Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Toxicol Lett ; 397: 141-150, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759937

ABSTRACT

Autophagy has been implicated in the developmental toxicity of multiple organs in offspring caused by adverse environmental conditions during pregnancy. We have previously found that prenatal caffeine exposure (PCE) can cause fetal overexposure to maternal glucocorticoids, leading to chondrodysplasia. However, whether autophagy is involved and what role it plays has not been reported. In this study, a PCE rat model was established by gavage of caffeine (120 mg/kg.d) on gestational day 9-20. The results showed that reduced cartilage matrix synthesis in male fetal rats in the PCE group was accompanied by increased autophagy compared to the control group. Furthermore, the expression of mTOR, miR-421-3p, and glucocorticoid receptor (GR) in male fetal rat cartilage of PCE group was increased. At the cellular level, we confirmed that corticosterone inhibited matrix synthesis in fetal chondrocytes while increasing autophagic flux. However, administration of autophagy enhancer (rapamycin) or inhibitor (bafilomycin A1 or 3-methyladenine) partially increased or further decreased aggrecan expression respectively. At the same time, we found that corticosterone could increase the expression of miR-421-3p through GR and target to inhibit the expression of mTOR, thereby enhancing autophagy. In conclusion, PCE can cause chondrodysplasia and autophagy enhancement in male fetal rats. Intrauterine high corticosterone activates GR/miR-421-3p signaling and down-regulates mTOR signaling in fetal chondrocytes, resulting in enhanced autophagy, which can partially compensate for corticosterone-induced fetal chondrodysplasia. This study confirmed the compensatory protective effect of autophagy on the developmental toxicity of fetal cartilage induced by PCE and its epigenetic mechanism, providing novel insights for exploring the early intervention and therapeutic target of fetal-originated osteoarthritis.


Subject(s)
Autophagy , Caffeine , MicroRNAs , Rats, Sprague-Dawley , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Male , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics , Pregnancy , Autophagy/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Female , Caffeine/toxicity , Rats , Signal Transduction/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Prenatal Exposure Delayed Effects/chemically induced
2.
Free Radic Biol Med ; 208: 684-699, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37743032

ABSTRACT

Epidemiological evidence has revealed that non-alcoholic fatty liver disease (NAFLD) harbors an intrauterine origin. Autophagy is known to be involved in the protective mechanism in the development of adult NAFLD, but whether it engages in the occurrence of fetal-originated NAFLD remains unclear. In this study, a rat model of fetal-originated NAFLD was established by giving a high-fat diet or chronic stress after birth on prenatal caffeine exposure (PCE) male offspring. The alterations of liver morphologic analysis, lipid metabolism, and autophagy before and after birth were determined to confirm autophagy mechanism, NAFLD susceptibility, and intrauterine origin in PCE male adult offspring. Our results revealed that PCE-induced intrauterine high concentration of corticosterone exposure blocked autophagic flux by inhibiting cathepsin D expression in hepatocytes, leading to ß-oxidation inhibition and lipid accumulation in the liver. Moreover, high concentration of corticosterone upregulated miR-665 by activating the glucocorticoid receptor to suppress cathepsin D, thus causing lysosomal degradation dysfunction and autophagy flux blockade. Notably, hepatic overexpression of cathepsin D could reverse PCE-induced postnatal NAFLD susceptibility in male rat offspring. This study elucidates the epigenetic programming mechanism of intrauterine autophagy-related fetal-originated NAFLD susceptibility, and identifies cathepsin D as its early intervention target, providing an experimental basis for exploring early prevention and treatment strategies for fetal-originated NAFLD.


Subject(s)
MicroRNAs , Non-alcoholic Fatty Liver Disease , Prenatal Exposure Delayed Effects , Pregnancy , Female , Humans , Rats , Male , Animals , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Caffeine/adverse effects , Corticosterone , Cathepsin D/genetics , Rats, Wistar , Prenatal Exposure Delayed Effects/metabolism , Autophagy
3.
FASEB J ; 37(7): e23011, 2023 07.
Article in English | MEDLINE | ID: mdl-37249374

ABSTRACT

Autophagy inhibition is known to be involved in the development of adult osteoarthritis. Dexamethasone, as a synthetic glucocorticoid, is widely used for premature delivery and related pregnancy diseases in clinics. We have previously shown that prenatal dexamethasone exposure (PDE) was associated with increased susceptibility to postnatal osteoarthritis in offspring. However, whether the occurrence of fetal-originated adult osteoarthritis induced by PDE is related to autophagy remains unclear. In this study, we first found that PDE could increase the mRNA and protein expression of cartilage matrix-degrading enzymes (MMP3, MMP13, and ADAMTS5) and decrease the cartilage matrix contents in adult offspring, and the in vitro results suggested that this might be related to the autophagy inhibition of chondrocytes. Further, we demonstrated a persistent autophagy inhibition with autolysosome accumulation, low expression of cathepsin D (CTSD), increased H3K9ac level, and expression of miR-1912-3p in the cartilage of PDE offspring from fetus to adulthood. In vitro experiments showed that dexamethasone inhibited autophagy flux and CTSD expression in fetal chondrocytes, while overexpression of CTSD could alleviate the inhibition of autophagic flux induced by dexamethasone. Finally, we confirmed that dexamethasone increased the H3K9ac level and expression of miR-1912-3p through activation of the glucocorticoid receptor (GR), resulting in the decreased expression of CTSD and inhibition of autophagy flux in fetal chondrocytes. In conclusion, intrauterine miR-1912-3p/CTSD programming-mediated autophagy inhibition promoted the susceptibility to osteoarthritis in PDE adult offspring rats. This study provides new ideas for exploring early prevention and therapeutic targets in fetal-originated osteoarthritis.


Subject(s)
MicroRNAs , Osteoarthritis , Prenatal Exposure Delayed Effects , Pregnancy , Humans , Female , Rats , Male , Animals , Rats, Wistar , Cathepsin D , Prenatal Exposure Delayed Effects/chemically induced , Osteoarthritis/chemically induced , Osteoarthritis/genetics , Osteoarthritis/metabolism , Dexamethasone/toxicity , MicroRNAs/genetics , Autophagy
5.
Chem Biol Interact ; 368: 110201, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36174738

ABSTRACT

PURPOSE: This study aims to further explore cartilage development in prenatal ethanol exposure (PEE) offspring at different times to explore the specific time points and mechanism of ethanol-induced fetal cartilage dysplasia. METHODS: On gestational day (GD)14, GD17, and GD20, PEE fetal cartilage was evaluated by morphological analysis. RT-qPCR, immunohistochemistry, and immunofluorescence were used to detect the expression of cartilage marker genes and their regulatory factors. Bone marrow mesenchymal stem cells (BMSCs) were used to explore the effect of ethanol on the differentiation of chondrocytes. Additionally, we used inhibitors, overexpression plasmids and a luciferase reporter assay on GD17 chondrocytes to verify the mechanism. RESULTS: PEE significantly reduced cartilage matrix content and the expression of marker genes on GD17 and GD20 but had no effect on GD14. The inhibition of chondrogenic differentiation by PEE mainly occurred on GD14-17. Furthermore, the expression of miR-200b-3p was increased, while that of ERG and PTHrP was markedly reduced in PEE fetal cartilage. In vitro, ethanol (30-120 mM) inhibited the differentiation of BMSCs into chondrocytes in a concentration-dependent manner, accompanied by strong expression of miR-200b-3p and low expression of ERG and PTHrP. Moreover, PTHLH and ERG overexpressed, as well as a miR-200b-3p inhibitor reversed the inhibitory effect of ethanol on the differentiation of fetal chondrocytes. Furthermore, miR-200b-3p could target and negatively regulate ERG. CONCLUSIONS: PEE can significantly inhibit the development of articular cartilage, especially during articular cartilage formation. The mechanism is related to the decreased differentiation of fetal cartilage into articular cartilage mediated by the miR-200b-3p/ERG/PTHrP axis.


Subject(s)
Cartilage, Articular , MicroRNAs , Female , Pregnancy , Cartilage, Articular/metabolism , Chondrocytes , Ethanol/pharmacology , Ethanol/metabolism , MicroRNAs/metabolism , Parathyroid Hormone-Related Protein/genetics , Parathyroid Hormone-Related Protein/metabolism , Parathyroid Hormone-Related Protein/pharmacology , Transcriptional Regulator ERG/metabolism
6.
Ecotoxicol Environ Saf ; 239: 113624, 2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35588619

ABSTRACT

Our previous study reported that prenatal caffeine exposure (PCE) could induce chondrodysplasia and increase the susceptibility to osteoarthritis in offspring rats. However, the potential mechanisms and initiating factors remain unknown. This study aims to investigate whether 11ß-HSD2, a glucocorticoid-metabolizing enzyme, is involved in the susceptibility of osteoarthritis induced by PCE and to further explore its potential mechanisms and initiating factors. Firstly, we found that PCE reduced cartilage matrix synthesis (aggrecan/Col2a1 expression) in male adult offspring rats and exhibited an osteoarthritis phenotype following chronic stress, which was associated with persistently reduced H3K9ac and H3K27ac levels at the promoter of 11ß-HSD2 as well as its expression in the cartilage from fetus to adulthood. The expression of 11ß-HSD2, aggrecan and Col2a1 were all decreased by corticosterone in the fetal chondrocytes, while overexpression of 11ß-HSD2 could partially alleviate the decrease of matrix synthesis induced by corticosterone in vitro. Furthermore, the glucocorticoid receptor (GR) activated by glucocorticoids directly bonded to the promoter region of 11ß-HSD2 to inhibit its expression. Meanwhile, the activated GR reduced the H3K9ac and H3K27ac levels of 11ß-HSD2 by recruiting HDAC4 and promoting GR-HDAC4 protein interaction to inhibit the 11ß-HSD2 expression. Moreover, caffeine could reduce the expression of 11ß-HSD2 by inhibiting the cAMP/PKA signaling pathway but without reducing the H3K9ac and H3K27ac levels of 11ß-HSD2, thereby synergistically enhancing the corticosterone effect. In conclusion, the persistently reduced H3K9ac and H3K27ac levels of 11ß-HSD2 from fetus to adulthood mediated the inhibition of cartilage matrix synthesis and the increased susceptibility to osteoarthritis. This epigenetic programming change in utero was induced by glucocorticoids with synergistic effect of caffeine.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 2 , Osteoarthritis , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , Aggrecans , Animals , Caffeine/toxicity , Cartilage , Corticosterone , Female , Glucocorticoids/metabolism , Male , Osteoarthritis/chemically induced , Osteoarthritis/genetics , Pregnancy , Rats
7.
Eur J Pharmacol ; 924: 174971, 2022 Jun 05.
Article in English | MEDLINE | ID: mdl-35469836

ABSTRACT

Osteoarthritis (OA), a progressive and degenerative joint disease, is characterized by cartilage degradation, synovitis, subchondral bone remodeling and osteophyte formation. Isorhynchophylline (IRN) is an oxindole alkaloid isolated from the traditional Chinese herb Uncaria rhynchophylla. In this study, we evaluated the protective effects of IRN on human OA chondrocytes. IRN treatment dose-dependently decreased the interleukin-1ß (IL-1ß)-induced expressions of nitric oxide (NO; p < 0.001), prostaglandin E2 (PGE2; p < 0.001), tumor necrosis factor alpha (TNF-α; p < 0.001), interleukin-6 (IL-6; p < 0.001), cyclooxygenase-2 (COX-2; p < 0.001) and inducible nitric oxide synthase (iNOS; p < 0.001) in chondrocytes. Meanwhile, the production of metalloproteinase 13 (MMP13; p < 0.001) and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5; p < 0.001) was inhibited by IRN treatment. Molecular docking studies revealed that IRN directly interacted with the nuclear factor kappa B (NF-κB) complex, which was associated with a reduced level of NF-κB nuclear translocation and the inhibition of NF-κB signaling activity. Furthermore, administration of IRN generated marked in vivo protective effects during OA development. Collectively, our results demonstrate that IRN may exhibit therapeutic benefits against OA, potentially by ameliorating the inflammative and degenerative progression of OA via inhibiting the NF-κB pathway.


Subject(s)
NF-kappa B , Osteoarthritis , Chondrocytes , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Molecular Docking Simulation , NF-kappa B/metabolism , Nitric Oxide/metabolism , Osteoarthritis/pathology , Oxindoles/metabolism , Oxindoles/pharmacology , Oxindoles/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Uncaria
8.
Arthroscopy ; 38(7): 2246-2254.e1, 2022 07.
Article in English | MEDLINE | ID: mdl-35093495

ABSTRACT

PURPOSE: This study aimed to verify whether transplantation of dedifferentiated osteogenic bone marrow mesenchymal stem cells (De-BMSCs) at the tendon-bone interface could result in more bone formation than BMSC transplantation in anterior cruciate ligament (ACL) reconstruction. METHODS: BMSCs from femur and tibia of New Zealand White rabbit were subjected to osteogenic induction and then cultured in osteogenic factor-free medium; the obtained cell population was termed De-BMSCs. Bilateral ACL reconstruction was performed in 48 adult rabbits. Three groups were established: control group with alginate gel injection, BMSCs group with the BMSCs injection, and De-BMSCs group with the De-BMSCs injection. At week 4 and 12 postoperatively, tendon-bone healing by histologic staining, micro-computed tomography examination, and biomechanical test were evaluated. RESULTS: The expression of α1 chain of type I collagen, osteocalcin, and osteopontin at the tendon-bone interface in the De-BMSCs group was greater than in the control or BMSCs group. The bone volume/total volume by micro-computed tomography scan was significantly greater in the De-BMSCs group than that in the control group (P = .013) or BMSCs group (P = .045) at 4 weeks, and greater than that in the control group (P = .014) or BMSCs group (P = .017) at 12 weeks. The tunnel area was significantly smaller in the De-BMSCs group than in the control group (P = .013) or BMSCs group (P = .044) at 12 weeks. The failure load and stiffness in De-BMSCs group were both significantly enhanced at 4 and 12 weeks than control group or De-BMSCs group. CONCLUSIONS: De-BMSCs transplantation can promote bone formation at the tendon-bone interface better than BMSCs transplantation in ACL reconstruction and increase the early biomechanical strength of the reconstructed ACL CLINICAL RELEVANCE: De-BMSCs transplantation is a potential choice for enhancing early bone formation in the tunnel in ACL reconstruction.


Subject(s)
Anterior Cruciate Ligament Reconstruction , Mesenchymal Stem Cells , Animals , Anterior Cruciate Ligament Reconstruction/methods , Bone Marrow Cells , Osteogenesis , Rabbits , Tendons/metabolism , X-Ray Microtomography
9.
BMC Med Genomics ; 14(1): 237, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34587952

ABSTRACT

BACKGROUND: The componential and structural change in the meniscus with aging would increase the tissue vulnerability of the meniscus, which would induce meniscus tearing. Here, we investigated the molecular mechanism of age-related meniscus degeneration with gene expression profiling analysis, and validate pivotal genes in vivo and in vitro models. METHODS: The GSE45233 dataset, including 6 elderly meniscus samples and 6 younger meniscus samples, was downloaded from the Gene Expression Omnibus (GEO) database. To screen the differential expression of mRNAs and identify the miRNAs targeting hub genes, we completed a series of bioinformatics analyses, including functional and pathway enrichment, protein-protein interaction network, hub genes screening, and construction of a lncRNA-miRNA-mRNA network. Furthermore, crucial genes were examined in human senescent menisci, mouse senescent meniscus tissues and mouse meniscus cells stimulated by IL-1ß. RESULTS: In total, the most significant 4 hub genes (RRM2, AURKB, CDK1, and TIMP1) and 5 miRNAs (hsa-miR-6810-5p, hsa-miR-4676-5p, hsa-miR-6877-5p, hsa-miR-8085, and hsa-miR-6133) that regulated such 4 hub genes, were finally identified. Moreover, these hub genes were decreased in meniscus cells in vitro and meniscus tissues in vivo, which indicated that hub genes were related to meniscus senescence and could serve as potential biomarkers for age-related meniscus tearing. CONCLUSIONS: In short, the integrated analysis of gene expression profile, co-expression network, and models detection identified pivotal genes, which elucidated the possible molecular basis underlying the senescence meniscus and also provided prognosis clues for early-onset age-related meniscus tearing.


Subject(s)
Aging/genetics , Gene Expression Profiling , Meniscus/metabolism , Animals , Gene Ontology , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Protein Interaction Maps , RNA, Long Noncoding/genetics , RNA, Messenger/genetics
10.
Life Sci ; 269: 119096, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33482192

ABSTRACT

AIMS: This study intends to explore the role of Vaspin and cholesterol metabolism in the process of osteoarthritis (OA) and its mechanism in vitro and in vivo. MAIN METHODS: In vitro, chondrocytes were treated with interleukin-1ß (IL-1ß, 20 ng/mL) in combination with Vaspin at different concentrations for 48 h. The expressions of Aggrecan (ACAN), Collagen 2a1 (Col2a1), A Disintegrin And Metalloproteinase with Thrombo Spondin type 1 motifs 5 (ADAMTS 5), and Matrix metalloproteinase 13 (MMP13) were detected. In vivo, the expression of liver X receptor (LXRα) and other Cholesterol efflux related genes were detected in the rat OA knee cartilage-induced by papain. KEY FINDINGS: In vitro, in a concentration-dependent manner, Vaspin reversed the decreased expression of ACAN and Col2a1, and the increased expression of ADAMTS 5 and MMP13 caused by IL-1ß. Besides, Vaspin promoted the expression of LXRα and other Cholesterol efflux related genes in a concentration-dependent manner in chondrocytes. However, miR155 mimics reversed the Vaspin-induced expression changes of cholesterol efflux pathway in chondrocytes. In vivo, the expression of LXRα and other Cholesterol efflux related genes were decreased in the rat OA knee cartilage-induced by papain. Besides, the level of Vaspin was reduced and the miroRNA155 (miR155) expression was increased in OA knee cartilage of rats. SIGNIFICANCE: In conclusion, the decreased expression of Vaspin inhibited the expression of Cholesterol efflux pathway via miR155/LXRα. Finally, the inhibited Cholesterol efflux pathway led to the cholesterol accumulation and OA in cartilage.


Subject(s)
Arthritis, Experimental/pathology , Cartilage, Articular/pathology , Cholesterol/metabolism , Liver X Receptors/metabolism , MicroRNAs/genetics , Osteoarthritis/pathology , Serpins/metabolism , Animals , Arthritis, Experimental/etiology , Arthritis, Experimental/metabolism , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes , Female , Gene Expression Regulation , Liver X Receptors/genetics , Osteoarthritis/etiology , Osteoarthritis/metabolism , Rats , Rats, Wistar , Serpins/genetics , Signal Transduction
11.
FASEB J ; 34(9): 12834-12846, 2020 09.
Article in English | MEDLINE | ID: mdl-32767431

ABSTRACT

Maternal dexamethasone decreases the body length of the newborn. However, whether dexamethasone inhibits the development of the growth plate of the fetal long bone is still unknown. Here, we found that lengths of fetal femur and growth plate were both shorter in the fetuses with maternal dexamethasone (0.2 mg/kg.d from gestation day 9 to 20), with a decreased proteoglycan content of the growth plate in the fetal rat. Notable decreases in both the gene expression and H3K9 acetylation of UDP-glucose dehydrogenase (Ugdh) gene, which codes a key enzyme in the proteoglycan biosynthesis in the chondrocyte, were also observed. Meanwhile, up-regulation of glucocorticoid receptor (GR), specific protein 3 (Sp3), and histone deacetylase 1 (Hdac1) gene expression were detected in the fetal growth plate. Similar changes were also observed in the chondrogenic rat bone marrow stromal cells (BMSCs) with excessive exogenous dexamethasone. However, antagonizing GR with RU486 and silencing Hdac1 or Sp3 with specific siRNAs could all stimulate the H3K9 acetylation and gene expression of Ugdh previously inhibited by dexamethasone. Meanwhile, dexamethasone also induced the nuclear translocation of GR, which further directly bound to the Ugdh promoter and interacted with HDAC1 and Sp3, respectively. Collectively, our study revealed that maternal dexamethasone induced the direct binding of GR to the Ugdh promoter of the chondrocyte in the rat fetal growth plate, which recruited HDAC1 and Sp3, induced deacetylation of the H3K9, and subsequently inhibited Ugdh gene expression. Such changes further led to attenuated proteoglycan synthesis in the developing chondrocyte and therefore disrupted the development of growth plate and fetal long bone.


Subject(s)
Dexamethasone/adverse effects , Femur , Fetal Development/drug effects , Growth Plate , Maternal Exposure/adverse effects , Mesenchymal Stem Cells/drug effects , Animals , Cells, Cultured , Female , Femur/drug effects , Femur/embryology , Femur/pathology , Gene Expression Regulation, Developmental/drug effects , Growth Plate/drug effects , Growth Plate/embryology , Growth Plate/pathology , Histone Deacetylase 1/metabolism , Male , Maternal-Fetal Exchange , Mesenchymal Stem Cells/cytology , Pregnancy , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Signal Transduction , Sp3 Transcription Factor/metabolism , Uridine Diphosphate Glucose Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL