Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.203
Filter
1.
Nat Commun ; 15(1): 6365, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39075094

ABSTRACT

Cell fate decisions remain poorly understood at the molecular level. Embryogenesis provides a unique opportunity to analyze molecular details associated with cell fate decisions. Works based on model organisms have provided a conceptual framework of genes that specify cell fate control, for example, transcription factors (TFs) controlling processes from pluripotency to immunity1. How TFs specify cell fate remains poorly understood. Here we report that SALL4 relies on NuRD (nucleosome-remodeling and deacetylase complex) to interpret BMP4 signal and decide cell fate in a well-controlled in vitro system. While NuRD complex cooperates with SALL4 to convert mouse embryonic fibroblasts or MEFs to pluripotency, BMP4 diverts the same process to an alternative fate, PrE (primitive endoderm). Mechanistically, BMP4 signals the dissociation of SALL4 from NuRD physically to establish a gene regulatory network for PrE. Our results provide a conceptual framework to explore the rich landscapes of cell fate choices intrinsic to development in higher organisms involving morphogen-TF-chromatin modifier pathways.


Subject(s)
Bone Morphogenetic Protein 4 , Cell Differentiation , Mi-2 Nucleosome Remodeling and Deacetylase Complex , Transcription Factors , Animals , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Bone Morphogenetic Protein 4/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Chromatin/metabolism , Gene Regulatory Networks , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Endoderm/metabolism , Endoderm/cytology , Signal Transduction , Cell Lineage , DNA-Binding Proteins
2.
Int Immunopharmacol ; 139: 112530, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39053231

ABSTRACT

Fibrosis is a pathological change mainly characterized by an increase of fibrous connective tissue and decrease of parenchymal cells. Its continuous progress may lead to the destruction of organ structure and function decline. An excess of alternatively activated M2 macrophages have been considered crucial candidates in the progression of fibrosis. Bone morphogenetic proteins (BMPs), a group of multifunctional growth factors, are essential for organ development and pathophysiological process, however, the roles that BMPs play in innate immune homeostasis in the development of fibrosis and the downstream signals have not been fully explored. In the current study, we firstly found that the expression of BMP4 was significantly down-regulated in human and mouse fibrosis samples. Then we investigated the effects of BMP4 on macrophage polarization in IL-4 environment and related molecular mechanisms, and found that BMP4 caused a decrease in polarized response towards M2, reflected in the expression of the markers Fizz1, Ym1 and Arg1, together with an inhibition in Stat6 phosphorylation. This relied on the Smad1/5/8 signaling, which had a crosstalk with Stat6. Moreover, the in vivo study showed that BMP4 treatment can reduce collagen deposition and delay the development of experimental pulmonary fibrosis in mice by inhibiting M2 macrophages through adoptive transfer experiment. These findings revealed a novel role of BMP4 in regulating macrophages, offering potential strategies for treating pulmonary fibrosis.


Subject(s)
Bone Morphogenetic Protein 4 , Macrophages , Mice, Inbred C57BL , Pulmonary Fibrosis , Signal Transduction , Animals , Bone Morphogenetic Protein 4/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Humans , Macrophages/immunology , Macrophages/metabolism , Mice , Male , STAT6 Transcription Factor/metabolism , Interleukin-4/metabolism , Macrophage Activation , Lung/pathology , Lung/immunology , Disease Models, Animal
3.
Genes (Basel) ; 15(7)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39062620

ABSTRACT

As an RNA binding protein (RBP), DDX5 is widely involved in the regulation of various biological activities. While recent studies have confirmed that DDX5 can act as a transcriptional cofactor that is involved in the formation of gametes, few studies have investigated whether DDX5 can be used as a transcription factor to regulate the formation of primordial germ cells (PGCs). In this study, we found that DDX5 was significantly up-regulated during chicken PGC formation. Under different PGC induction models, the overexpression of DDX5 not only up-regulates PGC markers but also significantly improves the formation efficiency of primordial germ cell-like cells (PGCLC). Conversely, the inhibition of DDX5 expression can significantly inhibit both the expression of PGC markers and PGCLC formation efficiency. The effect of DDX5 on PGC formation in vivo was consistent with that seen in vitro. Interestingly, DDX5 not only participates in the formation of PGCs but also positively regulates their migration and proliferation. In the process of studying the mechanism by which DDX5 regulates PGC formation, we found that DDX5 acts as a transcription factor to bind to the promoter region of BMP4-a key gene for PGC formation-and activates the expression of BMP4. In summary, we confirm that DDX5 can act as a positive transcription factor to regulate the formation of PGCs in chickens. The obtained results not only enhance our understanding of the way in which DDX5 regulates the development of germ cells but also provide a new target for systematically optimizing the culture and induction system of PGCs in chickens in vitro.


Subject(s)
Bone Morphogenetic Protein 4 , Chickens , DEAD-box RNA Helicases , Germ Cells , Animals , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Germ Cells/metabolism , Chickens/genetics , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Chick Embryo , Gene Expression Regulation, Developmental , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Proliferation , Cell Movement/genetics , Promoter Regions, Genetic
4.
ACS Appl Mater Interfaces ; 16(31): 40455-40468, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39072446

ABSTRACT

Bone morphogenetic proteins (BMPs) are important targets to incorporate in biomaterial scaffolds to orchestrate tissue repair. Glycosaminoglycans (GAGs) such as heparin allow the capture of BMPs and their retention at the surface of biomaterials at safe concentrations. Although heparin has strong affinities for BMP2 and BMP4, two important types of growth factors regulating bone and tissue repair, it remains difficult to embed stably at the surface of a broad range of biomaterials and degrades rapidly in vitro and in vivo. In this report, biomimetic poly(sulfopropyl methacrylate) (PSPMA) brushes are proposed as sulfated GAG mimetic interfaces for the stable capture of BMPs. The growth of PSPMA brushes via a surface-initiated activator regenerated by electron transfer polymerization is investigated via ellipsometry, prior to characterization of swelling and surface chemistry via X-ray photoelectron spectroscopy and Fourier transform infrared. The capacity of PSPMA brushes to bind BMP2 and BMP4 is then characterized via surface plasmon resonance. BMP2 is found to anchor particularly stably and at high density at the surface of PSPMA brushes, and a strong impact of the brush architecture on binding capacity is observed. These results are further confirmed using a quartz crystal microbalance with dissipation monitoring, providing some insights into the mode of adsorption of BMPs at the surface of PSPMA brushes. Primary adsorption of BMP2, with relatively little infiltration, is observed on thick dense brushes, implying that this growth factor should be accessible for further binding of corresponding cell membrane receptors. Finally, to demonstrate the impact of PSPMA brushes for BMP2 capture, dermal fibroblasts were then cultured at the surface of functionalized PSPMA brushes. The presence of BMP2 and the architecture of the brush are found to have a significant impact on matrix deposition at the corresponding interfaces. Therefore, PSPMA brushes emerge as attractive coatings for scaffold engineering and stable capture of BMP2 for regenerative medicine applications.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/chemistry , Bone Morphogenetic Protein 4/metabolism , Humans , Sulfonic Acids/chemistry , Methacrylates/chemistry , Surface Properties , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism
5.
Cell Biol Int ; 48(9): 1326-1342, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38890788

ABSTRACT

Chronic stress is a universal condition commonly associated with many psychiatric diseases. An extensive body of evidence discussed hippocampal affection upon chronic stress exposure, however, the underlying molecular pathways still need to be identified. We investigated the impact of chronic stress on miR200/BMP/Olig-2 signaling and hippocampal myelination. We also compared the effects of chronic administration of amitriptyline and cholecalciferol on chronically stressed hippocampi. Both amitriptyline and cholecalciferol significantly decreased serum cortisol levels, reduced immobility time in the forced swim test, increased the number of crossed squares in open field test, decreased the hippocampal expression of bone morphogenetic protein 4 (BMP4) and its messenger RNA (mRNA) levels, reduced miR200 expression as compared to untreated chronically stressed rats. Also, both drugs amended the hippocampal neuronal damage, enhanced the surviving cell count, and increased the pyramidal layer thickness of Cornu Ammonis subregion 1 (CA1) and granule cell layer of the dentate gyrus. Cholecalciferol was more effective in increasing the area percentage of myelin basic protein (MBP) and Olig-2 positive cells count in hippocampi of chronic stress-exposed rats than amitriptyline, thus enhancing myelination. We also found a negative correlation between the expression of BMP4, its mRNA, miR200, and the immunoexpression of MBP and Olig-2 proteins. This work underscores the amelioration of the stress-induced behavioral changes, inhibition of miR200/BMP4 signaling, and enhancement of hippocampal myelination following chronic administration of either amitriptyline or cholecalciferol, though cholecalciferol seemed more effective in brain remyelination.


Subject(s)
Amitriptyline , Bone Morphogenetic Protein 4 , Cholecalciferol , Hippocampus , MicroRNAs , Signal Transduction , Animals , Male , Rats , Amitriptyline/pharmacology , Bone Morphogenetic Protein 4/metabolism , Cholecalciferol/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Myelin Sheath/metabolism , Myelin Sheath/drug effects , Rats, Wistar , Signal Transduction/drug effects , Stress, Psychological/metabolism
6.
J Control Release ; 371: 386-405, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38844177

ABSTRACT

Recently, the formation of three-dimensional (3D) cell aggregates known as embryoid bodies (EBs) grown in media supplemented with HSC-specific morphogens has been utilized for the directed differentiation of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), into clinically relevant hematopoietic stem cells (HSCs). However, delivering growth factors and nutrients have become ineffective in inducing synchronous differentiation of cells due to their 3D conformation. Moreover, irregularly sized EBs often lead to the formation of necrotic cores in larger EBs, impairing differentiation. Here, we developed two gelatin microparticles (GelMPs) with different release patterns and two HSC-related growth factors conjugated to them. Slow and fast releasing GelMPs were conjugated with bone morphogenic factor-4 (BMP-4) and stem cell factor (SCF), respectively. The sequential presentation of BMP-4 and SCF in GelMPs resulted in efficient and effective hematopoietic differentiation, shown by the enhanced gene and protein expression of several mesoderm and HSC-related markers, and the increased concentration of released HSC-related cytokines. In the present study, we were able to generate CD34+, CD133+, and FLT3+ cells with similar cellular and molecular morphology as the naïve HSCs that can produce colony units of different blood cells, in vitro.


Subject(s)
Bone Morphogenetic Protein 4 , Cell Differentiation , Gelatin , Hematopoietic Stem Cells , Induced Pluripotent Stem Cells , Spheroids, Cellular , Stem Cell Factor , Bone Morphogenetic Protein 4/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Stem Cell Factor/metabolism , Gelatin/chemistry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Animals , Humans , Mice
7.
Differentiation ; 138: 100792, 2024.
Article in English | MEDLINE | ID: mdl-38935992

ABSTRACT

The role extracellular matrix (ECM) in multiple events of morphogenesis has been well described, little is known about its specific role in early eye development. One of the first morphogenic events in lens development is placodal thickening, which converts the presumptive lens ectoderm from cuboidal to pseudostratified epithelium. This process occurs in the anterior pre-placodal ectoderm when the optic vesicle approaches the cephalic ectoderm and is regulated by transcription factor Pax6 and secreted BMP4. Since cells and ECM have a dynamic relationship of interdependence and modulation, we hypothesized that the ECM evolves with cell shape changes during lens placode formation. This study investigates changes in optic ECM including both protein distribution deposition, extracellular gelatinase activity and gene expression patterns during early optic development using chicken and mouse models. In particular, the expression of Timp2, a metalloprotease inhibitor, corresponds with a decrease in gelatinase activity within the optic ECM. Furthermore, we demonstrate that optic ECM remodeling depends on BMP signaling in the placode. Together, our findings suggest that the lens placode plays an active role in remodeling the optic ECM during early eye development.


Subject(s)
Extracellular Matrix , Gene Expression Regulation, Developmental , Lens, Crystalline , PAX6 Transcription Factor , Animals , Extracellular Matrix/metabolism , Mice , Lens, Crystalline/metabolism , Lens, Crystalline/growth & development , Lens, Crystalline/cytology , PAX6 Transcription Factor/metabolism , PAX6 Transcription Factor/genetics , Eye Proteins/metabolism , Eye Proteins/genetics , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Chick Embryo , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction , Chickens/genetics , Eye/metabolism , Eye/growth & development , Eye/embryology
8.
Mol Cells ; 47(6): 100068, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759887

ABSTRACT

The coordinated movement of germ layer progenitor cells reaches its peak at the dorsal side, where the Bmp signaling gradient is low, and minimum at the ventral side, where the Bmp gradient is high. This dynamic cell movement is regulated by the interplay of various signaling pathways. The noncanonical Wnt signaling cascade serves as a pivotal regulator of convergence and extension cell movement, facilitated by the activation of small GTPases such as Rho, Rab, and Rac. However, the underlying cause of limited cell movement at the ventral side remains elusive. To explore the functional role of a key regulator in constraining gastrulation cell movement at the ventral side, we investigated the Bmp4-direct target gene, sizzled (szl), to assess its potential role in inhibiting noncanonical Wnt signaling. In our current study, we demonstrated that ectopic expression of szl led to gastrulation defects in a dose-dependent manner without altering cell fate specification. Overexpression of szl resulted in decreased elongation of Activin-treated animal cap and Keller explants. Furthermore, our immunoprecipitation assay unveiled the physical interaction of Szl with noncanonical Wnt ligand proteins (Wnt5 and Wnt11). Additionally, the activation of small GTPases involved in Wnt signaling mediation (RhoA and Rac1) was diminished upon szl overexpression. In summary, our findings suggest that Bmp4 signaling negatively modulates cell movement from the ventral side of the embryo by inducing szl expression during early Xenopus gastrulation.


Subject(s)
Bone Morphogenetic Protein 4 , Cell Movement , Gastrulation , Xenopus Proteins , Xenopus laevis , Animals , Bone Morphogenetic Protein 4/metabolism , Ligands , Wnt Proteins/metabolism , Wnt Signaling Pathway , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
9.
Dev Cell ; 59(15): 1940-1953.e10, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38776925

ABSTRACT

During neural tube (NT) development, the notochord induces an organizer, the floorplate, which secretes Sonic Hedgehog (SHH) to pattern neural progenitors. Conversely, NT organoids (NTOs) from embryonic stem cells (ESCs) spontaneously form floorplates without the notochord, demonstrating that stem cells can self-organize without embryonic inducers. Here, we investigated floorplate self-organization in clonal mouse NTOs. Expression of the floorplate marker FOXA2 was initially spatially scattered before resolving into multiple clusters, which underwent competition and sorting, resulting in a stable "winning" floorplate. We identified that BMP signaling governed long-range cluster competition. FOXA2+ clusters expressed BMP4, suppressing FOXA2 in receiving cells while simultaneously expressing the BMP-inhibitor NOGGIN, promoting cluster persistence. Noggin mutation perturbed floorplate formation in NTOs and in the NT in vivo at mid/hindbrain regions, demonstrating how the floorplate can form autonomously without the notochord. Identifying the pathways governing organizer self-organization is critical for harnessing the developmental plasticity of stem cells in tissue engineering.


Subject(s)
Bone Morphogenetic Protein 4 , Neural Tube , Notochord , Organoids , Animals , Mice , Organoids/metabolism , Organoids/cytology , Neural Tube/metabolism , Neural Tube/cytology , Notochord/metabolism , Notochord/cytology , Bone Morphogenetic Protein 4/metabolism , Signal Transduction , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Gene Expression Regulation, Developmental , Bone Morphogenetic Proteins/metabolism
10.
Brain Behav Immun ; 119: 836-850, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735405

ABSTRACT

INTRODUCTION: During postherpetic neuralgia (PHN), the cerebral spinal fluid (CSF) possesses the capability to trigger glial activation and inflammation, yet the specific changes in its composition remain unclear. Recent findings from our research indicate elevations of central bone morphogenetic protein 4 (BMP4) during neuropathic pain (NP), serving as an independent modulator of glial cells. Herein, the aim of the present study is to test the CSF-BMP4 expressions and its role in the glial modulation in the process of PHN. METHODS: CSF samples were collected from both PHN patients and non-painful individuals (Control) to assess BMP4 and its antagonist Noggin levels. Besides, intrathecal administration of both CSF types was conducted in normal rats to evaluate the impact on pain behavior, glial activity, and inflammation.; Additionally, both Noggin and STAT3 antagonist-Stattic were employed to treat the PHN-CSF or exogenous BMP4 challenged cultured astrocytes to explore downstream signals. Finally, microglial depletion was performed prior to the PHN-CSF intervention so as to elucidate the microglia-astrocyte crosstalk. RESULTS: BMP4 levels were significantly higher in PHN-CSF compared to Control-CSF (P < 0.001), with a positive correlation with pain duration (P < 0.05, r = 0.502). Comparing with the Control-CSF producing moderate paw withdrawal threshold (PWT) decline and microglial activation, PHN-CSF further exacerbated allodynia and triggered both microglial and astrocytic activation (P < 0.05). Moreover, PHN-CSF rather than Control-CSF evoked microglial proliferation and pro-inflammatory transformation, reinforced iron storage, and activated astrocytes possibly through both SMAD159 and STAT3 signaling, which were all mitigated by the Noggin application (P < 0.05). Next, both Noggin and Stattic effectively attenuated BMP4-induced GFAP and IL-6 upregulation, as well as SMAD159 and STAT3 phosphorylation in the cultured astrocytes (P < 0.05). Finally, microglial depletion diminished PHN-CSF induced astrogliosis, inflammation and endogenous BMP4 expression (P < 0.05). CONCLUSION: Our study highlights the role of CSF-BMP4 elevation in glial activation and allodynia during PHN, suggesting a potential therapeutic avenue for future exploration.


Subject(s)
Astrocytes , Bone Morphogenetic Protein 4 , Hyperalgesia , Microglia , Neuralgia, Postherpetic , Animals , Microglia/metabolism , Astrocytes/metabolism , Bone Morphogenetic Protein 4/metabolism , Male , Rats , Humans , Aged , Neuralgia, Postherpetic/cerebrospinal fluid , Neuralgia, Postherpetic/metabolism , Female , Hyperalgesia/metabolism , Middle Aged , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Carrier Proteins/metabolism
11.
Environ Pollut ; 351: 124101, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38710361

ABSTRACT

Both nanoplastics (NPs) and 3-tert-butyl-4-hydroxyanisole (3-BHA) are environmental contaminants that can bio-accumulate through the food chain. However, the combined effects of which on mammalian female reproductive system remain unclear. Here, the female ICR-CD1 mice were used to evaluate the damage effects of ovaries and uterus after NPs and 3-BHA co-treatment for 35 days. Firstly, co-exposure significantly reduced the body weight and organ index of ovaries and uterus in mice. Secondly, combined effects of NPs and 3-BHA exacerbated the histopathological abnormalities to the ovaries and uterus and decreased female sex hormones such as FSH and LH while increased antioxidant activities including CAT and GSH-Px. Moreover, the apoptotic genes, inflammatory cytokines and the key reproductive development genes such as FSTL1 were significantly up-regulated under co-exposure conditions. Thirdly, through transcriptional and bioinformatics analysis, immunofluorescence and western blotting assays, together with molecular docking simulation, we determined that co-exposure up-regulated the FSTL1, TGF-ß and p-Smad1/5/9 but down-regulated the expression of BMP4. Finally, the pharmacological rescue experiments further demonstrated that co-exposure of NPs and 3-BHA mainly exacerbated the female reproductive toxicity through FSTL1-mediated BMP4/TGF-ß/SMAD signaling pathway. Taken together, our studies provided the theoretical basis of new environmental pollutants on the reproductive health in female mammals.


Subject(s)
Mice, Inbred ICR , Ovary , Polystyrenes , Uterus , Animals , Female , Mice , Uterus/drug effects , Uterus/metabolism , Ovary/drug effects , Ovary/metabolism , Polystyrenes/toxicity , Reproduction/drug effects , Microplastics/toxicity , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein 4/metabolism , Nanoparticles/toxicity , Molecular Docking Simulation , Environmental Pollutants/toxicity , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/genetics
12.
Food Chem Toxicol ; 189: 114772, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38821392

ABSTRACT

Exposure to plastic-derived estrogen-mimicking endocrine-disrupting bisphenols can have a long-lasting effect on bone health. However, gestational exposure to bisphenol A (BPA) and its analogue, bisphenol S (BPS), on offspring's bone mineralization is unclear. The effects of in-utero bisphenol exposure were examined on the offspring's bone parameters. BPA and BPS (0.0, 0.4 µg/kg bw) were administered to pregnant Wistar rats via oral gavage from gestational day 4-21. Maternal exposure to BPA and BPS increased bone mineral content and density in the offspring aged 30 and 90 days (P < 0.05). Plasma analysis revealed that alkaline phosphatase, and Gla-type osteocalcin were significantly elevated in the BPS-exposed offspring (P < 0.05). The expression of BMP1, BMP4, and their signaling mediators SMAD1 mRNAs were decreased in BPS-exposed osteoblast SaOS-2 cells (P < 0.05). The expression of extracellular matrix proteins such as ALPL, COL1A1, DMP1, and FN1 were downregulated (P < 0.05). Bisphenol co-incubation with noggin decreased TGF-ß1 expression, indicating its involvement in bone mineralization. Altered mineralization could be due to dysregulated expression of bone morphogenetic proteins and signalling mediators in the osteoblast cells. Thus, bisphenol exposure during gestation altered growth and bone mineralization in the offspring, possibly by modulating the expression of Smad-dependent BMP/TGF-ß1 signalling mediators.


Subject(s)
Benzhydryl Compounds , Calcification, Physiologic , Phenols , Prenatal Exposure Delayed Effects , Rats, Wistar , Sulfones , Animals , Phenols/toxicity , Benzhydryl Compounds/toxicity , Female , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Calcification, Physiologic/drug effects , Rats , Sulfones/toxicity , Humans , Smad1 Protein/metabolism , Smad1 Protein/genetics , Alkaline Phosphatase/metabolism , Alkaline Phosphatase/blood , Maternal Exposure/adverse effects , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Osteocalcin/metabolism , Osteocalcin/genetics , Bone Morphogenetic Protein 1/metabolism , Bone Morphogenetic Protein 1/genetics , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Bone Density/drug effects , Endocrine Disruptors/toxicity , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Carrier Proteins
13.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727271

ABSTRACT

Vascular smooth muscle cells (VSMCs) play a key role in aortic aneurysm formation. Bone morphogenetic proteins (BMPs) have been implicated as important regulators of VSMC phenotype, and dysregulation of the BMP pathway has been shown to be associated with vascular diseases. The aim of this study was to investigate for the first time the effects of BMP-4 on the VSMC phenotype and to understand its role in the development of thoracic aortic aneurysms (TAAs). Using the angiotensin II (AngII) osmotic pump model in mice, aortas from mice with VSMC-specific BMP-4 deficiency showed changes similar to AngII-infused aortas, characterised by a loss of contractile markers, increased fibrosis, and activation of matrix metalloproteinase 9. When BMP-4 deficiency was combined with AngII infusion, there was a significantly higher rate of apoptosis and aortic dilatation. In vitro, VSMCs with mRNA silencing of BMP-4 displayed a dedifferentiated phenotype with activated canonical BMP signalling. In contrast, BMP-2-deficient VSMCs exhibited the opposite phenotype. The compensatory regulation between BMP-2 and BMP-4, with BMP-4 promoting the contractile phenotype, appeared to be independent of the canonical signalling pathway. Taken together, these results demonstrate the impact of VSMC-specific BMP-4 deficiency on TAA development.


Subject(s)
Aortic Aneurysm, Thoracic , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Male , Mice , Angiotensin II/pharmacology , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Apoptosis/drug effects , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction
14.
Cell Biochem Funct ; 42(4): e4068, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38817105

ABSTRACT

Evidence is accumulating that osteal macrophages, in addition to bone-resorbing osteoclasts and bone-forming osteoblasts, participate vitally in bone remodeling process. Oncostatin M (OSM), an inflammatory cytokine belonging to interleukin-6 superfamily, is recognized as an essential factor secreted by osteal macrophages to orchestrate bone remodeling. Osteoprotegerin (OPG) produced by osteoblasts regulates osteoclastogenesis. We have reported that bone morphogenetic protein-4 (BMP-4) stimulates OPG synthesis in MC3T3-E1 osteoblast-like cells, and that SMAD1/5/8(9), p38 mitogen-activated protein kinase (MAPK), and p70 S6 kinase are involved in the OPG synthesis. The present study aims to investigate the effect of OSM on the synthesis of OPG stimulated by BMP-4 in osteoblasts. OSM suppressed the release and the mRNA expression of OPG upregulated by BMP-4 in MC3T3-E1 cells. Neither the BMP-4-induced phosphorylation of SMAD1/5/9 nor that of p38 MAPK was affected by OSM. On the other hand, the phosphorylation of p70 S6 kinase stimulated by BMP-4 was considerably suppressed by OSM. These results strongly suggest that OSM suppresses the BMP-4-stimulated OPG synthesis via inhibition of the p70 S6 kinase-mediated pathway in osteoblast-like cells.


Subject(s)
Bone Morphogenetic Protein 4 , Oncostatin M , Osteoblasts , Osteoprotegerin , Ribosomal Protein S6 Kinases, 70-kDa , Animals , Mice , Oncostatin M/pharmacology , Oncostatin M/metabolism , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoprotegerin/metabolism , Osteoprotegerin/biosynthesis , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Cell Line
15.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38752427

ABSTRACT

Bone morphogenic protein (BMP) signaling plays an essential and highly conserved role in embryo axial patterning in animal species. However, in mammalian embryos, which develop inside the mother, early development includes a preimplantation stage, which does not occur in externally developing embryos. During preimplantation, the epiblast is segregated from extra-embryonic lineages that enable implantation and development in utero. Yet, the requirement for BMP signaling is imprecisely defined in mouse early embryos. Here, we show that, in contrast to previous reports, BMP signaling (SMAD1/5/9 phosphorylation) is not detectable until implantation when it is detected in the primitive endoderm - an extra-embryonic lineage. Moreover, preimplantation development appears to be normal following deletion of maternal and zygotic Smad4, an essential effector of canonical BMP signaling. In fact, mice lacking maternal Smad4 are viable. Finally, we uncover a new requirement for zygotic Smad4 in epiblast scaling and cavitation immediately after implantation, via a mechanism involving FGFR/ERK attenuation. Altogether, our results demonstrate no role for BMP4/SMAD4 in the first lineage decisions during mouse development. Rather, multi-pathway signaling among embryonic and extra-embryonic cell types drives epiblast morphogenesis postimplantation.


Subject(s)
Embryo Implantation , Germ Layers , Morphogenesis , Signal Transduction , Smad4 Protein , Animals , Smad4 Protein/metabolism , Smad4 Protein/genetics , Germ Layers/metabolism , Embryo Implantation/genetics , Mice , Morphogenesis/genetics , Female , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Developmental , Embryonic Development/genetics , Mice, Knockout , Embryo, Mammalian/metabolism , Endoderm/metabolism , Endoderm/embryology , Blastocyst/metabolism , Blastocyst/cytology
16.
Anat Histol Embryol ; 53(3): e13044, 2024 May.
Article in English | MEDLINE | ID: mdl-38695121

ABSTRACT

The vitamin D receptor (VDR) signalling has been implicated in vertebrate limb or fin formation. However, the involvement of VDR signalling in the early stages of limb/fin development remains to be elucidated. In this study, the role of VDR signalling in pectoral fin development was investigated in zebrafish embryos. Knockdown of vdr induced the severe impairment of pectoral fin development. The zebrafish larvae lacking vdr exhibited reduced pectoral fins with no skeletal elements. In situ hybridization revealed depletion of vdr downregulated fibroblast growth factor 24 (fgf24), a marker of early pectoral fin bud mesenchyme, in the presumptive fin field even before fin buds were visible. Moreover, a perturbed expression pattern of bone morphogenetic protein 4 (bmp4), a marker of the pectoral fin fold, was observed in the developing fin buds of zebrafish embryos that lost the vdr function. These findings suggest that VDR signalling is crucial in the early stages of fin development, potentially influencing the process by regulating other signalling molecules such as Fgf24 and Bmp4.


Subject(s)
Animal Fins , Bone Morphogenetic Protein 4 , Fibroblast Growth Factors , Receptors, Calcitriol , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/embryology , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Animal Fins/embryology , Animal Fins/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Gene Knockdown Techniques , Signal Transduction , Gene Expression Regulation, Developmental , In Situ Hybridization
17.
Cell Death Dis ; 15(4): 301, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684650

ABSTRACT

Understanding the mechanisms involved in colonic epithelial differentiation is key to unraveling the alterations causing inflammatory conditions and cancer. Organoid cultures provide an unique tool to address these questions but studies are scarce. We report a differentiation system toward enterocytes and goblet cells, the two major colonic epithelial cell lineages, using colon organoids generated from healthy tissue of colorectal cancer patients. Culture of these organoids in medium lacking stemness agents resulted in a modest ultrastructural differentiation phenotype with low-level expression of enterocyte (KLF4, KRT20, CA1, FABP2) and goblet cell (TFF2, TFF3, AGR2) lineage markers. BMP pathway activation through depletion of Noggin and addition of BMP4 resulted in enterocyte-biased differentiation. Contrarily, blockade of the Notch pathway using the γ-secretase inhibitor dibenzazepine (DBZ) favored goblet cell differentiation. Combination treatment with BMP4 and DBZ caused a balanced strong induction of both lineages. In contrast, colon tumor organoids responded poorly to BMP4 showing only weak signals of cell differentiation, and were unresponsive to DBZ. We also investigated the effects of 1α,25-dihydroxyvitamin D3 (calcitriol) on differentiation. Calcitriol attenuated the effects of BMP4 and DBZ on colon normal organoids, with reduced expression of differentiation genes and phenotype. Consistently, in normal organoids, calcitriol inhibited early signaling by BMP4 as assessed by reduction of the level of phospho-SMAD1/5/8. Our results show that BMP and Notch signaling play key roles in human colon stem cell differentiation to the enterocytic and goblet cell lineages and that calcitriol modulates these processes favoring stemness features.


Subject(s)
Bone Morphogenetic Protein 4 , Calcitriol , Carrier Proteins , Cell Differentiation , Colon , Dibenzazepines , Goblet Cells , Kruppel-Like Factor 4 , Organoids , Receptors, Notch , Signal Transduction , Humans , Organoids/drug effects , Organoids/metabolism , Cell Differentiation/drug effects , Bone Morphogenetic Protein 4/metabolism , Colon/drug effects , Colon/metabolism , Colon/cytology , Colon/pathology , Receptors, Notch/metabolism , Signal Transduction/drug effects , Calcitriol/pharmacology , Goblet Cells/drug effects , Goblet Cells/metabolism , Dibenzazepines/pharmacology , Cell Lineage/drug effects , Enterocytes/metabolism , Enterocytes/drug effects , Enterocytes/cytology , Vitamin D/pharmacology
18.
Cell Mol Gastroenterol Hepatol ; 18(2): 101347, 2024.
Article in English | MEDLINE | ID: mdl-38670488

ABSTRACT

BACKGROUND & AIM: Telocytes, a recently identified type of subepithelial interstitial cell, have garnered attention for their potential roles in tissue homeostasis and repair. However, their contribution to gastric metaplasia remains unexplored. This study elucidates the role of telocytes in the development of metaplasia within the gastric environment. METHODS: To investigate the presence and behavior of telocytes during metaplastic transitions, we used drug-induced acute injury models (using DMP-777 or L635) and a genetically engineered mouse model (Mist1-Kras). Lineage tracing via the Foxl1-CreERT2;R26R-tdTomato mouse model was used to track telocyte migratory dynamics. Immunofluorescence staining was used to identify telocyte markers and evaluate their correlation with metaplasia-related changes. RESULTS: We confirmed the existence of FOXL1+/PDGFRα+ double-positive telocytes in the stomach's isthmus region. As metaplasia developed, we observed a marked increase in the telocyte population. The distribution of telocytes expanded beyond the isthmus to encompass the entire gland and closely reflected the expansion of the proliferative cell zone. Rather than a general response to mucosal damage, the shift in telocyte distribution was associated with the establishment of a metaplastic cell niche at the gland base. Furthermore, lineage-tracing experiments highlighted the active recruitment of telocytes to the emerging metaplastic cell niche, and we observed expression of Wnt5a, Bmp4, and Bmp7 in PDGFRα+ telocytes. CONCLUSIONS: These results suggest that telocytes contribute to the evolution of a gastric metaplasia niche. The dynamic behavior of these stromal cells, their responsiveness to metaplastic changes, and potential association with Wnt5a, Bmp4, and Bmp7 signaling emphasize the significance of telocytes in tissue adaptation and repair.


Subject(s)
Bone Morphogenetic Protein 4 , Gastric Mucosa , Metaplasia , Receptor, Platelet-Derived Growth Factor alpha , Telocytes , Wnt-5a Protein , Animals , Metaplasia/pathology , Mice , Telocytes/metabolism , Telocytes/pathology , Wnt-5a Protein/metabolism , Gastric Mucosa/pathology , Gastric Mucosa/metabolism , Bone Morphogenetic Protein 4/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stomach/pathology , Bone Morphogenetic Protein 7/metabolism , Cell Movement , Mice, Transgenic , Disease Models, Animal , Forkhead Transcription Factors
19.
Tissue Cell ; 88: 102376, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38608407

ABSTRACT

OBJECTIVE: Heterotopic ossification (HO), also known as ossifying myositis, is a condition that produces abnormal bone and cartilage tissue in the soft tissues. Hypoxia inducible factor lα (HIF-lα) regulates the expression of various genes, which is closely related to the promotion of bone formation, and Drosophila mothers against decapentaplegic protein (SMAD) mediates the signal transduction in the Bone morphogenetic protein (BMP) signaling pathway, which affects the function of osteoblasts and osteoclasts, and thus plays a key role in the regulation of bone remodeling. We aimed to investigate the mechanism by which HIF-1α induces osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in a hypoxic environment. METHODS: A cellular hypoxia model was constructed to verify the expression of HIF-1α, while alizarin red staining was performed to observe the osteogenic differentiation ability of bone marrow mesenchymal stem cells (BMSCs). Alizarin red staining was used to analyze the late mineralization ability of the cells. Western blot analysis was performed to analyze the expression levels of osteogenesis-related factors OCN, OPN proteins as well as the pathway proteins BMP4, p-Smad1/5/8, and Smad1. We also constructed a rat model of ectopic bone formation, observed ectopic ossification by X-ray, and verified the success of the rat model by ELISA of HIF-1α. HE staining was used to observe the matrix and trabecular structure of bone, and Masson staining was used to observe the collagen and trabecular structure of bone. Immunohistochemistry analyzed the expression of OCN and OPN in ectopic bone tissues, and WB analyzed the expression of pathway proteins BMP4, p-Smad1/5/8 and Smad1 in ectopic bone tissues to verify the signaling pathway of ectopic bone formation. RESULTS: Our results indicate that hypoxic environment upregulates HIF-1a expression and activates BMP4/SMAD signaling pathway. This led to an increase in ALP content and enhanced expression of the osteogenesis-related factors OCN and OPN, resulting in enhanced osteogenic differentiation of BMSCs. The results of our in vivo experiments showed that rats inoculated with BMSCs overexpressing HIF-1α showed bony structures in tendon tissues, enhanced expression of the bone signaling pathways BMP4 and p-Smad1/5/8, and enhanced expression levels of the osteogenic-related factors OCN and OPN, resulting in the formation of ectopic bone. CONCLUSIONS: These data further suggest a novel mechanistic view that hypoxic bone marrow BMSCs activate the BMP4/SMAD pathway by up-regulating the expression level of HIF-1α, thereby promoting the secretion of osteogenic factors leading to ectopic bone formation.


Subject(s)
Bone Morphogenetic Protein 4 , Cell Differentiation , Cell Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit , Mesenchymal Stem Cells , Osteogenesis , Signal Transduction , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Rats , Bone Morphogenetic Protein 4/metabolism , Smad Proteins/metabolism , Rats, Sprague-Dawley , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Male
20.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38673725

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) offer a promising source for generating dental epithelial (DE) cells. Whereas the existing differentiation protocols were time-consuming and relied heavily on growth factors, herein, we developed a three-step protocol to convert hiPSCs into DE cells in 8 days. In the first phase, hiPSCs were differentiated into non-neural ectoderm using SU5402 (an FGF signaling inhibitor). The second phase involved differentiating non-neural ectoderm into pan-placodal ectoderm and simultaneously inducing the formation of oral ectoderm (OE) using LDN193189 (a BMP signaling inhibitor) and purmorphamine (a SHH signaling activator). In the final phase, OE cells were differentiated into DE through the application of Purmorphamine, XAV939 (a WNT signaling inhibitor), and BMP4. qRT-PCR and immunostaining were performed to examine the expression of lineage-specific markers. ARS staining was performed to evaluate the formation of the mineralization nodule. The expression of PITX2, SP6, and AMBN, the emergence of mineralization nodules, and the enhanced expression of AMBN and AMELX in spheroid culture implied the generation of DE cells. This study delineates the developmental signaling pathways and uses small molecules to streamline the induction of hiPSCs into DE cells. Our findings present a simplified and quicker method for generating DE cells, contributing valuable insights for dental regeneration and dental disease research.


Subject(s)
Cell Differentiation , Epithelial Cells , Induced Pluripotent Stem Cells , Morpholines , Purines , Pyrimidines , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation/drug effects , Epithelial Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Tooth/cytology , Ectoderm/cytology , Ectoderm/metabolism , Cells, Cultured , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Pyrazoles/pharmacology , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL