Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 328
Filter
1.
Int J Mol Sci ; 25(13)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38999967

ABSTRACT

Age-related macular degeneration (AMD) is a progressive neurodegenerative condition leading to vision loss and eventual blindness, with exudative AMD posing a heightened risk due to choroidal neovascularization and localized edema. Therapies targeting the VEGF pathway aim to address this mechanism for treatment effectiveness. Our study aimed to evaluate associations between specific genetic variants (RAD51B rs8017304, rs2588809; TRIB1 rs6987702, rs4351379; COL8A1 rs13095226; COL10A1 rs1064583; IL-9 rs1859430, rs2069870, rs11741137, rs2069885, rs2069884; IL-10 rs1800871, rs1800872, rs1800896; VEGFA rs1570360, rs699947, rs3025033, rs2146323) and the response to anti-VEGF treatment for exudative AMD. We enrolled 119 patients with exudative AMD categorized as responders or non-responders based on their response to anti-VEGF treatment. Statistical analysis revealed that RAD51B rs8017304 heterozygous and homozygous minor allele carriers had increased CMT before treatment compared to wild-type genotype carriers (p = 0.004). Additionally, TRIB1 rs4351379 heterozygous and homozygous minor allele carriers exhibited a greater decrease in central macular thickness (CMT) after 6 months of treatment than wild-type genotype carriers (p = 0.030). IL-9 rs1859430, rs2069870, and rs2069884 heterozygous and homozygous minor allele carriers had worse BCVA before treatment than wild-type genotype carriers (p = 0.018, p = 0.012, p = 0.041, respectively). Conversely, IL-9 rs2069885 heterozygous and homozygous minor allele carriers showed greater improvement in BCVA after 6 months compared to wild-type genotype carriers (p = 0.032). Furthermore, VEGFA rs699947 heterozygous and homozygous minor allele carriers had better BCVA before treatment and after 3 and 6 months of treatment than wild-type genotype carriers (p = 0.003, p = 0.022, respectively), with these carriers also exhibiting higher CMT after 6 months of anti-VEGF treatment (p = 0.032). Not all results remained statistically significant under this stringent correction for multiple comparisons. The comparisons of the serum concentrations of IL-10, VEGF-A, and VEGF-R2/KDR between non-responders and responders did not yield statistically significant differences. Our study identified significant associations between genetic variants, including RAD51B rs8017304, TRIB1 rs4351379, IL-9 rs1859430, rs2069870, rs2069884, rs2069885, and VEGFA rs699947, and parameters related to the efficacy of exudative AMD treatment, such as BCVA and CMT.


Subject(s)
Collagen Type X , Interleukin-10 , Interleukin-9 , Intracellular Signaling Peptides and Proteins , Polymorphism, Single Nucleotide , Protein Serine-Threonine Kinases , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/genetics , Male , Female , Aged , Interleukin-10/genetics , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Interleukin-9/genetics , Collagen Type X/genetics , Treatment Outcome , Macular Degeneration/genetics , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Aged, 80 and over , DNA-Binding Proteins/genetics , Middle Aged , Genotype , Collagen Type VIII
2.
Int J Biol Macromol ; 273(Pt 1): 133074, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38866293

ABSTRACT

Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer, has a poor prognosis and limited access to efficient targeted treatments. Chronic unpredictable mild stress (CUMS) is highly risk factor for TNBC occurrence and development. Type X collagen (COL10A1), a crucial protein component of the extracellular matrix, ranks second among all aberrantly expressed genes in TNBC, and it is significantly up-regulated under CUMS. Nevertheless, the impact of CUMS and COL10A1 on TNBC, along with the underlying mechanisms are still unclear. In this research, we studied the effect of CUMS-induced norepinephrine (NE) elevation on TNBC, and uncovered that it notably enhanced TNBC cell proliferation, migration, and invasion in vitro, and also fostering tumor growth and lung metastasis in vivo. Additionally, our investigation found that COL10A1 directly interacted with integrin subunit beta 1 (ITGB1), then activates the downstream PI3K/AKT signaling pathway, thereby promoting TNBC growth and metastasis, while it was reversed by knocking down of COL10A1 or ITGB1. Our study demonstrated that the TNBC could respond to CUMS, and advocate for COL10A1 as a pivotal therapeutic target in TNBC treatment.


Subject(s)
Cell Proliferation , Collagen Type X , Integrin beta1 , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Integrin beta1/metabolism , Integrin beta1/genetics , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Female , Animals , Cell Line, Tumor , Signal Transduction/drug effects , Cell Proliferation/drug effects , Collagen Type X/metabolism , Collagen Type X/genetics , Disease Progression , Mice , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques
3.
Mol Carcinog ; 63(8): 1588-1598, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38780151

ABSTRACT

Triple-negative breast cancer (TNBC) is a malignant tumor with high degree of malignancy and lack of effective target treatment. The research aims to explore the role and mechanism of X collagen alpha-1 chain protein (COL10A1 gene) in TNBC. UALCAN and Kaplan-Meier were used to detect the expression of COL10A1 and its role in the prognosis of breast cancer patients. The cells with stably expressing high levels of COL10A1 were obtained by recombinant lentivirus infection. The expression of COL10A1 in cells was temporarily downregulated by siRNA interference fragments. Real-time quantitative polymerase chain reaction and western blot analysis were utilized to detect the changes of COL10A1 mRNA and protein expression. The biological functions of the cells were evaluated by colony formation, cell counting kit-8, cell invasion and wound healing experiments. In addition, the effect of COL10A1 on angiogenesis was investigated by tube formation assay. Xenograft tumor model was used to confirm the effect of COL10A1 on tumorigenicity in vivo and multiplex fluorescent immunohistochemistry to detect multiple proteins simultaneously. The possible molecular mechanism of the function of COL10A1 was speculated through the detection of proteins in functionally related pathways. COL10A1 is highly expressed and is significantly associated with worse overall survival (OS) and recurrence-free survival (RFS) in TNBC. Overexpression of COL10A1 increased the clone formation rate and cell migration capacity of TNBC cells. In the COL10A1 overexpression group, the clone formation rates of MD-MB-231 and BT-549 cells (21.5 ± 0.62, 27.83 ± 3.72)% were significantly higher than those in the control group(15.23 ± 2.79, 19.4 ± 1.47)%, and the relative migration ratio (47.40 ± 3.09, 41.26 ± 4.33)% were higher than those in the control group (34.48 ± 2.03, 21.80 ± 1.03)%. When the expression of COL10A1 was downregulated, the ability of clone formation and wound-healing migration capacity in TNBC cells was weakened. Upregulated COL10A1 in TNBC cells generated more junctions and longer total segments between vascular endothelial cells, and promoted angiogenesis of the cells, and thus enhanced the tumorigenesis. In TNBC, it was found that COL10A1 might affect epithelial-mesenchymal transition (EMT) of the cells through Wnt/ß-catenin signaling pathway by the detection of the related pathway proteins. COL10A1 is highly expressed in TNBC, and its high expression leads to poor OS and RFS. COL10A1 may enhance TNBC cell proliferation, migration and tumor-related angiogenesis, and promote tumorigenesis in vivo via Wnt/ß-catenin signaling.


Subject(s)
Cell Movement , Cell Proliferation , Collagen Type X , Gene Expression Regulation, Neoplastic , Triple Negative Breast Neoplasms , Wnt Signaling Pathway , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/metabolism , Humans , Female , Wnt Signaling Pathway/genetics , Animals , Mice , Cell Movement/genetics , Cell Line, Tumor , Collagen Type X/genetics , Collagen Type X/metabolism , Prognosis , Up-Regulation , Mice, Nude , beta Catenin/metabolism , beta Catenin/genetics , Xenograft Model Antitumor Assays , Middle Aged , Mice, Inbred BALB C
4.
In Vitro Cell Dev Biol Anim ; 60(6): 609-615, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38727898

ABSTRACT

Osteoblast-derived semaphorin3A (Sema3A) has been reported to be involved in bone protection, and Sema3A knockout mice have been reported to exhibit chondrodysplasia. From these reports, Sema3A is considered to be involved in chondrogenic differentiation and skeletal formation, but there are many unclear points about its function and mechanism in chondrogenic differentiation. This study investigated the pharmacological effects of Sema3A in chondrogenic differentiation. The amount of Sema3A secreted into the culture supernatant was measured using an enzyme-linked immunosorbent assay. The expression of chondrogenic differentiation-related factors, such as Type II collagen (COL2A1), Aggrecan (ACAN), hyaluronan synthase 2 (HAS2), SRY-box transcription factor 9 (Sox9), Runt-related transcription factor 2 (Runx2), and Type X collagen (COL10A1) in ATDC5 cells treated with Sema3A (1,10 and 100 ng/mL) was examined using real-time reverse transcription polymerase chain reaction. Further, to assess the deposition of total glycosaminoglycans during chondrogenic differentiation, ATDC5 cells were stained with Alcian Blue. Moreover, the amount of hyaluronan in the culture supernatant was measured by enzyme-linked immunosorbent assay. The addition of Sema3A to cultured ATDC5 cells increased the expression of Sox9, Runx2, COL2A1, ACAN, HAS2, and COL10A1 during chondrogenic differentiation. Moreover, it enhanced total proteoglycan and hyaluronan synthesis. Further, Sema3A was upregulated in the early stages of chondrogenic differentiation, and its secretion decreased later. Sema3A increases extracellular matrix production and promotes chondrogenic differentiation. To the best of our knowledge, this is the first study to demonstrate the role of Sema3A on chondrogenic differentiation.


Subject(s)
Cell Differentiation , Chondrogenesis , Semaphorin-3A , Animals , Cell Differentiation/drug effects , Semaphorin-3A/metabolism , Chondrogenesis/drug effects , Mice , Chondrocytes/metabolism , Chondrocytes/cytology , Hyaluronic Acid/metabolism , Hyaluronic Acid/pharmacology , Cell Line , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Collagen Type II/metabolism , Collagen Type II/genetics , Aggrecans/metabolism , Aggrecans/genetics , Hyaluronan Synthases/metabolism , Hyaluronan Synthases/genetics , Glycosaminoglycans/metabolism , Collagen Type X/metabolism , Collagen Type X/genetics
5.
Biomolecules ; 14(2)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38397376

ABSTRACT

Zebrafish are now widely used to study skeletal development and bone-related diseases. To that end, understanding osteoblast differentiation and function, the expression of essential transcription factors, signaling molecules, and extracellular matrix proteins is crucial. We isolated Sp7-expressing osteoblasts from 4-day-old larvae using a fluorescent reporter. We identified two distinct subpopulations and characterized their specific transcriptome as well as their structural, regulatory, and signaling profile. Based on their differential expression in these subpopulations, we generated mutants for the extracellular matrix protein genes col10a1a and fbln1 to study their functions. The col10a1a-/- mutant larvae display reduced chondrocranium size and decreased bone mineralization, while in adults a reduced vertebral thickness and tissue mineral density, and fusion of the caudal fin vertebrae were observed. In contrast, fbln1-/- mutants showed an increased mineralization of cranial elements and a reduced ceratohyal angle in larvae, while in adults a significantly increased vertebral centra thickness, length, volume, surface area, and tissue mineral density was observed. In addition, absence of the opercle specifically on the right side was observed. Transcriptomic analysis reveals up-regulation of genes involved in collagen biosynthesis and down-regulation of Fgf8 signaling in fbln1-/- mutants. Taken together, our results highlight the importance of bone extracellular matrix protein genes col10a1a and fbln1 in skeletal development and homeostasis.


Subject(s)
Collagen Type X , Extracellular Matrix Proteins , Osteoblasts , Zebrafish , Animals , Cell Differentiation , Extracellular Matrix/genetics , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Homeostasis/genetics , Minerals/metabolism , Osteoblasts/metabolism , Transcriptome/genetics , Zebrafish/genetics , Zebrafish/growth & development , Collagen Type X/genetics , Collagen Type X/physiology
6.
J Nanobiotechnology ; 22(1): 72, 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38374072

ABSTRACT

Osteoarthritis (OA) is one of the most prevalent chronic musculoskeletal diseases among the elderly population. In this study, macrophage-derived exosomes were isolated and identified. Exosomes were subjected to microRNA (miRNA) sequencing and bioinformatic analysis, and differentially expressed miRNAs were verified. miR-26b-5p target genes were confirmed through target-site mutation combined with a dual-luciferase reporter assay. The effects of miR-26b-5p on macrophage polarization and chondrocyte hypertrophy were assessed in vitro. miR-26b-5p agomir was applied to mice with OA induced by anterior cruciate ligament transection (ACLT). The therapeutic effects of miR-26b-5p were evaluated via pain behavior experiments and histological observations. In vitro, miR-26b-5p repolarized M1 macrophages to an anti-inflammatory M2 type by targeting the TLR3 signaling pathway. miR-26b-5p could target COL10A1, further inhibiting chondrocyte hypertrophy induced by M1 macrophage-conditioned medium (M1-CM). In vivo, miR-26b-5p agomir ameliorated gait abnormalities and mechanical allodynia in OA mice. miR-26b-5p treatment attenuated synovitis and cartilage degeneration, thereby delaying OA progression. In conclusion, M2 macrophage-derived exosomal miR-26b-5p could protect articular cartilage and ameliorate gait abnormalities in OA mice by targeting TLR3 and COL10A1. miR-26b-5p further affected macrophage polarization and chondrocyte hypertrophy. Thus, this exosomal miR-26b-5p-based strategy might be a potential method for OA treatment.


Subject(s)
MicroRNAs , Osteoarthritis , Aged , Animals , Humans , Mice , Chondrocytes/metabolism , Hypertrophy/metabolism , Hypertrophy/pathology , Macrophages/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoarthritis/metabolism , Toll-Like Receptor 3/metabolism , Collagen Type X/genetics , Collagen Type X/metabolism , Exosomes/genetics
7.
Aging (Albany NY) ; 15(24): 15134-15160, 2023 12 21.
Article in English | MEDLINE | ID: mdl-38147021

ABSTRACT

BACKGROUND: Type X collagen (COL10) is a homologous trimeric non-fibrillar collagen found in the extracellular matrix of human tissues, and it exhibits a distinctive white appearance. Type X collagen α1 chain (COL10A1) is a specific cleaved fragment of type X collagen. However, the expression, prognostic significance, clinicopathological attributes and immune-related associations of COL10A1 in prostate cancer as well as in pan-cancer contexts remain poorly understood. METHODS: Using bioinformatic analysis of data from the most recent databases (TCGA, GTEx and GEO databases), we have extensively elucidated the role played by COL10A1 in terms of its expression patterns, prognostic implications, and immune efficacy across a pan-cancer spectrum. Subsequently, the biological functions of COL10A1 in prostate cancer were elucidated by experimental validation. RESULTS: Our findings have confirmed that COL10A1 was highly expressed in most cancers and was associated with poorer prognosis in cancer patients. Immune correlation analysis of COL10A1 in various cancers showed its significant correlation with Tumor mutational burden (TMB), microsatellite instability (MSI) and immune cell infiltration. In addition, knockdown of COL10A1 in prostate cancer resulted in a substantial reduction in the proliferation, migration, and invasive potential of prostate cancer cells. CONCLUSION: Our pan-cancer analysis of COL10A1 gene provided novel insights into its pivotal role in cancer initiation, progression, and therapeutic implications, underscoring its potential significance in prognosis and immunotherapeutic interventions for cancer, particularly prostate cancer.


Subject(s)
Collagen Type X , Prostatic Neoplasms , Humans , Male , Collagen Type X/genetics , Oncogenes/genetics , Prognosis , Prostate , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy
8.
Microvasc Res ; 139: 104239, 2022 01.
Article in English | MEDLINE | ID: mdl-34520774

ABSTRACT

With the dramatic rise in the aging population, researching age-related macular degeneration (AMD), especially the severe form neovascular AMD (nAMD), has become more important than ever. In this study, we found that collagen type X was increased in retina-choroid tissue of mice with laser-induced choroidal neovascularization (CNV) based on immunohistofluorescence. RNA sequencing and bioinformatic analyses were performed to compare the retina-choroid tissue complex of the CNV mouse model to normal controls. Collagen type X alpha 1 chain (Col10a1) was among the most significantly upregulated genes, and the results were validated with an animal model at the mRNA and protein levels by quantitative real-time polymerase chain reaction (qPCR) and western blotting, respectively. COL10A1 was also upregulated in human retinal microvascular endothelial cells (HRMECs), human umbilical vein endothelial cells (HUVECs), RPE19 cells and RF/6A cells under hypoxic conditions. Next, in vitro and in vivo experiments were performed to study the effect of COL10A1 on neovascularization. siRNA knockdown of COL10A1 suppressed the proliferation and tube formation ability of HRMECs under hypoxic conditions. Snail family transcriptional repressor 1 (SNAIL1) and angiopoietin-2 (ANGPT2) were downregulated in COL10A1 knockdown HRMECs under hypoxic conditions and thus were potential downstream genes. Significant decreases in CNV leakage and CNV lesion area, as assessed by fundus fluorescein angiography (FFA) and immunofluorescence of choroidal flat mounts, respectively, were observed in a mouse model intravitreally injected with anti-collagen X monoclonal antibody (mAb) compared to the controls. In conclusion, COL10A1 promotes CNV formation and may represent a new candidate target for the treatment and diagnosis of nAMD and other neovascular diseases.


Subject(s)
Choroid/blood supply , Choroidal Neovascularization/metabolism , Collagen Type X/metabolism , Endothelial Cells/metabolism , Macular Degeneration/metabolism , Neovascularization, Physiologic , Angiopoietin-2/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Cell Hypoxia , Cell Line , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Choroidal Neovascularization/prevention & control , Collagen Type X/antagonists & inhibitors , Collagen Type X/genetics , Collagen Type X/immunology , Disease Models, Animal , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Macular Degeneration/prevention & control , Male , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Signal Transduction , Snail Family Transcription Factors/metabolism
9.
Exp Cell Res ; 408(2): 112841, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34563516

ABSTRACT

Osteoarthritis (OA) patients undergo cartilage degradation and experience painful joint swelling. OA symptoms are caused by inflammatory molecules and the upregulation of catabolic genes leading to the breakdown of cartilage extracellular matrix (ECM). Here, we investigate the effects of gallic acid (GA) and mechanical stretching on the expression of anabolic and catabolic genes and restoring ECM production by osteoarthritic human articular chondrocytes (hAChs) cultured in monolayers. hAChs were seeded onto conventional plates or silicone chambers with or without 100 µM GA. A 5% cyclic tensile strain (CTS) was applied to the silicone chambers and the deposition of collagen and glycosaminoglycan, and gene expressions of collagen types II (COL2A1), XI (COL11A2), I (COL1A1), and X (COL10A1), and matrix metalloproteinases (MMP-1 and MMP-13) as inflammation markers, were quantified. CTS and GA acted synergistically to promote the deposition of collagen and glycosaminoglycan in the ECM by 14- and 7-fold, respectively. Furthermore, the synergistic stimuli selectively upregulated the expression of cartilage-specific proteins, COL11A2 by 7-fold, and COL2A1 by 47-fold, and, in contrast, downregulated the expression of MMP-1 by 2.5-fold and MMP-13 by 125-fold. GA supplementation with CTS is a promising approach for restoring osteoarthritic hAChs ECM production ability making them suitable for complex tissue engineering applications.


Subject(s)
Cartilage, Articular/drug effects , Extracellular Matrix/genetics , Inflammation/therapy , Muscle Stretching Exercises , Osteoarthritis/therapy , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type I, alpha 1 Chain/genetics , Collagen Type II/genetics , Collagen Type X/genetics , Collagen Type XI/genetics , Extracellular Matrix/drug effects , Gallic Acid/pharmacology , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/pathology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 13/genetics , Osteoarthritis/genetics , Osteoarthritis/pathology
10.
Mol Genet Genomic Med ; 9(10): e1758, 2021 10.
Article in English | MEDLINE | ID: mdl-34423584

ABSTRACT

BACKGROUND: The collagen alpha-1(X) chain gene (COL10A1) is a known causative gene for Schmid metaphyseal chondrodysplasia (SMCD). This study clinically examined a Chinese family (n = 42) for SMCD and inheritance pattern. Fifteen individuals were diagnosed with SMCD based on characteristic skeletal phenotypes with autosomal dominant inheritance mode. METHODS: Four clinically diagnosed patients and three healthy relatives were selected for subsequent genetic tests. Trio-whole exome sequencing (Trio-WES) followed by Sanger sequencing and familial co-segregation analysis were performed to identify SMCD-associated variants. RESULTS: COL10A1 (NM_000493.4):c.1952 G>T(p.Trp651Leu) variant was detected only in the four patients and not in the three healthy relatives. The variant was evaluated as "likely pathogenic" according to the American College of Medical Genetics and Genomics variation classification guidelines with evidence of PM2, PM5, PP1, and PP3. To test the presence of the target variant in proband's fetal offspring, we developed a noninvasive prenatal testing method by extracting cell-free fetal DNA in maternal plasma followed by high-depth sequencing. The variant was also detected in the fetus and later confirmed by amniocentesis. CONCLUSION: We identified a new disease-causing variant in COL10A1. Cell-free fetal DNA in maternal peripheral blood can be used as the rapid and noninvasive prenatal diagnostic method to detect the pathogenic/or likely pathogenic variant.


Subject(s)
Alleles , Collagen Type X/genetics , Genetic Predisposition to Disease , Mutation , Noninvasive Prenatal Testing , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/etiology , Adult , Aged , Female , Genes, Dominant , Genetic Association Studies , Genetic Testing , Genotype , Humans , Male , Middle Aged , Noninvasive Prenatal Testing/methods , Pedigree , Phenotype , Pregnancy , Radiography , Sequence Analysis, DNA , Exome Sequencing
11.
J Orthop Surg Res ; 16(1): 512, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-34407854

ABSTRACT

BACKGROUND: Developmental dysplasia of the hip (DDH) is a highly prevalent hip disease among children. However, its pathogenesis remains unclear. MicroRNAs (miRNA) are important regulators of cartilage development. In a previous study, high-throughput miRNA sequencing of tissue samples from an animal model of DDH showed a low level of miR-1-3p in the cartilage of the acetabular roof (ARC), but its role in DDH pathogenesis was not addressed. Therefore, our aim here was to investigate the effects of miR-1-3p in the ARC. METHODS: The diagnosis of acetabular dysplasia was confirmed with X-ray examination, while imaging and HE staining were conducted to further evaluate the ARC thickness in each animal model. FISH was employed to verify miR-1-3p expression in the ARC and chondrocytes. The miR-1-3p target genes were predicted by a bioinformatics database. A dual-luciferase reporter assay was used to confirm the targeting relationship between miR-1-3p and SOX9. The gene expression of miR-1-3p, SOX9, RUNX2 and collagen type X was evaluated by qPCR analysis. The protein expression of SOX9, RUNX2 and collagen type X was detected by western blot analysis. The levels of SOX9, RUNX2, and collagen type X in the ARC were further assessed via immunohistochemistry analysis. Finally, Alizarin Red S staining was used to observe the mineralized nodules produced by the chondrocytes. RESULTS: We observed low expression of miR-1-3p in the ARC of animals with DDH. SOX9 is a miR-1-3p target gene. Using miR-1-3p silencing technology in vitro, we demonstrated significantly reduced chondrocyte-generated mineralized nodules compared to those of the control. We also confirmed that with miR-1-3p silencing, SOX9 expression was upregulated, whereas the expression of genes associated with endochondral osteogenesis such as RUNX2 and collagen type X was downregulated. To confirm the involvement of miR-1-3p silencing in abnormal ossification through SOX9, we also performed a rescue experiment in which SOX9 silencing restored the low expression of RUNX2 and collagen type X produced by downregulated miR-1-3p expression. Finally, the elevated SOX9 levels and reduced RUNX2 and collagen type X levels in the ARC of rabbits with DDH were also verified using immunohistochemistry, RT-PCR, and western blots. CONCLUSION: The relatively low expression of miR-1-3p in the ARC may be the cause of abnormal endochondral ossification in the acetabular roof of animals with DDH.


Subject(s)
Chondrocytes , MicroRNAs , Animals , Chondrocytes/metabolism , Collagen Type X/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Down-Regulation , Hypertrophy , MicroRNAs/genetics , Rabbits
12.
Mol Genet Genomic Med ; 9(5): e1668, 2021 05.
Article in English | MEDLINE | ID: mdl-33764685

ABSTRACT

BACKGROUND: Schmid-type metaphyseal chondrodysplasia (SMCD) is a rare autosomal dominant skeletal dysplasia caused by heterozygous mutations in COL10A1, the gene which encodes collagen type X alpha 1 chain. However, its genotype-phenotype relationship has not been fully determined. Subjects and Methods The proband is a 2-year-old boy, born of non-consanguineous Chinese parents. We conducted a systematic analysis of the clinical and radiological characteristics and a follow-up study of the proband. Whole-exome sequencing was applied for the genetic analysis, together with bioinformatic analysis of predicted consequences of the identified variant. A homotrimer model was built to visualize the affected region and predict possible outcomes of this variant. Furthermore, a literature review and genotype-phenotype analysis were performed by online searching all cases with SMCD. RESULTS: A novel heterozygous variant (NM_000493.4: c.1863_1866delAATG, NP_000484.2: p.(Met622 Thrfs*54)) was identified in COL10A1 gene in the affected child. And it was predicted to be pathogenic by in silico analysis. Protein modeling revealed that the variant was located in the NC1 domain, which was predicted to produce truncated collagen and impair the trimerization of collagen type X alpha 1 chain and combination with molecules in the matrix. Moreover, genotype-phenotype correlation analysis demonstrated that patients with truncating variants or variants in NC1 domain often presented earlier onset and severer symptoms compared with those with non-truncating or variants in non-NC1 domains. CONCLUSION: The NC1 domain of COL10A1 was proved to be the hotspot region underlying SMCD, patients with variants in NC1 domain were more likely to present severer manifestations at an earlier age.


Subject(s)
Collagen Type X/genetics , Osteochondrodysplasias/genetics , Child, Preschool , Collagen Type X/chemistry , Collagen Type X/metabolism , Heterozygote , Humans , Male , Mutation , Osteochondrodysplasias/pathology , Phenotype , Protein Multimerization
13.
Stem Cell Reports ; 16(3): 610-625, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33636111

ABSTRACT

Chondrodysplasias are hereditary diseases caused by mutations in the components of growth cartilage. Although the unfolded protein response (UPR) has been identified as a key disease mechanism in mouse models, no suitable in vitro system has been reported to analyze the pathology in humans. Here, we developed a three-dimensional culture protocol to differentiate hypertrophic chondrocytes from induced pluripotent stem cells (iPSCs) and examine the phenotype caused by MATN3 and COL10A1 mutations. Intracellular MATN3 or COL10 retention resulted in increased ER stress markers and ER size in most mutants, but activation of the UPR was dependent on the mutation. Transcriptome analysis confirmed a UPR with wide-ranging changes in bone homeostasis, extracellular matrix composition, and lipid metabolism in the MATN3 T120M mutant, which further showed altered cellular morphology in iPSC-derived growth-plate-like structures in vivo. We then applied our in vitro model to drug testing, whereby trimethylamine N-oxide led to a reduction of ER stress and intracellular MATN3.


Subject(s)
Cartilage/physiology , Chondrocytes/physiology , Collagen Type X/metabolism , Induced Pluripotent Stem Cells/physiology , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Animals , Bone and Bones/metabolism , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chondrogenesis , Collagen Type X/genetics , Endoplasmic Reticulum Stress , Extracellular Matrix/metabolism , Gene Editing , Gene Expression Profiling , Homeostasis , Humans , Induced Pluripotent Stem Cells/cytology , Male , Matrilin Proteins/genetics , Matrilin Proteins/metabolism , Mice , Models, Biological , Mutation , Osteochondrodysplasias/pathology , Phenotype , Unfolded Protein Response
14.
Med Sci Monit ; 27: e928919, 2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33637669

ABSTRACT

BACKGROUND Breast cancer, a common malignant tumor, has been considered as the leading cause of cancer-related death in women. Collagen type X alpha 1 (COL10A1) is overexpressed in breast cancer. The current study was designed to determine the functional involvement and regulatory mechanism of COL10A1 on the growth and metastasis of breast cancer. MATERIAL AND METHODS COL10A1 and Prolyl 4-hydroxylase beta polypeptide (P4HB) expressions in normal tissues and tumor tissues of breast cancer patients were obtained from the GEPIA dataset. COL10A1 and P4HB levels in breast cancer cell lines were detected by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot analysis. Furthermore, the interaction between COL10A1 and P4HB was confirmed by co-immunoprecipitation (Co-IP) assay. Cell Counting Kit-8 (CCK-8) and colony formation assay were applied to evaluate cell proliferation and clone-forming abilities of breast cancer cells. In addition, wound healing assay and transwell assay were performed to measure cell migration and invasion capabilities, respectively, in breast cancer. RESULTS The GEPIA dataset presented overexpressed COL10A1 and P4HB in tumor tissues of breast cancer patients. COL10A1 and P4HB expression levels were greatly upregulated in breast cancer cell lines. In addition, COL10A1 could directly interact with P4HB. Functionally, overexpressed COL10A1 boosted the proliferation and metastasis of breast cancer cells and silenced COL10A1 impeded the progression of breast cancer. More importantly, knockdown of P4HB weakened the promoting effects of overexpressed COL10A1 on cell proliferation, migration, and invasion in breast cancer. CONCLUSIONS COL10A1 promotes the malignant progression of breast cancer by upregulating P4HB expression, indicating that COL10A1 functions as an oncogene in breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Collagen Type X/metabolism , Procollagen-Proline Dioxygenase/genetics , Protein Disulfide-Isomerases/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Collagen Type X/genetics , Databases, Genetic , Disease Progression , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , MCF-7 Cells , Neoplasm Metastasis/genetics , Procollagen-Proline Dioxygenase/metabolism , Prolyl Hydroxylases/genetics , Prolyl Hydroxylases/metabolism , Protein Disulfide-Isomerases/metabolism
15.
Bioengineered ; 12(1): 127-137, 2021 12.
Article in English | MEDLINE | ID: mdl-33371777

ABSTRACT

Collagen type X alpha 1 (COL10A1) is a member of the collagen family and the main matrix component. However, COL10A1 expression and prognosis relationship remains unclear in gastric cancer (GC). Through the analysis of database of Oncomine, the Cancer Genome Atlas (TCGA) as well as the Gene Expression Omnibus (GEO), in contrast to the tissue of normal gastric, COL10A1 in gastric cancer, had been upregulated. The high expression of COL10A1 was obviously related to T stage (P = 0.025) and lymph node metastasis (P = 0.025). It has been illustrated by the analysis of logistic regression that COL10A1's heightened expression in gastric cancer had been essentially linked with pathological stage, tumor differentiation, and T classification. The Kaplan-Meier curve in the Kaplan-Meier plotter database (P = 0.0371) and GSE84437 (P = 0.002) indicate that patients with high COL10A1 expression possess poor prognosis, specifically GC patients with lymph node metastasis have it. TCGA's Multivariate analysis (P = 0.025) and GSE84437 dataset (P = 0.034) show that high expression COL10A1 is a key independent predictor of poor overall survival. Searching KEGG pathway enrichment by GSEA, the results suggested that 29 pathways were enriched. qRT-PCR technique was used for verification of the COL10A1's high expression in gastric cancer in contrast to the normal gastric tissues. In conclusion, COL10A1 is of great importance in predicting the survival rate of GC patients.


Subject(s)
Collagen Type X , Stomach Neoplasms , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Collagen Type X/analysis , Collagen Type X/genetics , Collagen Type X/metabolism , Computational Biology , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
16.
Mol Med Rep ; 22(5): 4031-4040, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33000215

ABSTRACT

Histone deacetylase 4 (HDAC4) plays a vital role in chondrocyte hypertrophy and bone formation. To investigate the function of HDAC4 in postnatal skeletal development, the present study developed lineage­specific HDAC4­knockout mice [collagen type 2α1 (Col2α1)­Cre, HDAC4d/d mice] by crossing transgenic mice expressing Cre recombinase. Thus, a specific ablation of HDAC4 was performed in Col2α1­expressing mice cells. The knee joints of HDAC4fl/fl and Col2α1­Cre, HDAC4d/d mice were analyzed at postnatal day (P)2­P21 using an in vivo bromodeoxyuridine (BrdU) assay, and Safranin O, Von Kossa and whole­body staining were used to evaluate the developmental growth plate, hypertrophic differentiation, mineralization and skeletal mineralization patterns. The trabecular bone was analyzed using microcomputed tomography. The expressions of BrdU, proliferating cell nuclear antigen (PCNA), matrix metalloproteinase (MMP)­13, runt­related transcription factor (Runx)­2, osteoprotegerin (OPG), CD34, type X collagen (ColX), osteocalcin and Wnt5a were determined using immunohistochemistry, in situ hybridization (ISH) and reverse transcription­quantitative (RT­q)PCR. The results demonstrated that HDAC4­null mice (HDAC4d/d mice) were severely runted; these mice had a shortened hypertrophic zone (histopathological evaluation), accelerated vascular invasion and articular mineralization (Von Kossa staining), elevated expressions of MMP­13, Runx2, OPG and CD34 (RT­qPCR and immunohistochemistry), downregulated expression of the proliferative marker BrdU and PCNA (immunohistochemistry), increased expression of ColX and decreased expression of Wnt5a (ISH). In conclusion, chondrocyte­derived HDAC4 was responsible for regulating chondrocyte proliferation and differentiation as well as endochondral bone formation.


Subject(s)
Cell Enlargement , Chondrocytes/metabolism , Collagen Type II/metabolism , Gene Deletion , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Osteogenesis/genetics , Animals , Cancellous Bone/pathology , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Chondrogenesis/genetics , Collagen Type X/genetics , Collagen Type X/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Knockout , X-Ray Microtomography
17.
Inflamm Res ; 69(11): 1123-1132, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32809048

ABSTRACT

BACKGROUND AND PURPOSE: Osteoarthritis (OA) impacts the quality of life in middle-aged and elderly people by inducing immobility. The severe inflammation in chondrocytes is reported to be related to the development and process of OA. The present study aims to investigate the protective effects of Apremilast on injured chondrocytes induced by interleukin-1α (IL-1α) and the underlying mechanism. METHODS: 10 ng/mL IL-1α was used to induce the in vitro injured chondrocytes. QRT-PCR was used to evaluate the expression level of Sry-type high-mobility-group box 9 (SOX-9), collagen type II alpha-1 gene (COL2A1), Aggrecan (ACAN) and collagen type X alpha 1 chain (COL10A1). SiRNA technology was utilized to knock down the expression of SOX-9 in the chondrocytes. The expression of SOX-9 was determined by Western Blot assay and/or immunofluorescence assay. Western Blot was used to evaluate the expression level of phosphorylated cyclic AMP response element binding (CREB). RESULTS: SOX9, Col2a1 and Acan were significantly up-regulated and Col10a1 was significantly down-regulated in the chondrocytes by Apremilast in a dose-dependent manner. IL-1α induced the injured chondrocytes by decreasing the expression of SOX9, Col2a1, Acan and increasing the expression of Col10a1, which were greatly reversed by Apremilast. By silencing SOX-9, the effects of Apremilast on SOX9 and marker genes were abolished. Phosphorylated CREB was up-regulated by Apremilast in a time-dependent manner. The up-regulated SOX-9 by Apremilast was reversed by the protein kinase A (PKA)/CREB pathway inhibitor H89. CONCLUSION: Apremilast may protect chondrocytes from inflammation by up-regulating SOX9.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , SOX9 Transcription Factor/genetics , Thalidomide/analogs & derivatives , Aggrecans/genetics , Cell Line , Chondrocytes/metabolism , Collagen Type II/genetics , Collagen Type X/genetics , Down-Regulation/drug effects , Humans , Interleukin-1alpha/pharmacology , Thalidomide/pharmacology , Up-Regulation/drug effects
18.
Biomed Res ; 41(4): 187-197, 2020.
Article in English | MEDLINE | ID: mdl-32801268

ABSTRACT

As chondrocytes fail to retain their chondrogenic potential in two-dimensional monolayer cultures, several three-dimensional culture systems have been employed for investigating the physiology and pathophysiology in articular cartilage tissues. In this study, we introduced a polyethylene glycol-coated microfabricated chip that enables spheroid formation from ATDC5 cell line, commonly used as a model for in vitro chondrocyte research. ATDC5 cells cultured in our devices aggregated immediately and generated a single spheroid per well within 24 h. Most cells in spheroids cultured in differentiation medium were viable and the circular shape and smooth surface of the spheroid were maintained up to 14 d in culture. We also detected potent hypoxia conditions, a key factor in chondrogenesis, in whole lesions of ATDC5 spheroids. Expression of chondrogenesis-related genes and type X collagen protein was significantly increased in ATDC5 spheroids grown in differentiation medium, compared with monolayer-cultured ATDC5 cells. We also demonstrated that the differentiation medium-induced Akt protein phosphorylation was upregulated in ATDC5 cells cultured in our spheroid device, suggesting that enhancement of chondrogenic potential in ATDC5 spheroids results from PI3/Akt signaling activation. These results indicated that our spheroid culture system could constitute a high-throughput strategy approach towards elucidating the molecular mechanisms that regulate chondrogenesis.


Subject(s)
Cell Culture Techniques , Cell Proliferation/drug effects , Chondrocytes/drug effects , Coated Materials, Biocompatible/pharmacology , Polyethylene Glycols/pharmacology , Spheroids, Cellular/drug effects , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/genetics , Cell Line , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/drug effects , Chondrogenesis/genetics , Coated Materials, Biocompatible/chemistry , Collagen Type II/genetics , Collagen Type II/metabolism , Collagen Type X/genetics , Collagen Type X/metabolism , Gene Expression , Hyaluronan Synthases/genetics , Hyaluronan Synthases/metabolism , Lab-On-A-Chip Devices , Mice , Platinum/chemistry , Polymethyl Methacrylate/chemistry , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism
19.
Ecotoxicol Environ Saf ; 204: 111058, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32739676

ABSTRACT

Skeletal fluorosis causes growth plate impairment and growth retardation during bone development. However, the mechanism of how fluoride impairs chondrocyte is unclear. To explore the effect of fluoride on chondrocyte differentiation and the regulation of circadian clock signaling pathway during chondrogenesis, we treated ATDC5 cells with fluoride and carried out a series of experiments. 10-3 M fluoride inhibited cell viability and significantly decreased the expression of Sox9 and Col2a1 (P < 0.05). Fluoride inhibited proteoglycan synthesis and decreased significantly the expression of Aggrecan, Ihh and Col10a1 (P < 0.05). Meanwhile, fluoride significantly inhibited the expression of Bmal1 and disrupted circadian clock signaling pathway (P < 0.05). Furthermore, fluoride disrupted the time-dependent expression of circadian clock molecules and stage-specific differentiation markers. Overexpression of Bmal1 by lentivirus reversed the adverse effects of fluoride on chondrogenesis. These results suggested that fluoride inhibited chondrocyte viability and delayed chondrocyte differentiation. Fluoride delayed chondrogenesis partly via interfering with Bmal1 and circadian clock signaling pathway. Nevertheless, the specific mechanism of circadian clock in fluoride-induced cartilage damage needs to be further studied.


Subject(s)
Cell Differentiation/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects , Circadian Clocks , Environmental Pollutants/toxicity , Fluorides/toxicity , Animals , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis/physiology , Collagen Type X/genetics , Collagen Type X/metabolism , Growth Plate/drug effects , Growth Plate/metabolism , Mice , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction
20.
PLoS One ; 15(8): e0237479, 2020.
Article in English | MEDLINE | ID: mdl-32790806

ABSTRACT

OBJECTIVE: As native cartilage consists of different phenotypical zones, this study aims to fabricate different types of neocartilage constructs from collagen hydrogels and human mesenchymal stromal cells (MSCs) genetically modified to express different chondrogenic factors. DESIGN: Human MSCs derived from bone-marrow of osteoarthritis (OA) hips were genetically modified using adenoviral vectors encoding sex-determining region Y-type high-mobility-group-box (SOX) 9, transforming growth factor beta (TGFB) 1 or bone morphogenetic protein (BMP) 2 cDNA, placed in type I collagen hydrogels and maintained in serum-free chondrogenic media for three weeks. Control constructs contained unmodified MSCs or MSCs expressing GFP. The respective constructs were analyzed histologically, immunohistochemically, biochemically, and by qRT-PCR for chondrogenesis and hypertrophy. RESULTS: Chondrogenesis in MSCs was consistently and strongly induced in collagen I hydrogels by the transgenes SOX9, TGFB1 and BMP2 as evidenced by positive staining for proteoglycans, chondroitin-4-sulfate (CS4) and collagen (COL) type II, increased levels of glycosaminoglycan (GAG) synthesis, and expression of mRNAs associated with chondrogenesis. The control groups were entirely non-chondrogenic. The levels of hypertrophy, as judged by expression of alkaline phosphatase (ALP) and COL X on both the protein and mRNA levels revealed different stages of hypertrophy within the chondrogenic groups (BMP2>TGFB1>SOX9). CONCLUSIONS: Different types of neocartilage with varying levels of hypertrophy could be generated from human MSCs in collagen hydrogels by transfer of genes encoding the chondrogenic factors SOX9, TGFB1 and BMP2. This technology may be harnessed for regeneration of specific zones of native cartilage upon damage.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Hydrogels/chemistry , SOX9 Transcription Factor/genetics , Transforming Growth Factor beta1/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bone Morphogenetic Protein 2/metabolism , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrogenesis/genetics , Collagen Type I/chemistry , Collagen Type X/genetics , Culture Media, Serum-Free/chemistry , Glycosaminoglycans/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , RNA, Messenger/metabolism , SOX9 Transcription Factor/metabolism , Transforming Growth Factor beta1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL