Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 16(9): 7870-7888, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38709288

ABSTRACT

BMP9 has demonstrated significant osteogenic potential. In this study, we investigated the effect of Leptin on BMP9-induced osteogenic differentiation. Firstly, we found Leptin was decreased during BMP9-induced osteogenic differentiation and serum Leptin concentrations were increased in the ovariectomized (OVX) rats. Both in vitro and in vivo, exogenous expression of Leptin inhibited the process of osteogenic differentiation, whereas silencing Leptin enhanced. Exogenous Leptin could increase the malonylation of ß-catenin. However, BMP9 could increase the level of Sirt5 and subsequently decrease the malonylation of ß-catenin; the BMP9-induced osteogenic differentiation was inhibited by silencing Sirt5. These data suggested that Leptin can inhibit the BMP9-induced osteogenic differentiation, which may be mediated through reducing the activity of Wnt/ß-catenin signalling via down-regulating Sirt5 to increase the malonylation level of ß-catenin partly.


Subject(s)
Down-Regulation , Growth Differentiation Factor 2 , Leptin , Osteogenesis , Sirtuins , Wnt Signaling Pathway , beta Catenin , Animals , beta Catenin/metabolism , beta Catenin/genetics , Sirtuins/metabolism , Sirtuins/genetics , Female , Rats , Osteogenesis/drug effects , Leptin/metabolism , Leptin/pharmacology , Growth Differentiation Factor 2/metabolism , Wnt Signaling Pathway/drug effects , Ovariectomy , Cell Differentiation/drug effects , Rats, Sprague-Dawley
2.
Int J Biochem Cell Biol ; 164: 106476, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37802385

ABSTRACT

Bone morphogenetic protein 9 (BMP9) has been validated as one of the most potent osteoinduction factors, but its underlying mechanism remains unclear. As a member of the matrix metalloproteinase (MMP) family, MMP13 may be involved in regulating the lineage-specific differentiation of mouse embryonic fibroblasts (MEFs). The goal of this study was to determine whether MMP13 regulates the osteoinduction potential of BMP9 in MEFs, which are multipotent progenitor cells widely used for stem cell biology research. In vitro and in vivo experiments showed that BMP9-induced osteogenic markers and/or bone were enhanced by exogenous MMP13 in MEFs, but were reduced by MMP13 knockdown or inhibition. The expression of hypoxia inducible factor 1 alpha (HIF-1α) was induced by BMP9, which was enhanced by MMP13. The protein expression of ß-catenin and phosphorylation level of glycogen synthase kinase-3 beta (GSK-3ß) were increased by BMP9 in MEFs, as was the translocation of ß-catenin from the cytoplasm to the nucleus; all these effects of BMP9 were enhanced by MMP13. Furthermore, the MMP13 effects of increasing BMP9-induced ß-catenin protein expression and GSK-3ß phosphorylation level were partially reversed by HIF-1α knockdown. These results suggest that MMP13 can enhance the osteoinduction potential of BMP9, which may be mediated, at least in part, through the HIF-1α/ß-catenin axis. Our findings demonstrate a novel role of MMP13 in the lineage decision of progenitor cells and provide a promising strategy to speed up bone regeneration.


Subject(s)
Growth Differentiation Factor 2 , beta Catenin , Animals , Mice , beta Catenin/metabolism , Cell Differentiation , Fibroblasts/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Growth Differentiation Factor 2/pharmacology , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/pharmacology , Osteogenesis , Up-Regulation
3.
Tissue Eng Regen Med ; 20(5): 705-723, 2023 08.
Article in English | MEDLINE | ID: mdl-37010733

ABSTRACT

BACKGROUND: All-trans retinoic acid (ATRA) promotes the osteogenic differentiation induced by bone morphogenetic protein 9 (BMP9), but the intrinsic relationship between BMP9 and ATRA keeps unknown. Herein, we investigated the effect of Cyp26b1, a critical enzyme of ATRA degradation, on the BMP9-induced osteogenic differentiation in mesenchymal stem cells (MSCs), and unveiled possible mechanism through which BMP9 regulates the expression of Cyp26b1. METHODS: ATRA content was detected with ELISA and HPLC-MS/MS. PCR, Western blot, and histochemical staining were used to assay the osteogenic markers. Fetal limbs culture, cranial defect repair model, and micro-computed tomographic were used to evaluate the quality of bone formation. IP and ChIP assay were used to explore possible mechanism. RESULTS: We found that the protein level of Cyp26b1 was increased with age, whereas the ATRA content decreased. The osteogenic markers induced by BMP9 were increased by inhibiting or silencing Cyp26b1 but reduced by exogenous Cyp26b1. The BMP9-induced bone formation was enhanced by inhibiting Cyp26b1. The cranial defect repair was promoted by BMP9, which was strengthened by silencing Cyp26b1 and reduced by exogenous Cyp26b1. Mechanically, Cyp26b1 was reduced by BMP9, which was enhanced by activating Wnt/ß-catenin, and reduced by inhibiting this pathway. ß-catenin interacts with Smad1/5/9, and both were recruited at the promoter of Cyp26b1. CONCLUSIONS: Our findings suggested the BMP9-induced osteoblastic differentiation was mediated by activating retinoic acid signalling, viadown-regulating Cyp26b1. Meanwhile, Cyp26b1 may be a novel potential therapeutic target for the treatment of bone-related diseases or accelerating bone-tissue engineering.


Subject(s)
Growth Differentiation Factor 2 , Mesenchymal Stem Cells , Wnt Signaling Pathway , beta Catenin/metabolism , Growth Differentiation Factor 2/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis , Tandem Mass Spectrometry , Tretinoin/pharmacology
4.
Curr Stem Cell Res Ther ; 18(8): 1160-1171, 2023.
Article in English | MEDLINE | ID: mdl-36941809

ABSTRACT

BACKGROUND: Due to its effective osteogenic ability, BMP9 is a promising candidate for bone regeneration medicine. Whereas, BMP9 can also induce adipogenesis simultaneously. LCN2 is a cytokine associated with osteogenesis and adipogenesis. Reducing the adipogenic potential may be a feasible measure to enhance the osteogenic capability of BMP9. OBJECTIVE: The objective of the study was to explore the role of LCN2 in regulating the BMP9-initialized osteogenic and adipogenic differentiation in mouse embryonic fibroblasts (MEFs), and clarify the possible underlying mechanism. METHODS: Histochemical stain, western blot, real-time PCR, laser confocal, immunoprecipitation, cranial defect repair, and fetal limb culture assays were used to evaluate the effects of LCN2 on BMP9-induced osteogenic and adipogenic differentiation, as well as Wnt/ß-catenin signaling. RESULTS: LCN2 was down-regulated by BMP9. The BMP9-induced osteogenic markers were inhibited by LCN2 overexpression, but the adipogenic markers were increased; LCN2 knockdown exhibited opposite effects. Similar results were found in bone defect repair and fetal limb culture tests. The level of ß-catenin nucleus translocation was found to be reduced by LCN2 overexpression, but increased by LCN2 knockdown. The inhibitory effect of LCN2 overexpression on the osteogenic capability of BMP9 was reversed by ß-catenin overexpression; whereas, the effect of LCN2 knockdown on promoting BMP9 osteogenic potential was almost eliminated by ß-catenin knockdown. LCN2 could bind with LRP6 specifically, and the inhibitory effect of LCN2 on the osteogenic potential of BMP9 could not be enhanced by LRP6 knockdown. CONCLUSION: LCN2 inhibits the BMP9-induced osteogenic differentiation but promotes its adipogenic potential in MEFs, which may be partially mediated by reducing Wnt/ß-catenin signaling via binding with LRP6.


Subject(s)
Growth Differentiation Factor 2 , Osteogenesis , Animals , Mice , Growth Differentiation Factor 2/metabolism , Growth Differentiation Factor 2/pharmacology , beta Catenin/metabolism , Fibroblasts , Wnt Signaling Pathway , Cell Differentiation
5.
Oxid Med Cell Longev ; 2023: 3563663, 2023.
Article in English | MEDLINE | ID: mdl-36778210

ABSTRACT

Studies reported the positive and negative osteogenic effects of MEG3 in mesenchymal stem cells (MSCs). This study aims at clarifying the osteogenic potential of MEG3 and the underlying mechanism. Bone morphogenetic protein 9- (BMP9-) transfected MSCs were recruited as an osteogenic model in vitro, and ectopic bone formation were used in vivo to explore the effect of MEG3 on osteogenesis. We found that overexpression of MEG3 facilitated BMP9-induced osteogenic markers, ALP activities, and matrix mineralization. However, knockdown of MEG3 attenuated BMP9-induced osteogenic markers. MEG3 increased the phosphorylation of GSK-3ß and the protein level of ß-catenin. Pyruvate dehydrogenase kinase 4 (PDK4) can also combine with GSK-3ß and increase the latter phosphorylation. Moreover, MEG3 increased the mRNA level of PDK4. The ceRNA analysis showed that MEG3 may regulate the expression of PDK4 via microRNA 532-5p (miR-532-5p). The MEG3-enhanced GSK-3ß/ß-catenin axis can be attenuated by miR-532-5p, and miR-532-5p inhibitor partly rescued endogenous PDK4 and MEG3-mediated expression of PDK4. MEG3 may potentiate PDK4 and GSK-3ß/ß-catenin by inhibiting miR-532-5p.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Glycogen Synthase Kinase 3 beta/genetics , Cell Differentiation/physiology , RNA, Long Noncoding/genetics , beta Catenin/genetics , beta Catenin/metabolism , Osteogenesis , MicroRNAs/genetics , MicroRNAs/metabolism , Cells, Cultured
6.
Aging (Albany NY) ; 15(3): 718-733, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36750182

ABSTRACT

Interleukin 6 (IL-6) plays a dual role in regulating bone metabolism, although the concrete mechanism is unclear. Bone morphogenetic protein 9 (BMP9) is one of the most potent osteogenic inducers, and a promising alternative for bone tissue engineering. The relationship between IL-6 and BMP9 in osteogenic differentiation remains to be elucidated, and the osteoblastic potential of BMP9 needs to be enhanced to overcome certain shortcomings of BMP9. In this study, we used real-time PCR, western blot, immunofluorescent stain, fetal limb culture and cranial defects repair model to explore the IL-6 role in BMP9-induced osteogenic differentiation in mouse embryonic fibroblasts (MEFs). We found that the rat serum level of IL-6 was increased in the dexamethasone-induced osteoporosis model, and IL-6 expression was detectable in several progenitor cells and MEFs. BMP9 upregulated IL-6 in MEFs, and the BMP9-induced osteoblastic markers were elevated by IL-6, but reduced by IL-6 knockdown. BMP9 and/or IL-6 both activated mTOR, and the IL-6 effect on BMP9-induced osteoblastic markers and bone formation were reduced greatly by mTOR inhibition. Raptor was up-regulated by IL-6 and/or BMP9 specifically, and the osteoblastic markers induced by IL-6 and/or BMP9 were reduced by Raptor knockdown. Meanwhile, Stat-3 was activated by IL-6 and/or BMP9, and the increase of Raptor or osteoblastic markers by IL-6 and/or BMP9 were reduced by Stat-3 inhibition. The Raptor promoter activity was regulated by p-Stat-3. Our finding suggested that IL-6 can promote the BMP9 osteoblastic potential, which may be mediated through activating Stat-3/mTORC1 pathway.


Subject(s)
Growth Differentiation Factor 2 , Interleukin-6 , Animals , Mice , Rats , Cell Differentiation , Fibroblasts/metabolism , Growth Differentiation Factor 2/metabolism , Interleukin-6/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Osteogenesis , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
7.
Regen Biomater ; 10: rbac095, 2023.
Article in English | MEDLINE | ID: mdl-36683747

ABSTRACT

Silk as a natural biomaterial is considered as a promising bone substitute in tissue regeneration. Sericin and fibroin are the main components of silk and display unique features for their programmable mechanical properties, biocompatibility, biodegradability and morphological plasticity. It has been reported that sericin recombinant growth factors (GFs) can support cell proliferation and induce stem cell differentiation through cross-talk of signaling pathways during tissue regeneration. The transgenic technology allows the productions of bioactive heterologous GFs as fusion proteins with sericin, which are then fabricated into solid matrix or hydrogel format. Herein, using an injectable hydrogel derived from transgenic platelet-derived GF (PDGF)-BB silk sericin, we demonstrated that the PDGF-BB sericin hydrogel effectively augmented osteogenesis induced by bone morphogenetic protein (BMP9)-stimulated mesenchymal stem cells (MSCs) in vivo and in vitro, while inhibiting adipogenic differentiation. Further gene expression and protein-protein interactions studies demonstrated that BMP9 and PDGF-BB synergistically induced osteogenic differentiation through the cross-talk between Smad and Stat3 pathways in MSCs. Thus, our results provide a novel strategy to encapsulate osteogenic factors and osteoblastic progenitors in transgenic sericin-based hydrogel for robust bone tissue engineering.

8.
Stem Cells Int ; 2023: 5915988, 2023.
Article in English | MEDLINE | ID: mdl-36698376

ABSTRACT

Mesenchymal stem cells (MSCs) can self-renew and differentiate into multiple lineages, making MSC transplantation a promising option for bone regeneration. Both matricellular proteins and growth factors play an important role in regulating stem cell fate. In this study, we investigated the effects of matricellular protein SMOC2 (secreted modular calcium-binding protein 2) on bone morphogenetic protein 9 (BMP9) in mouse embryonic fibroblasts (MEFs) and revealed a possible molecular mechanism underlying this process. We found that SMOC2 was detectable in MEFs and that exogenous SMOC2 expression potentiated BMP9-induced osteogenic markers, matrix mineralization, and ectopic bone formation, whereas SMOC2 knockdown inhibited these effects. BMP9 increased the levels of p-FAK and p-AKT, which were either enhanced or reduced by SMOC2 and FAK silencing, respectively. BMP9-induced osteogenic markers were increased by SMOC2, and this increase was partially abolished by silencing FAK or LY290042. Furthermore, we found that general transcription factor 2I (GTF2I) was enriched at the promoter region of SMOC2 and that integrin ß1 interacted with SMOC2 in BMP9-treated MEFs. Our findings demonstrate that SMOC2 can promote BMP9-induced osteogenic differentiation by enhancing the FAK/PI3K/AKT pathway, which may be triggered by facilitating the interaction between SMOC2 and integrin ß1.

9.
Int J Biochem Cell Biol ; 154: 106341, 2023 01.
Article in English | MEDLINE | ID: mdl-36442735

ABSTRACT

Bone morphogenetic protein 9 (BMP9) is an effective osteogenic factor and a promising candidate for bone tissue engineering. The osteoblastic potential of BMP9 needs to be further increased to overcome its shortcomings. However, the details of how BMP9 triggers osteogenic differentiation in mesenchymal stem cells (MSCs) are unclear. In this study, we used real-time PCR, western blot, histochemical staining, mouse ectopic bone formation model, immunofluorescence, immunoprecipitation, and chromatin immunoprecipitation to investigate the role of pyruvate dehydrogenase kinase 4 (PDK4) in BMP9-induced osteogenic differentiation of C3H10T1/2 cells, as well as the underlying mechanism. We found that PDK4 was upregulated by BMP9 in C3H10T1/2 cells. BMP9-induced osteogenic markers and bone mass were increased by PDK4 overexpression, but decreased by PDK4 silencing. ß-catenin protein level was increased by BMP9, which was enhanced by PDK overexpression and decreased by PDK4 silencing. BMP9-induced osteogenic markers were reduced by PDK4 silencing, which was almost reversed by ß-catenin overexpression. PDK4 increased the BMP9-induced osteogenic markers, which was almost eliminated by ß-catenin silencing. Sclerostin was mildly decreased by BMP9 or PDK4, and significantly decreased by combined BMP9 and PDK4. In contrast, sclerostin increased significantly when BMP9 was combined with PDK4 silencing. BMP9-induced p-SMAD1/5/9 was increased by PDK4 overexpression, but was reduced by PDK4 silencing. PDK4 interacts with p-SMAD1/5/9 and regulates the sclerostin promoter. These findings suggest that PDK4 can increase the osteogenic potential of BMP9 by enhancing Wnt/ß-catenin signaling via the downregulation of sclerostin. PDK4 may be an effective target to strengthen BMP9-induced osteogenesis.


Subject(s)
Growth Differentiation Factor 2 , Mesenchymal Stem Cells , Osteogenesis , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Wnt Signaling Pathway , Animals , Mice , beta Catenin/genetics , beta Catenin/metabolism , Cell Differentiation , Growth Differentiation Factor 2/genetics , Growth Differentiation Factor 2/metabolism , Mesenchymal Stem Cells/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism
10.
Am J Cancer Res ; 11(9): 4515-4527, 2021.
Article in English | MEDLINE | ID: mdl-34659902

ABSTRACT

TNBG-5602, a new synthesized derivative of tetrazanbigen, is a potential chemotherapeutic agent against cancer. However, its underlying mechanism is complex and still unknown. In this investigation, the anticancer effects of TNBG-5602 were determined in vitro and in vivo. Small RNA retroviral library plasmids that overexpress 19-bp fragments were used to generate TNBG-5602-resistant cells. After validation, the overexpressed 19-bp fragments were sequenced using next-generation sequencing (NGS) in the drug-resistant cells. Furthermore, the relationship of TNBG-5602, phosphatase and tensin homolog deleted on Chromosome 10 (PTEN), and the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt) pathway was explored. The results showed that TNBG-5602 can effectively inhibit cancer cell proliferation and induce apoptosis in vitro and in vivo. Drug-resistant cells were screened using the small RNA library. Compared with naïve cells, drug-resistant cells were more resistant to TNBG-5602 in vitro and in vivo. NGS results revealed that the second highest overexpressed 19-bp fragment perfectly matched the PTEN gene, so the expression of PTEN in various cells and tissues was verified. Further research showed that exogenous overexpression of PTEN strengthened the anticancer effects of TNBG-5602 on p-Akt expression, whereas silencing of PTEN weakened these effects in naïve cells. Taken together, by using this library, we confirmed that PTEN is the target gene to the anticancer effects of TNBG-5602 via the PI3K/Akt pathway.

11.
Genes Dis ; 8(3): 373-383, 2021 May.
Article in English | MEDLINE | ID: mdl-33997184

ABSTRACT

Despite advances in screening and treatment, colon cancer remains one of the leading causes of cancer-related death. Finding novel and useful drug treatment targets is also an urgent need for clinical applications. Tetrandrine (Tet) is extracted from the Chinese medicinal herbal medicine, which is a well-known calcium blocker with a variety of pharmacological activities, including anti-cancer. In this study, we recruited cell viability assay, flow cytometry analysis, cloning formation to confirm that Tet can inhibit the proliferation of SW620 cells, and induce apoptosis. Mechanically, we confirmed that Tet up-regulates the mRNA and protein level of BMP9 in SW620 cells. Over-expression BMP9 enhances the anti-cancer effects of Tet in SW620 cells, but these effects can be partly reversed by silencing BMP9. Also, Tet reduces phosphorylation of Aktl/2/3 in SW620 cells, which could be elevated by overexpressed BMP9 and impaired by silencing BMP9. Furthermore, we demonstrated that Tet reduces phosphorylated PTEN, which can be promoted by overexpressed BMP9, analogously also be attenuated through silencing BMP9. Finally, we introduced a xenograft tumor model to investigate the anti-proliferative effect of Tet, further to explore the effects of BMP9 and PTEN in SW620 cells. Our findings suggested that the anti-cancer activity of Tet in SW620 cells may be mediated partly by up-regulating BMP9, followed by inactivation PI3K/Akt through up-regulating PTEN at least.

12.
Aging (Albany NY) ; 13(8): 11336-11351, 2021 04 04.
Article in English | MEDLINE | ID: mdl-33833129

ABSTRACT

This study investigated the effects of transforming growth factor-ß1 (TGF-ß1) and cyclooxygenase-2 (COX-2) on bone morphogenetic protein 9 (BMP9) in mesenchymal stem cells (MSCs). We found that BMP9 increased mRNA levels of TGF-ß1 and COX-2 in C3H10T1/2 cells. BMP9-induced osteogenic markers were enhanced by TGF-ß1 and reduced by TGF-ßRI-specific inhibitor LY364947. BMP9 increased level of p-Smad2/3, which were either enhanced or reduced by COX-2 and its inhibitor NS398. BMP9-induced osteogenic markers were decreased by NS398 and it was partially reversed by TGF-ß1. COX-2 increased BMP9-induced osteogenic marker levels, which almost abolished by LY364947. BMP9-induced bone formation was enhanced by TGF-ß1 but reduced by silencing TGF-ß1 or COX-2. BMP9's osteogenic ability was inhibited by silencing COX-2 but partially reversed by TGF-ß1. TGF-ß1 and COX-2 enhanced activation of p38 signaling, which was induced by BMP9 and reduced by LY364947. The ability of TGF-ß1 to increase the BMP9-induced osteogenic markers was reduced by p38-specific inhibitor, while BMP9-induced TGF-ß1 expression was reduced by NS398, but enhanced by COX-2. Furthermore, CREB interacted with Smad1/5/8 to regulate TGF-ß1 expression in MSCs. These findings suggest that COX-2 overexpression leads to increase BMP9's osteogenic ability, resulting from TGF-ß1 upregulation which then activates p38 signaling in MSCs.


Subject(s)
Cyclooxygenase 2/metabolism , Growth Differentiation Factor 2/metabolism , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , Transforming Growth Factor beta1/metabolism , Animals , Cell Line , Female , Gene Knockdown Techniques , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Models, Animal , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Transforming Growth Factor beta1/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Front Endocrinol (Lausanne) ; 12: 815245, 2021.
Article in English | MEDLINE | ID: mdl-35095774

ABSTRACT

Osteoporosis is a common systemic bone disease caused by the imbalance between osteogenic activity and osteoclastic activity. Aged women are at higher risk of osteoporosis, partly because of estrogen deficiency. However, the underlying mechanism of how estrogen deficiency affects osteoclast activity has not yet been well elucidated. In this study, GSE2208 and GSE56815 datasets were downloaded from GEO database with 25 PreH BMD women and 25 PostL BMD women in total. The RRA algorithm determined 38 downregulated DEGs and 30 upregulated DEGs. Through GO analysis, we found that downregulated DEGs were mainly enriched in myeloid cell differentiation, cytokine-related functions while upregulated DEGs enriched in immune-related biological processes; pathways like Notch signaling and MAPK activation were found in KEGG/Rectome pathway database; a PPI network which contains 66 nodes and 91 edges was constructed and three Modules were obtained by Mcode; Correlation analysis helped us to find highly correlated genes in each module. Moreover, three hub genes FOS, PTPN6, and CTSD were captured by Cytohubba. Finally, the hub genes were further confirmed in blood monocytes of ovariectomy (OVX) rats by real-time PCR assay. In conclusion, the integrative bioinformatics analysis and real-time PCR analysis were utilized to offer fresh light into the role of monocytes in premenopausal osteoporosis and identified FOS, PTPN6, and CTSD as potential biomarkers for postmenopausal osteoporosis.


Subject(s)
Osteoporosis, Postmenopausal , Osteoporosis , Aged , Animals , Computational Biology , Female , Gene Expression Profiling , Humans , Monocytes , Osteoporosis/genetics , Osteoporosis, Postmenopausal/genetics , Rats
14.
Aging (Albany NY) ; 12(21): 21220-21235, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159018

ABSTRACT

In this study, we studied the effect and possible mechanism of TGF-ß1 on vascular calcification. We found that the serum levels of TGF-ß1 and cycloxygenase-2 (COX-2) were significantly increased in patients with chronic kidney disease. Phosphate up regulated TGF-ß1 in vascular smooth muscle cells (VSMCs). TGF-ß1 decreased the markers of VSMCs, but increased osteogenic markers and calcification in aortic segments. The phosphate-induced osteogenic markers were reduced by the TGFßR I inhibitor (LY364947), which also attenuated the potential of phosphate to reduce VSMC markers in VSMCs. Both phosphate and TGF-ß1 increased the protein level of ß-catenin, which was partially mitigated by LY364947. TGF-ß1 decreased sclerostin, and exogenous sclerostin decreased the mineralization induced by TGF-ß1. LY364947 reduced the phosphate and TGF-ß1 induced COX-2. Meanwhile, the effects of TGF-ß1 on osteogenic markers, ß-catenin, and sclerostin, were partially reversed by the COX-2 inhibitor. Mechanistically, we found that p-Smad2/3 and p-CREB were both enriched at the promoter regions of sclerostin and ß-catenin. TGF-ß1 and COX-2 were significantly elevated in serum and aorta of rats undergoing renal failure. Therapeutic administration of meloxicam effectively ameliorated the renal lesion. Our results suggested that COX-2 may mediate the effect of TGF-ß1 on vascular calcification through down-regulating sclerostin in VMSCs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcinosis/metabolism , Cyclooxygenase 2/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Renal Insufficiency/metabolism , Renal Insufficiency/pathology , Transforming Growth Factor beta1/metabolism , Animals , Biomarkers/blood , Cells, Cultured , Cyclooxygenase 2/blood , HEK293 Cells , Humans , Male , Rats, Sprague-Dawley , Renal Insufficiency/blood , Transforming Growth Factor beta1/blood
15.
Oncol Rep ; 44(5): 2093-2107, 2020 11.
Article in English | MEDLINE | ID: mdl-32901874

ABSTRACT

Honokiol (HNK), a natural pharmaceutically active component extracted from magnolia bark, has been used for clinical treatments and has anti­inflammatory, antiviral and antioxidative effects. In recent years, anticancer research has become a major hotspot. However, the underlying molecular mechanisms of how HNK inhibits colorectal cancer have remained elusive. The present study focused on elucidating the effects of HNK on the expression of bone morphogenetic protein (BMP)7 and its downstream interaction with transforming growth factor (TGF)­ß1 and p53 in colon cancer. In in vitro assays, cell viability, cell cycle distribution and apoptosis were examined using Cell Counting Kit­8, flow cytometry and reverse transcription­quantitative PCR, respectively. In addition, the expression of BMP7, TGF­ß1 and relevant signaling proteins was determined by western blot analysis. In vivo, the anticancer effect of HNK was assessed in xenografts in nude mice. Furthermore, immunohistochemistry was performed to evaluate the association between BMP7 and TGF­ß1 expression in colon cancer. The results indicated that HNK inhibited the proliferation of colon cancer cell lines, with SW620 cells being more sensitive than other colon cancer cell lines. Furthermore, HNK markedly promoted the expression of BMP7 at the mRNA and protein level. Exogenous BMP7 potentiated the effect of HNK on SW620 cells, while knocking down BMP7 inhibited it. As a downstream mechanism, HNK increased the expression of TGF­ß1 and p53, which was enhanced by exogenous BMP7 in SW620 cells. In addition, immunohistochemical analysis indicated a positive association between BMP7 and TGF­ß1 expression. Hence, the present results suggested that HNK is a promising agent for the treatment of colon cancer and enhanced the expression TGF­ß1 and p53 through stimulating BMP7 activity via the non­canonical TGF­ß signaling pathway.


Subject(s)
Biphenyl Compounds/pharmacology , Bone Morphogenetic Protein 7/metabolism , Colonic Neoplasms/drug therapy , Lignans/pharmacology , Transforming Growth Factor beta1/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
16.
Front Pharmacol ; 11: 492, 2020.
Article in English | MEDLINE | ID: mdl-32372963

ABSTRACT

Traditional Chinese medicine is an accepted and integral part of clinical cancer management alongside Western medicine in China. However, historically TCM physicians were unaware of the chemical constituents of their formulations, and the specific biological targets in the body. Through HPLC, flow cytometry, and other processes, researchers now have a much clearer picture of how herbal medicine works in conjunction with the immune system in cancer therapy. Among them, the regulation of tumor-related T cells plays the most important role in modulating tumor immunity by traditional Chinese medicine. Encouraging results have been well-documented, including an increase in T cell production along with their associated cytokines, enhanced regulation of Tregs and important T cell ratios, the formation and function of Tregs in tumor microenvironments, and the promotion of the number and function of normal T Cells to reduce conventional cancer therapy side effects. Chinese herbal medicine represents a rich field of research from which to draw further inspiration for future studies. While promising agents have already been identified, the vast majority of Chinese herbal mechanisms remain undiscovered. In this review, we summarize the effects and mechanisms of specific Chinese herbs and herbal decoctions on tumor related T cells.

17.
Oncol Rep ; 43(2): 415-426, 2020 02.
Article in English | MEDLINE | ID: mdl-31894286

ABSTRACT

Colon cancer is one of the most common malignancies. Although there has been great development in treatment regimens over the last few decades, its prognosis remains poor. There is still a clinical need to find new drugs for colon cancer. Evodiamine (Evo) is a quinolone alkaloid extracted from the traditional herbal medicine plant Evodia rutaecarpa. In the present study, CCK­8, flow cytometry, reverse transcription quantitative polymerase chain reaction, western blot analysis and a xenograft tumor model were used to evaluate the anti­cancer activity of Evo in human colon cancer cells and determine the possible mechanism underlying this process. It was revealed that Evo exhibited prominent anti­proliferation and apoptosis­inducing effects in HCT116 cells. Bone morphogenetic protein 9 (BMP9) was notably upregulated by Evo in HCT116 cells. Exogenous BMP9 potentiated the anti­cancer activity of Evo, and BMP9 silencing reduced this effect. In addition, HIF­1α was also upregulated by Evo. The anticancer activity of Evo was enhanced by HIF­1α, but was reduced by HIF­1α silencing. BMP9 potentiated the effect of Evo on the upregulation of HIF­1α, and enhanced the antitumor effect of Evo in colon cancer, which was clearly reduced by HIF­1α silencing. In HCT116 cells, Evo increased the phosphorylation of p53, which was enhanced by BMP9 but reduced by BMP9 silencing. Furthermore, the effect of Evo on p53 was potentiated by HIF­1α and reduced by HIF­1α silencing. The present findings therefore strongly indicated that the anticancer activity of Evo may be partly mediated by BMP9 upregulation, which can activate p53 through upregulation of HIF­1α, at least in human colon cancer.


Subject(s)
Colonic Neoplasms/drug therapy , Growth Differentiation Factor 2/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Quinazolines/administration & dosage , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Mice , Phosphorylation , Quinazolines/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
18.
Front Cell Dev Biol ; 8: 608544, 2020.
Article in English | MEDLINE | ID: mdl-33614622

ABSTRACT

Bone morphogenetic protein 9 (BMP9) is one of the most efficacious osteogenic cytokines. PTEN and Wnt10b are both implicated in regulating the osteogenic potential of BMP9, but the potential relationship between them is unknown. In this study, we determined whether PTEN could reduce the expression of Wnt10b during the osteogenic process initialized by BMP9 in mesenchymal stem cells (MSCs) and the possible molecular mechanism. We find that PTEN is inhibited by BMP9 in MSCs, but Wnt10b is increased simultaneously. The BMP9-induced osteogenic markers are reduced by PTEN but increased by silencing PTEN. The effects of knockdown PTEN on elevating BMP9-induced osteogenic markers are almost abolished by knockdown of Wnt10b. On the contrary, the BMP9-increased ALP activities and mineralization are both inhibited by PTEN but almost reversed by the combination of Wnt10b. Bone masses induced by BMP9 are enhanced by knockdown of PTEN, which is reduced by knockdown of Wnt10b. The BMP9-increased Wnt10b is decreased by PTEN but enhanced by knockdown of PTEN. Meanwhile, the BMP9-induced Wnt10b is also reduced by a PI3K-specific inhibitor (Ly294002) or rapamycin, respectively. The BMP9-induced phosphorylation of CREB or Smad1/5/9 is also reduced by PTEN, but enhanced by PTEN knockdown. In addition, p-CREB interacts with p-Smad1/5/9 in MSCs, and p-CREB or p-Smad1/5/9 are both enriched at the promoter region of Wnt10b. Our findings indicate that inhibitory effects of PTEN on BMP9's osteogenic potential may be partially mediated through decreasing the expression of Wnt10b via the disturbance of interaction between CREB and BMP/Smad signaling.

19.
Biomed Pharmacother ; 118: 109279, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31376651

ABSTRACT

COX-2 specific inhibitor, which has been widely used, can delay bone fracture healing and reduce osteogenic potential of bone marrow stromal cells. However, it remains unknown how to prevent these side-effects of COX-2 inhibitor. In this study, we introduced BMP9-induced osteogenic differentiation as model to evaluate whether all-trans retinoic acid (ATRA) could ameliorate these adverse effects of COX-2 specific inhibitor on bone metabolism with in vitro and in vivo experiments, and uncover the possible mechanism underlying this process. Results showed that ATRA enhanced the potential of BMP9 to induce the osteogenic markers, such as alkaline phosphates (ALP) and mineralization; but retinoic acid receptor a (RARa) inhibitor showed the reversal effects. COX-2 specific inhibitor (NS398) reduced the osteogenic markers induced by BMP9, and ATRA almost eliminated the inhibitory effect of NS398. BMP9 up-regulated the protein level of ß-catenin and promoted it translocate to nucleus, and both were reduced by NS398. On the contrary, ATRA notablely attenuated the inhibitory effect of NS398 on BMP9-increased ß-catenin. Exogenous RXRa obviously ameliorated the inhibitory effect of silencing COX-2 on ectopic bone formation induced by BMP9. NS398 reduced the level of phosphorylated CREB, which was almost reversed by ATRA. Besides, RXRa interacted with phosphorylated CREB directly and both were recruited at ß-catenin promoter region. Thus, we demonstrated that ATRA may reverse the side-effects of COX-2 inhibitor on bone metabolism through increasing the activation of Wnt/ß-catenin pathway partly.


Subject(s)
Cell Differentiation/drug effects , Cyclooxygenase 2/metabolism , Growth Differentiation Factor 2/pharmacology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects , Animals , Biomarkers/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase Inhibitors/pharmacology , HEK293 Cells , Humans , Mesenchymal Stem Cells/drug effects , Mice
20.
J Steroid Biochem Mol Biol ; 191: 105363, 2019 07.
Article in English | MEDLINE | ID: mdl-31018166

ABSTRACT

Glucocorticoid-Induced Osteoporosis (GIOP) is a prevalent clinical complication caused by large dose administration of glucocorticoids, such as Dexamethasone (Dex) and Prednisone. GIOP may lead to fractures and even Osteonecrosis of the Femoral Head (ONFH). It has been reported that glucocorticoids inhibit osteogenesis via the suppression of osteogenic differentiation in Mesenchymal Stem Cells (MSCs), but the precise mechanism underlying this suppression awaits further investigation. Meanwhile, novel and efficacious therapies are recommended to cope with GIOP. In this study, we demonstrated that Dex had the inhibitory effect on Bone Morphogenetic Protein 9 (BMP9)-induced ALP activities and matrix mineralization in Mouse Embryonic Fibroblasts (MEFs). In addition, the study confirmed that Dex decreased the expression of osteogenic markers such as Runx2 and OPN. However, the inhibitory effect of Dex on these osteogenic markers can be reversed when combined with insulin-like growth factor 1 (IGF-1). Regarding the inhibitory mechanism, we found that the level of AKT and p-AKT can be decreased by Dex and that Ly294002, the PI3K inhibitor, can block the reversal effect of IGF-1. Moreover, the knockdown or inhibition of COX-2 produced similar results to those of Ly294002. Our findings indicated that IGF-1 may reverse the osteogenic inhibitory effect of Dex via PI3K/AKT pathway, which may be associated with the up-regulation of COX-2. This study may provide new clinical management strategy for GIOP cases.


Subject(s)
Dexamethasone/adverse effects , Fibroblasts/drug effects , Glucocorticoids/adverse effects , Growth Differentiation Factor 2/metabolism , Insulin-Like Growth Factor I/metabolism , Osteogenesis/drug effects , Animals , Cell Line , Cells, Cultured , Cyclooxygenase 2/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...