Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 567
Filter
1.
Neoplasia ; 52: 100997, 2024 06.
Article in English | MEDLINE | ID: mdl-38669760

ABSTRACT

Neurodevelopmental cell communication plays a crucial role in neuroblastoma prognosis. However, determining the impact of these communication pathways on prognosis is challenging due to limited sample sizes and patchy clinical survival information of single cell RNA-seq data. To address this, we have developed the cell communication pathway prognostic model (CCPPM) in this study. CCPPM involves the identification of communication pathways through single-cell RNA-seq data, screening of prognosis-significant pathways using bulk RNA-seq data, conducting functional and attribute analysis of these pathways, and analyzing the post-effects of communication within these pathways. By employing the CCPPM, we have identified ten communication pathways significantly influencing neuroblastoma, all related to axongenesis and neural projection development, especially the BMP7-(BMPR1B-ACVR2B) communication pathway was found to promote tumor cell migration by activating the transcription factor SMAD1 and regulating UNK and MYCBP2. Notably, BMP7 expression was higher in neuroblastoma samples with distant metastases. In summary, CCPPM offers a novel approach to studying the influence of cell communication pathways on disease prognosis and identified detrimental communication pathways related to neurodevelopment.


Subject(s)
Cell Communication , Neuroblastoma , Signal Transduction , Neuroblastoma/pathology , Neuroblastoma/metabolism , Neuroblastoma/genetics , Humans , Prognosis , Gene Expression Regulation, Neoplastic , Single-Cell Analysis/methods , Computational Biology/methods , Cell Line, Tumor , Gene Expression Profiling , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/genetics , Cell Movement
2.
Cell Rep ; 43(5): 114162, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38678558

ABSTRACT

Zebrafish have a lifelong cardiac regenerative ability after damage, whereas mammals lose this capacity during early postnatal development. This study investigated whether the declining expression of growth factors during postnatal mammalian development contributes to the decrease of cardiomyocyte regenerative potential. Besides confirming the proliferative ability of neuregulin 1 (NRG1), interleukin (IL)1b, receptor activator of nuclear factor kappa-Β ligand (RANKL), insulin growth factor (IGF)2, and IL6, we identified other potential pro-regenerative factors, with BMP7 exhibiting the most pronounced efficacy. Bmp7 knockdown in neonatal mouse cardiomyocytes and loss-of-function in adult zebrafish during cardiac regeneration reduced cardiomyocyte proliferation, indicating that Bmp7 is crucial in the regenerative stages of mouse and zebrafish hearts. Conversely, bmp7 overexpression in regenerating zebrafish or administration at post-mitotic juvenile and adult mouse stages, in vitro and in vivo following myocardial infarction, enhanced cardiomyocyte cycling. Mechanistically, BMP7 stimulated proliferation through BMPR1A/ACVR1 and ACVR2A/BMPR2 receptors and downstream SMAD5, ERK, and AKT signaling. Overall, BMP7 administration is a promising strategy for heart regeneration.


Subject(s)
Bone Morphogenetic Protein 7 , Cell Proliferation , Myocytes, Cardiac , Regeneration , Zebrafish , Animals , Zebrafish/metabolism , Myocytes, Cardiac/metabolism , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/genetics , Mice , Signal Transduction , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Neuregulin-1/metabolism , Neuregulin-1/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type I/genetics , Mice, Inbred C57BL , Smad5 Protein/metabolism
3.
Int J Nanomedicine ; 19: 3475-3495, 2024.
Article in English | MEDLINE | ID: mdl-38623080

ABSTRACT

Purpose: Human umbilical cord mesenchymal stem cell (hucMSC)-derived small extracellular vesicles (sEVs) are natural nanocarriers with promising potential in treating liver fibrosis and have widespread applications in the fields of nanomedicine and regenerative medicine. However, the therapeutic efficacy of natural hucMSC-sEVs is currently limited owing to their non-specific distribution in vivo and partial removal by mononuclear macrophages following systemic delivery. Thus, the therapeutic efficacy can be improved through the development of engineered hucMSC-sEVs capable to overcome these limitations. Patients and Methods: To improve the anti-liver fibrosis efficacy of hucMSC-sEVs, we genetically engineered hucMSC-sEVs to overexpress the anti-fibrotic gene bone morphogenic protein 7 (BMP7) in parental cells. This was achieved using lentiviral transfection, following which BMP7-loaded hucMSC-sEVs were isolated through ultracentrifugation. First, the liver fibrosis was induced in C57BL/6J mice by intraperitoneal injection of 50% carbon tetrachloride (CCL4) twice a week for 8 weeks. These mice were subsequently treated with BMP7+sEVs via tail vein injection, and the anti-liver fibrosis effect of BMP7+sEVs was validated using small animal in vivo imaging, immunohistochemistry (IHC), tissue immunofluorescence, and enzyme-linked immunosorbent assay (ELISA). Finally, cell function studies were performed to confirm the in vivo results. Results: Liver imaging and liver histopathology confirmed that the engineered hucMSC-sEVs could reach the liver of mice and aggregate around activated hepatic stellate cells (aHSCs) with a significantly stronger anti-liver fibrosis effect of BMP7-loaded hucMSC-sEVs compared to those of blank or negative control-transfected hucMSC-sEVs. In vitro, BMP7-loaded hucMSC-sEVs promoted the phenotypic reversal of aHSCs and inhibited their proliferation to enhance the anti-fibrotic effects. Conclusion: These engineered BMP7-loaded hucMSC-sEVs offer a novel and promising strategy for the clinical treatment of liver fibrosis.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Mice , Humans , Hepatic Stellate Cells/pathology , Mice, Inbred C57BL , Liver Cirrhosis/chemically induced , Liver Cirrhosis/therapy , Liver Cirrhosis/metabolism , Fibrosis , Extracellular Vesicles/pathology , Mesenchymal Stem Cells/metabolism , Umbilical Cord , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism
4.
J Pharm Pharmacol ; 76(6): 656-671, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38429940

ABSTRACT

BACKGROUND: We administered Bushen Huoxue Huazhuo Formula (BSHXHZF) and transplanted bone marrow mesenchymal stem cells (BMSCs) into mice with Wilson's disease (WD)-related liver fibrosis to evaluate the liver-protecting mechanism of this prescription. METHODS: Mice, randomly divided into different treatment groups, showed histopathological changes and degree of hepatocyte apoptosis. For hepatic hydroxyproline (Hyp) determination, transforming growth factor-ß1 (TGF-ß1) and bone morphogenetic protein-7 (BMP-7) mRNA and protein were measured. Chemical profiling of the extract of BSHXHZF using The liquid chromatography-mass spectrometry (LC-MS/MS) and revealing its antifibrosis mechanism using metabolomics. RESULTS: TCM+BMSC group livers exhibited few inflammatory cells. TUNEL revealed abundant brown apoptotic cells in model control groups, while the TCM+BMSC groups showed a significant increase in blue negative expression of liver cells. Hyp in toxic milk (TX) mice groups was significantly lower than that in model control groups (MG). Compared with MG, TGF-ß1 expression was significantly lower than all other groups, while BMP-7 expression was significantly higher. Metabolic analysis identified 20 potential biomarkers and 10 key pathways, indicating that BSHXHZF+BMSC intervention has a significant regulatory effect on metabolic disorders of these small molecule substances. CONCLUSION: BSHXHZF combined with BMSCs can inhibit liver fibrosis and hepatocyte apoptosis by improving related metabolic disorders, and achieving therapeutic effects in WD-related liver fibrosis.


Subject(s)
Bone Morphogenetic Protein 7 , Disease Models, Animal , Drugs, Chinese Herbal , Hepatolenticular Degeneration , Liver Cirrhosis , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Metabolomics , Transforming Growth Factor beta1 , Animals , Liver Cirrhosis/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Metabolomics/methods , Drugs, Chinese Herbal/pharmacology , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Hepatolenticular Degeneration/therapy , Hepatolenticular Degeneration/metabolism , Hepatolenticular Degeneration/drug therapy , Bone Morphogenetic Protein 7/metabolism , Transforming Growth Factor beta1/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Apoptosis/drug effects , Medicine, Chinese Traditional/methods , Proton Magnetic Resonance Spectroscopy , Liver/metabolism , Liver/drug effects , Liver/pathology , Hepatocytes/metabolism , Hepatocytes/drug effects , Hydroxyproline/metabolism
5.
Dev Growth Differ ; 66(3): 182-193, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38342985

ABSTRACT

Newts can regenerate functional elbow joints after amputation at the joint level. Previous studies have suggested the potential contribution of cells from residual tendon tissues to joint cartilage regeneration. A serum-free tissue culture system for tendons was established to explore cell dynamics during joint regeneration. Culturing isolated tendons in this system, stimulated by regeneration-related factors, such as fibroblast growth factor (FGF) and platelet-derived growth factor, led to robust cell migration and proliferation. Moreover, cells proliferating in an FGF-rich environment differentiated into Sox9-positive chondrocytes upon BMP7 introduction. These findings suggest that FGF-stimulated cells from tendons may aid in joint cartilage regeneration during functional elbow joint regeneration in newts.


Subject(s)
Bone Morphogenetic Protein 7 , Chondrocytes , Fibroblast Growth Factors , Animals , Cell Differentiation , Chondrocytes/metabolism , Fibroblast Growth Factors/pharmacology , Fibroblast Growth Factors/metabolism , Salamandridae/metabolism , Tendons/metabolism , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology
6.
Hum Cell ; 37(2): 435-450, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38218754

ABSTRACT

Continuously progressive hepatic fibrosis might cause chronic liver diseases, resulting in hepatic failure. The activation of hepatic stellate cells (HSCs) residing in the liver might induce and influence hepatic fibrosis. In the present study, microRNA 3074 (miR-3074) was found increased within transforming growth factor-ß (TGF-ß)-activated HSCs and enriched within the TGF-ß signaling. In activated HSCs by TGF-ß, miR-3074 overexpression aggravated TGF-ß-induced fibrotic changes, whereas miR-3074 inhibition exerted opposite effects. miR-3074 directly targeted bone morphogenetic protein 7 (BMP7) and inhibited BMP7 expression. Under TGF-ß induction, overexpressed BMP7 notably attenuated the promotive roles of miR-3074 overexpression in TGF-ß-activated HSCs. Within carbon tetrachloride (CCl4)-caused liver fibrosis murine model, miR-3074 agomir administration promoted, while LV-BMP7 administration alleviated CCl4-induced fibrotic changes; LV-BMP7 significantly attenuated the effects of miR-3074 agomir. Lastly, mmu-miR-3074 also targeted mouse BMP7 and inhibited mouse BMP7 expression. In conclusion, the miR-3074/BMP7 axis regulates TGF-ß-caused activation of HSCs in vitro and CCl4-caused murine liver fibrosis in vivo. BMP7-mediated Smad1/5/8 activation might be involved.


Subject(s)
Hepatic Stellate Cells , MicroRNAs , Animals , Mice , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/adverse effects , Bone Morphogenetic Protein 7/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/chemically induced , MicroRNAs/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism
7.
Protein Cell ; 15(1): 21-35, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37300483

ABSTRACT

The seat of human intelligence is the human cerebral cortex, which is responsible for our exceptional cognitive abilities. Identifying principles that lead to the development of the large-sized human cerebral cortex will shed light on what makes the human brain and species so special. The remarkable increase in the number of human cortical pyramidal neurons and the size of the human cerebral cortex is mainly because human cortical radial glial cells, primary neural stem cells in the cortex, generate cortical pyramidal neurons for more than 130 days, whereas the same process takes only about 7 days in mice. The molecular mechanisms underlying this difference are largely unknown. Here, we found that bone morphogenic protein 7 (BMP7) is expressed by increasing the number of cortical radial glial cells during mammalian evolution (mouse, ferret, monkey, and human). BMP7 expression in cortical radial glial cells promotes neurogenesis, inhibits gliogenesis, and thereby increases the length of the neurogenic period, whereas Sonic Hedgehog (SHH) signaling promotes cortical gliogenesis. We demonstrate that BMP7 signaling and SHH signaling mutually inhibit each other through regulation of GLI3 repressor formation. We propose that BMP7 drives the evolutionary expansion of the mammalian cortex by increasing the length of the neurogenic period.


Subject(s)
Ependymoglial Cells , Hedgehog Proteins , Animals , Mice , Humans , Ependymoglial Cells/metabolism , Hedgehog Proteins/metabolism , Ferrets/metabolism , Cerebral Cortex , Neurogenesis , Mammals/metabolism , Neuroglia/metabolism , Bone Morphogenetic Protein 7/metabolism
8.
Protein & Cell ; (12): 21-35, 2024.
Article in English | WPRIM (Western Pacific) | ID: wpr-1010788

ABSTRACT

The seat of human intelligence is the human cerebral cortex, which is responsible for our exceptional cognitive abilities. Identifying principles that lead to the development of the large-sized human cerebral cortex will shed light on what makes the human brain and species so special. The remarkable increase in the number of human cortical pyramidal neurons and the size of the human cerebral cortex is mainly because human cortical radial glial cells, primary neural stem cells in the cortex, generate cortical pyramidal neurons for more than 130 days, whereas the same process takes only about 7 days in mice. The molecular mechanisms underlying this difference are largely unknown. Here, we found that bone morphogenic protein 7 (BMP7) is expressed by increasing the number of cortical radial glial cells during mammalian evolution (mouse, ferret, monkey, and human). BMP7 expression in cortical radial glial cells promotes neurogenesis, inhibits gliogenesis, and thereby increases the length of the neurogenic period, whereas Sonic Hedgehog (SHH) signaling promotes cortical gliogenesis. We demonstrate that BMP7 signaling and SHH signaling mutually inhibit each other through regulation of GLI3 repressor formation. We propose that BMP7 drives the evolutionary expansion of the mammalian cortex by increasing the length of the neurogenic period.


Subject(s)
Animals , Mice , Humans , Ependymoglial Cells/metabolism , Hedgehog Proteins/metabolism , Ferrets/metabolism , Cerebral Cortex , Neurogenesis , Mammals/metabolism , Neuroglia/metabolism , Bone Morphogenetic Protein 7/metabolism
9.
Altern Lab Anim ; 51(6): 363-375, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37831588

ABSTRACT

Human umbilical cord-derived mesenchymal stem cells (hucMSCs) can differentiate into multiple cell lineages, but few methods have been developed to generate kidney lineage cells. Due to their human origin, pluripotent nature and immunomodulatory properties, these stem cells are attractive candidates for clinical applications such as the repair or regeneration of damaged organs. This study evaluated the renal differentiation potential of hucMSCs, when exposed for 10 days to optimised concentrations of retinoic acid, activin-A and bone morphogenetic protein-7 (BMP-7) in various combinations, with and without the priming of the cells with a Wnt signalling pathway activator (CHIR99021). The hucMSCs were isolated and characterised according to surface marker expression (CD73, CD90, CD44, CD146 and CD8) and tri-lineage differentiation potential. The expression of key marker genes (OSR1, TBXT, HOXA13, SIX2, PAX2, KRT18 and ZO1) was examined by qRT-PCR. Specific marker protein expression (E-cadherin, cytokeratin-8 and cytokeratin-19) was analysed by immunocytochemistry. CHIR99021-primed cells treated with the retinoic acid, activin-A and BMP-7 cocktail showed epithelial cell-like differentiation - i.e. distinct phenotypic changes, as well as upregulated gene and protein expression, were observed that were consistent with an epithelial cell phenotype. Thus, our results showed that hucMSCs can efficiently differentiate into renal epithelial-like cells. This work may help in the development of focused therapeutic strategies, in which lineage-defined human stem cells can be used for renal regeneration.


Subject(s)
Bone Morphogenetic Protein 7 , Mesenchymal Stem Cells , Humans , Bone Morphogenetic Protein 7/metabolism , Umbilical Cord , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Epithelial Cells , Tretinoin/metabolism , Activins/pharmacology , Activins/metabolism , Cells, Cultured
10.
Genes (Basel) ; 14(6)2023 05 23.
Article in English | MEDLINE | ID: mdl-37372307

ABSTRACT

Bone morphogenetic proteins (BMPs) play important roles in a lot of biological processes, such as bone development, cell proliferation, cell differentiation, growth, etc. However, the functions of abalone BMP genes are still unknown. This study aimed to better understand the characterization and biological function of BMP7 of Haliotis discus hannai (hdh-BMP7) via cloning and sequencing analysis. The coding sequence (CDS) length of hdh-BMP7 is 1251 bp, which encodes 416 amino acids including a signal peptide (1-28 aa), a transforming growth factor-ß (TGF-ß) propeptide (38-272 aa), and a mature TGF-ß peptide (314-416 aa). The analysis of expression showed that hdh-BMP7 mRNA was widely expressed in all the examined tissues of H. discus hannai. Four SNPs were related to growth traits. The results of RNA interference (RNAi) showed that the mRNA expression levels of hdh-BMPR I, hdh-BMPR II, hdh-smad1, and hdh-MHC declined after hdh-BMP7 was silenced. After RNAi experiment for 30 days, the shell length, shell width, and total weight were found to be reduced in H. discus hannai (p < 0.05). The results of real-time quantitative reverse transcription PCR revealed that the hdh-BMP7 mRNA was lower in abalone of the S-DD-group than in the L-DD-group. Based on these data, we hypothesized that BMP7 gene has a positive role in the growth of H. discus hannai.


Subject(s)
Bone Morphogenetic Protein 7 , Gastropoda , Animals , Bone Morphogenetic Protein 7/metabolism , Gastropoda/genetics , Gastropoda/metabolism , Peptides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Cell Mol Life Sci ; 80(7): 186, 2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37344704

ABSTRACT

Mammalian cardiomyocytes (CMs) undergo maturation during postnatal heart development to meet the increased demands of growth. Here, we found that omentin-1, an adipokine, facilitates CM cell cycle arrest and metabolic maturation. Deletion of omentin-1 causes mouse heart enlargement and dysfunction in adulthood and CM maturation retardation in juveniles, including delayed cell cycle arrest and reduced fatty acid oxidation. Through RNA sequencing, molecular docking analysis, and proximity ligation assays, we found that omentin-1 regulates CM maturation by interacting directly with bone morphogenetic protein 7 (BMP7). Omentin-1 prevents BMP7 from binding to activin type II receptor B (ActRIIB), subsequently decreasing the downstream pathways mothers against DPP homolog 1 (SMAD1)/Yes-associated protein (YAP) and p38 mitogen-activated protein kinase (p38 MAPK). In addition, omentin-1 is required and sufficient for the maturation of human embryonic stem cell-derived CMs. Together, our findings reveal that omentin-1 is a pro-maturation factor for CMs that is essential for postnatal heart development and cardiac function maintenance.


Subject(s)
Bone Morphogenetic Protein 7 , Myocytes, Cardiac , Animals , Humans , Mice , Bone Morphogenetic Protein 7/metabolism , Cell Cycle Checkpoints , Cell Differentiation , Molecular Docking Simulation , Myocytes, Cardiac/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Chem Biol Interact ; 382: 110559, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37247809

ABSTRACT

BACKGROUND: Nephrotic syndrome (NS) is a chronic kidney disease mainly caused by impaired podocytes, ultimately resulting in massive proteinuria or even end-stage renal disease (ESRD). METHODS: The objective of this study was to explore the potential pathogenesis of NS caused by podocyte injury, and further explore the underlying mechanism through data mining, bioinformatics analysis, and experimental verification. The integrated analyses including Seurat, CellChat, gene ontology (GO), and molecular docking were performed based on the single-cell RNA-seq data (scRNA-seq). The adriamycin (ADR)-induced podocyte injury model in vitro was established to conduct the experimental verification for bioinformatics analysis results through western blot and real-time quantitative PCR (RT-qPCR). RESULTS: The results of bioinformatics analysis revealed that the bone morphogenetic protein (BMP) signaling pathway was involved in the podocyte-to-podocyte communication, which plays a crucial role in podocyte injury. The expression of BMP7 was significantly increased in ADR-induced podocytes through activating the Adenosine-monophosphate activated-protein kinase/Mammalian target of rapamycin (AMPK/mTOR) mediated autophagy pathway, and these findings were confirmed by in vitro experiments. CONCLUSION: This study first demonstrated that BMP7 participated in ADR-induced podocyte injury. The BMP7/AMPK/mTOR mediated autophagy pathway may play a crucial role in podocyte injury, which may be the potential therapeutic target for NS patients.


Subject(s)
Podocytes , Animals , Humans , Podocytes/metabolism , Podocytes/pathology , Sirolimus/pharmacology , AMP-Activated Protein Kinases/metabolism , Molecular Docking Simulation , Single-Cell Gene Expression Analysis , TOR Serine-Threonine Kinases/metabolism , Doxorubicin/toxicity , Doxorubicin/metabolism , Mammals/metabolism , Autophagy , Apoptosis , Bone Morphogenetic Protein 7/metabolism
13.
Int J Mol Sci ; 24(7)2023 Apr 02.
Article in English | MEDLINE | ID: mdl-37047609

ABSTRACT

Alterations in the expression of numerous genes and the miRNAs that are recognized as their regulators in the endometrial cells of women with endometriosis may disrupt the intracellular signaling pathways associated with epithelial-mesenchymal transition (EMT). So far, the functional role of BMP7 in endometrial physiology has been confirmed, especially in the context of fertility, but the role of the activation of a specific mechanism operating through the BMP-SMAD-CDH1 axis in the formation of endometrial lesions remains unexplored. The aim of this study was to evaluate the expression profile of miR-542-3p and the EMT markers (BMP7, SMAD4, CDH1) in matched eutopic endometrium (EUE) and ectopic endometrium (ECE) samples from women with endometriosis in relation to healthy women. The levels of expression of the studied genes and miRNA in peripheral blood mononuclear cells (PBMCs) obtained from women diagnosed with endometriosis and those without the disease were also evaluated. Fifty-four patients (n = 54: with endometriosis-n = 29 and without endometriosis-n = 25) were included in the study. A comparative analysis of the relative mean expression values (RQ) of the studied mRNA and miRNA assessed by RT-qPCR demonstrated downregulation of BMP7, SMAD4, and CDH1 expression in ectopic lesions and upregulation in the eutopic endometrium compared with the control group. In the eutopic tissue of women with endometriosis, miR-542-3p expression was similar to that of the control but significantly lower than in endometrial lesions. We also confirmed a trend towards a negative correlation between miR-542-3p and BMP7 in ectopic tissue, and in PBMC, a significant negative correlation of miR-542-3p with further BMP signaling genes, i.e., SMAD4 and CDH1, was observed. These results indicate that the miRNA selected by us may be a potential negative regulator of BMP7-SMAD4-CDH1 signaling associated with EMT. The different patterns of BMP7, SMAD4, and CDH1 gene expression in ECE, EUE, and the control endometrium observed by us suggests the loss of the endometrial epithelium phenotype in women with endometriosis and demonstrates their involvement in the pathogenesis and pathomechanism of this disease.


Subject(s)
Endometriosis , MicroRNAs , Uterine Diseases , Humans , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Endometriosis/metabolism , Leukocytes, Mononuclear/metabolism , Uterine Diseases/pathology , Endometrium/metabolism , Smad4 Protein/genetics , Smad4 Protein/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism
14.
Auton Neurosci ; 247: 103085, 2023 07.
Article in English | MEDLINE | ID: mdl-37031474

ABSTRACT

Autonomic dysfunction has been observed in Alzheimer's disease (AD); however, the effects of genes involved in AD on the peripheral nervous system are not well understood. Previous studies have shown that presenilin-1 (PSEN1), the catalytic subunit of the gamma secretase (γ-secretase) complex, mutations in which are associated with familial AD function, regulates dendritic growth in hippocampal neurons. In this study, we examined whether the γ-secretase pathway also influences dendritic growth in primary sympathetic neurons. Using immunoblotting and immunocytochemistry, molecules of the γ-secretase complex, PSEN1, PSEN2, PEN2, nicastrin and APH1a, were detected in sympathetic neurons dissociated from embryonic (E20/21) rat sympathetic ganglia. Addition of bone morphogenetic protein-7 (BMP-7), which induces dendrites in these neurons, did not alter expression or localization of γ-secretase complex proteins. BMP-7-induced dendritic growth was inhibited by siRNA knockdown of PSEN1 and by three γ-secretase inhibitors, γ-secretase inhibitor IX (DAPT), LY-411575 and BMS-299897. These effects were specific to dendrites and concentration-dependent and did not alter early downstream pathways of BMP signaling. In summary, our results indicate that γ-secretase activity enhances BMP-7 induced dendritic growth in sympathetic neurons. These findings provide insight into the normal cellular role of the γ-secretase complex in sympathetic neurons.


Subject(s)
Amyloid Precursor Protein Secretases , Bone Morphogenetic Protein 7 , Rats , Animals , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/pharmacology , Dendrites/metabolism , Cells, Cultured , Neurons/metabolism
15.
Neurochem Res ; 48(9): 2687-2700, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37071344

ABSTRACT

Excessive activation of pro-inflammatory (M1) microglia phenotypes after spinal cord injury (SCI) disrupts tissue repair and increases the risk of secondary SCI. We previously reported that adeno-associated virus (AAV) mediated delivery of bone morphogenetic protein 7 (BMP7) promotes functional recovery after SCI by reducing oligodendrocyte loss and demyelination; however, little is known about the early effects of BMP7 in ameliorating neuroinflammation in the acute SCI phase. Herein, we demonstrate that treatment with recombinant human BMP7 (rhBMP7) suppresses the viability of LPS-induced HMC3 microglia cells and increases the proportion with the M2 phenotype. Consistently, in a rat SCI model, rhBMP7 decreases the activation of microglia and promotes M2 polarization. After rhBMP7 administration, the STAT3 signaling pathway was activated in LPS-induced HMC3 cells and microglia in spinal cord lesions. Furthermore, the levels of TNF-α and IL-1ß were significantly decreased in cell culture supernatants, lesion sites of injured spinal cords, and cerebrospinal fluid circulation after rhBMP7 administration, thus reducing neuron loss in the injured spinal cord and promoting functional recovery after SCI. These results provide insight into the immediate early mechanisms by which BMP7 may ameliorate the inflammation response to secondary SCI.


Subject(s)
Microglia , Spinal Cord Injuries , Humans , Rats , Animals , Microglia/metabolism , Neuroinflammatory Diseases , Inflammation/metabolism , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology , Lipopolysaccharides/toxicity , Spinal Cord Injuries/pathology , Spinal Cord/metabolism , STAT3 Transcription Factor/metabolism
16.
Mol Med ; 29(1): 30, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36858954

ABSTRACT

BACKGROUND: Accumulating evidence indicates that intervertebral disc degeneration (IDD) is associated with diabetes mellitus (DM), while the underlying mechanisms still remain elusive. Herein, the current study sought to explore the potential molecular mechanism of IDD in diabetic rats based on transcriptome sequencing data. METHODS: Streptozotocin (STZ)-induced diabetes mellitus type 1 (T1DM) rats were used to obtain the nucleus pulposus tissues for transcriptome sequencing. Next, differentially expressed genes (DEGs) in transcriptome sequencing data and GSE34000 microarray dataset were obtained and intersected to acquire the candidate genes. Moreover, GO and KEGG enrichment analyses were performed to analyze the cellular functions and molecular signaling pathways primarily regulated by candidate DEGs. RESULTS: A total of 35 key genes involved in IDD of T1DM rats were mainly enriched in the extracellular matrix (ECM) and cytokine adhesion binding-related pathways. NLRP3 inflammasome activation promoted the pyroptosis of nucleus pulposus cells (NPCs). Besides, BMP7 could affect the IDD of T1DM rats by regulating the inflammatory responses. Additionally, NPCs were isolated from STZ-induced T1DM rats to illustrate the effects of BMP7 on IDD of T1DM rats using the ectopic expression method. Both in vitro and in vivo experiments validated that BMP7 alleviated IDD of T1DM rats by inhibiting NLRP3 inflammasome activation and pyroptosis of NPCs. CONCLUSION: Collectively, our findings provided novel mechanistic insights for understanding of the role of BMP7 in IDD of T1DM, and further highlighted BMP7 as a potential therapeutic target for preventing IDD in T1DM.


Subject(s)
Bone Morphogenetic Protein 7 , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Intervertebral Disc Degeneration , Nucleus Pulposus , Animals , Rats , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Streptozocin , Bone Morphogenetic Protein 7/metabolism
17.
Turk J Med Sci ; 53(1): 10-18, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36945919

ABSTRACT

BACKGROUND: Diabetes is a multifactorial and growing disease, one of the severe complications of which is diabetic nephropathy (DN), which is the most common cause of chronic renal failure. FERM domain containing 3 (FRMD3) is responsible for maintaining the shape and integrity of nephron cells, and bone morphogenetic protein 7 (BMP7) helps maintain function and reduce kidney damage. This study aimed to evaluate the effect of crocin and losartan on biochemical parameters and the expression of FRMD3 and BMP7 genes in streptozotocin (STZ)-induced diabetic rats. METHODS: Forty male Wistar rats were randomly divided into five experimental groups as healthy, diabetic control (D), crocin, losartan, and diabetic rats treated with losartan-crocin (n = 8). A single dose of STZ (50 mg/kg intraperitoneally injection) was used to induce diabetes. Four weeks after induction of diabetes, rats received crocin (50 mg/kg) and losartan (25 mg/kg) daily for four weeks orally. Rats were sacrificed at the end of the intervention, and blood samples were taken to determine serum levels of glucose, urea, creatinine (Cr), malondialdehyde (MDA), and thiol. Real-time polymerase chain reaction (PCR) was used to assess the expression of the FRMD3 and BMP7 genes in the kidney samples. RESULTS: Diabetes induction increased serum levels of glucose, Cr, urea, MDA, and thiol, but decreased BMP7 and FRMD3 genes expression. Treatment with crocin and losartan decreased these biochemical parameters and increased the expression of the BMP7 and FRMD3 genes. DISCUSSION: Crocin may be a promising therapeutic agent for preventing and improving diabetes-related kidney disease due to its antidiabetic and antioxidant properties.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Rats , Male , Animals , Losartan , Rats, Wistar , Diabetes Mellitus, Experimental/metabolism , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/pharmacology , Diabetic Nephropathies/drug therapy , Streptozocin/adverse effects , Glucose/adverse effects , Sulfhydryl Compounds , Oxidative Stress
18.
Int J Mol Sci ; 24(5)2023 Feb 21.
Article in English | MEDLINE | ID: mdl-36901687

ABSTRACT

Bone effects attributed to bisphenols (BPs) include the inhibition of growth and differentiation. This study analyzes the effect of BPA analogs (BPS, BPF, and BPAF) on the gene expression of the osteogenic markers RUNX2, osterix (OSX), bone morphogenetic protein-2 (BMP-2), BMP-7, alkaline phosphatase (ALP), collagen-1 (COL-1), and osteocalcin (OSC). Human osteoblasts were obtained by primary culture from bone chips harvested during routine dental work in healthy volunteers and were treated with BPF, BPS, or BPAF for 24 h at doses of 10-5, 10-6, and 10-7 M. Untreated cells were used as controls. Real-time PCR was used to determine the expression of the osteogenic marker genes RUNX2, OSX, BMP-2, BMP-7, ALP, COL-1, and OSC. The expression of all studied markers was inhibited in the presence of each analog; some markers (COL-1; OSC, BMP2) were inhibited at all three doses and others only at the highest doses (10-5 and 10-6 M). Results obtained for the gene expression of osteogenic markers reveal an adverse effect of BPA analogs (BPF, BPS, and BPAF) on the physiology of human osteoblasts. The impact on ALP, COL-1, and OSC synthesis and therefore on bone matrix formation and mineralization is similar to that observed after exposure to BPA. Further research is warranted to determine the possible contribution of BP exposure to the development of bone diseases such as osteoporosis.


Subject(s)
Bone Morphogenetic Protein 7 , Core Binding Factor Alpha 1 Subunit , Humans , Bone Morphogenetic Protein 7/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/metabolism , Osteogenesis , Gene Expression , Benzhydryl Compounds/pharmacology
19.
Protein Expr Purif ; 206: 106245, 2023 06.
Article in English | MEDLINE | ID: mdl-36805029

ABSTRACT

Human bone morphogenetic protein 2 (hBMP-2) plays a leading role in the process of osteogenesis and is one of the key components of osteoplastic materials, ensuring their high osteoinduction. In order to obtain a homodimeric form hBMP-2 using the E. coli expression system, a number of problems associated with refolding in vitro and purification from monomer and oligomeric forms must be solved. The developed method for co-expression of the target protein with chaperone proteins makes it possible to obtain the biologically active homodimeric form of hBMP-2 in vivo. Purification with simple ion-exchange sorbents without the use of denaturing reagents affecting the structure of the protein molecule provides a chromatographic purity of the product of at least 97%. The expressed hBMP-2 was identified by Western blotting and the LC-ESI-TOF mass spectrometry confirmed its molecular weight of 26052.72 Da. Circular dichroism spectroscopy showed that recombinant hBMP-2 has a native secondary structure.


Subject(s)
Bone Morphogenetic Protein 2 , Escherichia coli , Humans , Bone Morphogenetic Protein 2/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Recombinant Proteins/chemistry , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Osteogenesis , Bone Morphogenetic Protein 7/metabolism
20.
Sci Rep ; 13(1): 2404, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36765143

ABSTRACT

Endothelial dysfunction and inflammatory immune response trigger dedifferentiation of vascular smooth muscle cells (SMCs) from contractile to synthetic phenotype and initiate arterial occlusion. However, the complex vascular remodeling process playing roles in arterial occlusion initiation is largely unknown. We performed bulk sequencing of small and messenger RNAs in a rodent arterial injury model. Bioinformatic data analyses reveal that six miRNAs are overexpressed in injured rat carotids as well as synthetic-type human vascular SMCs. In vitro cell-based assays show that four miRNAs (miR-130b-5p, miR-132-3p, miR-370-3p, and miR-410-3p) distinctly regulate the proliferation of and monocyte adhesion to the vascular SMCs. Individual inhibition of the four selected miRNAs strongly prevents the neointimal hyperplasia in the injured rat carotid arteries. Mechanistically, miR-132-3p and miR-370-3p direct the cell cycle progression, triggering SMC proliferation. Gene ontology analysis of mRNA sequencing data consistently reveal that the miRNA targets include gene clusters that direct proliferation, differentiation, and inflammation. Notably, bone morphogenic protein (BMP)-7 is a prominent target gene of miR-370-3p, and it regulates vascular SMC proliferation in cellular and animal models. Overall, this study first reports that the miR-370-3p/BMP-7 axis determines the vascular SMC phenotype in both rodent and human systems.


Subject(s)
MicroRNAs , Muscle, Smooth, Vascular , Animals , Humans , Rats , Bone Morphogenetic Protein 7/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , MicroRNAs/genetics , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...