growth factors and modulate their activity in the neural tissues lining the ventricles.


Subject(s)
Bone Morphogenetic Protein 4/pharmacology , Extracellular Matrix/drug effects , Heparitin Sulfate/pharmacology , Lateral Ventricles/metabolism , Neurogenesis/drug effects , Animals , Blood Vessels/metabolism , Bone Morphogenetic Protein 4/administration & dosage , Cell Count , Cell Proliferation/drug effects , Choroid Plexus/drug effects , Choroid Plexus/metabolism , Female , Injections, Intraventricular , Lateral Ventricles/drug effects , Male , Mice , Mice, Inbred BALB C
20.
Biomed Res Int ; 2013: 140639, 2013.
Article in English | MEDLINE | ID: mdl-24371815

ABSTRACT

The generation and application of porcine induced pluripotent stem cells (iPSCs) may enable the testing for safety and efficacy of therapy in the field of human regenerative medicine. Here, the generation of iPSCs from the Massachusetts General Hospital miniature pig (MGH minipig) established for organ transplantation studies is reported. Fibroblasts were isolated from the skin of the ear of a 10-day-old MGH minipig and transduced with a cocktail of six human factors: POU5F1, NANOG, SOX2, C-MYC, KLF4, and LIN28. Two distinct types of iPSCs were generated that were positive for alkaline phosphatase activity, as well as the classical pluripotency markers: Oct4, Nanog, Sox2, and the surface marker Ssea-1. Only one of two porcine iPSC lines differentiated into three germ layers both in vitro and in vivo. Western blot analysis showed that the porcine iPSCs were dependent on LIF or BMP-4 to sustain self-renewal and pluripotency. In conclusion, the results showed that human pluripotent factors could reprogram porcine ear fibroblasts into the pluripotent state. These cells may provide a useful source of cells that could be used for the treatment of degenerative and genetic diseases and agricultural research and application.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Leukemia Inhibitory Factor/administration & dosage , Animals , Bone Morphogenetic Protein 4/metabolism , Hospitals, General , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Leukemia Inhibitory Factor/metabolism , Massachusetts , Regenerative Medicine , Swine , Swine, Miniature
SELECTION OF CITATIONS
SEARCH DETAIL