Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Adv Sci (Weinh) ; 11(29): e2306860, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38864559

ABSTRACT

Breast tumor-initiating cells (BTICs) of triple-negative breast cancer (TNBC) tissues actively repair DNA and are resistant to treatments including chemotherapy, radiotherapy, and targeted therapy. Herein, it is found that a previously reported secreted protein, sclerostin domain containing 1 (SOSTDC1), is abundantly expressed in BTICs of TNBC cells and positively correlated with a poor patient prognosis. SOSTDC1 knockdown impairs homologous recombination (HR) repair, BTIC maintenance, and sensitized bulk cells and BTICs to Olaparib. Mechanistically, following Olaparib treatment, SOSTDC1 translocates to the nucleus in an importin-α dependent manner. Nuclear SOSTDC1 interacts with the N-terminus of the nucleoprotein, chromatin helicase DNA-binding factor (CHD1), to promote HR repair and BTIC maintenance. Furthermore, nuclear SOSTDC1 bound to ß-transducin repeat-containing protein (ß-TrCP) binding motifs of CHD1 is found, thereby blocking the ß-TrCP-CHD1 interaction and inhibiting ß-TrCP-mediated CHD1 ubiquitination and degradation. Collectively, these findings identify a novel nuclear SOSTDC1 pathway in regulating HR repair and BTIC maintenance, providing insight into the TNBC therapeutic strategies.


Subject(s)
Adaptor Proteins, Signal Transducing , DNA-Binding Proteins , Phthalazines , Piperazines , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/drug therapy , Female , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Piperazines/pharmacology , Phthalazines/pharmacology , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Line, Tumor , Animals , Drug Resistance, Neoplasm/genetics , Recombinational DNA Repair/genetics , Disease Progression , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Disease Models, Animal , Cell Nucleus/metabolism , DNA Helicases
2.
J Anim Sci Biotechnol ; 14(1): 88, 2023 Jul 08.
Article in English | MEDLINE | ID: mdl-37420295

ABSTRACT

BACKGROUND: Wool fibers are valuable materials for textile industry. Typical wool fibers are divided into medullated and non-medullated types, with the former generated from primary wool follicles and the latter by either primary or secondary wool follicles. The medullated wool is a common wool type in the ancestors of fine wool sheep before breeding. The fine wool sheep have a non-medullated coat. However, the critical period determining the type of wool follicles is the embryonic stage, which limits the phenotypic observation and variant contrast, making both selection and studies of wool type variation fairly difficult. RESULTS: During the breeding of a modern fine (MF) wool sheep population with multiple-ovulation and embryo transfer technique, we serendipitously discovered lambs with ancestral-like coarse (ALC) wool. Whole-genome resequencing confirmed ALC wool lambs as a variant type from the MF wool population. We mapped the significantly associated methylation locus on chromosome 4 by using whole genome bisulfite sequencing signals, and in turn identified the SOSTDC1 gene as exons hypermethylated in ALC wool lambs compare to their half/full sibling MF wool lambs. Transcriptome sequencing found that SOSTDC1 was expressed dozens of times more in ALC wool lamb skin than that of MF and was at the top of all differentially expressed genes. An analogy with the transcriptome of coarse/fine wool breeds revealed that differentially expressed genes and enriched pathways at postnatal lamb stage in ALC/MF were highly similar to those at the embryonic stage in the former. Further experiments validated that the SOSTDC1 gene was specifically highly expressed in the nucleus of the dermal papilla of primary wool follicles. CONCLUSION: In this study, we conducted genome-wide differential methylation site association analysis on differential wool type trait, and located the only CpG locus that strongly associated with primary wool follicle development. Combined with transcriptome analysis, SOSTDC1 was identified as the only gene at this locus that was specifically overexpressed in the primary wool follicle stem cells of ALC wool lamb skin. The discovery of this key gene and its epigenetic regulation contributes to understanding the domestication and breeding of fine wool sheep.

3.
J Bone Miner Res ; 38(5): 765-774, 2023 05.
Article in English | MEDLINE | ID: mdl-36891756

ABSTRACT

The development of Wnt-based osteoanabolic agents has progressed rapidly in recent years, given the potent effects of Wnt modulation on bone homeostasis. Simultaneous pharmacologic inhibition of the Wnt antagonists sclerostin and Dkk1 can be optimized to create potentiated effects in the cancellous bone compartment. We looked for other candidates that might be co-inhibited along with sclerostin to potentiate the effects in the cortical compartment. Sostdc1 (Wise), like sclerostin and Dkk1, also binds and inhibits Lrp5/6 coreceptors to impair canonical Wnt signaling, but Sostdc1 has greater effects in the cortical bone. To test this concept, we deleted Sostdc1 and Sost from mice and measured the skeletal effects in cortical and cancellous compartments individually. Sost deletion alone produced high bone mass in all compartments, whereas Sostdc1 deletion alone had no measurable effects on either envelope. Mice with codeletion of Sostdc1 and Sost had high bone mass and increased cortical properties (bone mass, formation rates, mechanical properties), but only among males. Combined administration of sclerostin antibody and Sostdc1 antibody in wild-type female mice produced potentiation of cortical bone gain despite no effect of Sostdc1 antibody alone. In conclusion, Sostdc1 inhibition/deletion can work in concert with sclerostin deficiency to improve cortical bone properties. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Glycoproteins , Intercellular Signaling Peptides and Proteins , Male , Female , Animals , Mice , Intercellular Signaling Peptides and Proteins/metabolism , Glycoproteins/metabolism , Bone and Bones/metabolism , Cortical Bone/metabolism , Cancellous Bone/metabolism , Adaptor Proteins, Signal Transducing/metabolism
4.
Front Physiol ; 13: 1029646, 2022.
Article in English | MEDLINE | ID: mdl-36338475

ABSTRACT

Sclerostin domain-containing protein-1 (Sostdc1) is a member of the sclerostin family and encodes a secreted 28-32 kDa protein with a cystine knot-like domain and two N-linked glycosylation sites. Sostdc1 functions as an antagonist to bone morphogenetic protein (BMP), mediating BMP signaling. It also interacts with LRP6, mediating LRP6 and Wnt signaling, thus regulating cellular proliferation, differentiation, and programmed cell death. Sostdc1 plays various roles in the skin, intestines, brain, lungs, kidneys, and vasculature. Deletion of Sostdc1 gene in mice resulted in supernumerary teeth and improved the loss of renal function in Alport syndrome. In the skeletal system, Sostdc1 is essential for bone metabolism, bone density maintenance, and fracture healing. Recently, Sostdc1 has been found to be closely related to the development and progression of multiple cancer types, including breast, renal, gastric, and thyroid cancers. This article summarises the role of Sostdc1 in skeletal biology and related cancers to provide a theoretical basis for the treatment of related diseases.

5.
Curr Issues Mol Biol ; 44(5): 2167-2174, 2022 May 12.
Article in English | MEDLINE | ID: mdl-35678675

ABSTRACT

In our previous study, we found that lymphatic vessels stimulate hair follicle growth through paracrine effects on dermal papilla cells. However, the paracrine factors secreted from cutaneous lymphatic vessels that can activate dermal papilla cells are still unknown. In this study, we investigated whether lymphatic endothelial cells might secrete paracrine factors that activate dermal papilla cells in vitro. We found that Sostdc1 was more expressed in lymphatic endothelial cells compared with blood vascular endothelial cells. In addition, Sostdc1 expression levels were significantly increased during the anagen phase in the back skin of C57BL/6J mice, as compared to the telogen phase. We also observed that incubation of dermal papilla cells with 200 ng/mL Sostdc1 for 72 h induced the expression levels of Lef-1, a downstream target of Wnt signaling. Taken together, our results reveal that Sostdc1, a BMP antagonist, secreted from cutaneous lymphatic vessels, may act as a paracrine factor for hair follicle growth.

6.
Environ Toxicol ; 37(8): 1934-1943, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35442555

ABSTRACT

Sclerostin domain-containing 1 (SOSTDC1) has been documented as a key tumor-associated protein that is differentially expressed in multiple malignancies. However, the function of SOSTDC1 in acute myeloid leukemia (AML) is unexplored. The goal of this work was to assess the possible role of SOSTDC1 in AML. Our data showed decreased SOSTDC1 level in bone marrow from AML patients, and patients with low levels of SOSTDC1 had a reduced survival rate. SOSTC1 upregulation restrained the proliferative ability and promoted the apoptotic rate of AML cells. SOSTDC1 suppressed the activation of the Wnt/ß-catenin pathway in AML cells. Reactivation of the Wnt/ß-catenin pathway reversed SOSTDC1-mediated antitumor effects. SOSTDC1 upregulation weakened the tumorigenicity of AML cells in vivo. Collectively, our work demonstrates that SOSTDC1 has a tumor-inhibiting role in AML via downregulation of the Wnt/ß-catenin pathway. This work underscores a key function for the SOSTDC1/Wnt/ß-catenin pathway in AML.


Subject(s)
Leukemia, Myeloid, Acute , beta Catenin , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Neoplasm Proteins , Wnt Signaling Pathway , beta Catenin/metabolism
7.
Biochem Genet ; 60(6): 1895-1913, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35138469

ABSTRACT

Gastric cancer (GC) is the fifth most common cancer, which has a significant impact on human health. Recent researches have shown that circular RNAs (circRNAs) could affect the progress of GC, but the mechanism still indistinct. In this work, we explored the roles of circ_0001190 in GC. The levels of circ_0001190, microRNA-586 (miR-586) and sclerostin domain containing 1 (SOSTDC1) were detected by quantitative RT-PCR and western blot in GC. The cell functions were scrutinized by cell counting kit-8 assay, 5-Ethynyl-29-deoxyuridine assay, flow cytometry assay, tube formation assay, transwell assay, and western blot. Furthermore, the relationship between miR-586 and circ_0001190 or SOSTDC1 was identified by dual-luciferase reporter assay. Finally, the xenograft model test was implemented to demonstrate the effect of exosomal circ_0001190 in vivo. The levels of circ_0001190 and SOSTDC1 were downregulated, and the miR-586 level was increased in GC. For functional assay, circ _0001190 overexpression inhibited cell vitality, cell proliferation, angiogenesis, cell migration and invasion, whereas stimulated cell apoptosis in GC cells. Circ _0001190 served as a miR-586 sponge to adjust the expression of SOSTDC1. Additionally, miR-586 could promote the advancement of GC by interfering SOSTDC1. Exosomal circ_0001190 overexpression inhibited the development of GC by miR-586/SOSTDC1 axis, which proposed a potential targeted therapy for GC cure.


Subject(s)
Adaptor Proteins, Signal Transducing , MicroRNAs , RNA, Circular , Stomach Neoplasms , Humans , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA, Circular/genetics , Stomach Neoplasms/genetics
8.
Cell Cycle ; 20(12): 1147-1162, 2021 06.
Article in English | MEDLINE | ID: mdl-34110955

ABSTRACT

Ovarian cancer (OC) is the fifth most common female malignant tumor and the leading cause of cancer-related death in women worldwide. Epithelial ovarian cancer (EOC) is the predominant type of OC. Investigating the mechanism underlying tumorigenesis and progression of EOC is urgent. Our previous research has shown that long non-coding RNAs (lncRNAs) CDKN2A-AS1 is upregulated in EOC tissues and cells. Furthermore, we have predicted that CDKN2A-AS1 is associated with the bone morphogenetic protein (BMP)-SMAD signaling pathway, which is negatively regulated by the sclerostin domain containing 1 (SOSTDC1). Therefore, we conjecture that the CDKN2A-AS1 regulate BMP-SMAD signaling pathway via interacting with SOSTDC1, which need more investigation. Moreover, the functions of the BMP-SMAD signaling pathway and the SOSTDC1 on EOC are still unclear. Herein, we unearthed that CDKN2A-AS1, BMP2/4/7, SMAD1/5/9 and phosphorylation of SMAD1/5/9 (p-SMAD1/5/9) were upregulated in EOC tissues and cells, whereas SOSTDC1 was downregulated in EOC tissues and cells. We firstly demonstrated that CDKN2A-AS1 bound directly with the SOSTDC1. CDKN2A-AS1 downregulated the expression of SOSTDC1, but upregulated the expression of BMP2/4/7, SMAD1/5/9, and p-SMAD1/5/9. CDKN2A-AS1 promoted the proliferation, migration, invasion of EOC cells and tumor growth in vivo, whereas SOSTDC1 inhibited the proliferation, migration, invasion of EOC cells. Knockdown SOSTDC1 rescued the inhibitory effect of si-lncRNA CDKN2A-AS1 on the EOC cells proliferation, migration and invasion. These results demonstrated that CDKN2A-AS1activated the BMP-SMAD signaling pathway by directly bind with SOSTDC1 to promote EOC tumor growth. CDKN2A-AS1/SOSTDC1 axis may provide a novel therapeutic strategy for EOC treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Proteins/metabolism , Carcinogenesis/metabolism , Carcinoma, Ovarian Epithelial/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Disease Progression , Ovarian Neoplasms/metabolism , RNA, Antisense , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Smad Proteins, Receptor-Regulated/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Down-Regulation/genetics , Female , Gene Knockdown Techniques , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Long Noncoding/genetics , Transfection , Up-Regulation/genetics
9.
Biochem J ; 477(17): 3167-3182, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32779697

ABSTRACT

The DAN (differential screening-selected gene aberrative in neuroblastoma) family are a group of secreted extracellular proteins which typically bind to and antagonize BMP (bone morphogenetic protein) ligands. Previous studies have revealed discrepancies between the oligomerization state of certain DAN family members, with SOST (a poor antagonist of BMP signaling) forming a monomer while Grem1, Grem2, and NBL1 (more potent BMP antagonists) form non-disulfide linked dimers. The protein SOSTDC1 (Sclerostin domain containing protein 1) is sequentially similar to SOST, but has been shown to be a better BMP inhibitor. In order to determine the oligomerization state of SOSTDC1 and determine what effect dimerization might have on the mechanism of DAN family antagonism of BMP signaling, we isolated the SOSTDC1 protein and, using a battery of biophysical, biochemical, and structural techniques, showed that SOSTDC1 forms a highly stable non-covalent dimer. Additionally, this SOSTDC1 dimer was shown, using an in vitro cell based assay system, to be an inhibitor of multiple BMP signaling growth factors, including GDF5, while monomeric SOST was a very poor antagonist. These results demonstrate that SOSTDC1 is distinct from paralogue SOST in terms of both oligomerization and strength of BMP inhibition.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Avian Proteins/chemistry , Protein Multimerization , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cell Line , Chickens , Humans , Signal Transduction
10.
Am J Cancer Res ; 9(11): 2331-2348, 2019.
Article in English | MEDLINE | ID: mdl-31815038

ABSTRACT

Gastric cancer is commonly diagnosed at an advanced stage when metastasis is almost inevitable. Despite numerous novel regulators have been identified in driving gastric cancer progression, much remains unclear due to the complex nature of cancer. Comparison of the transcriptome profiles of gastric primary tumor tissue, with its matched non-tumor and lymph node metastasis revealed frequent stepwise down-regulation of sclerostin domain containing 1 (SOSTDC1) related with tumor progression. Clinically, deficiency of this gene is associated with shortened survival of patients. Our results suggest that SOSTDC1 confers tumor-suppressive features in gastric cancer and silencing of it accelerates tumor growth and promotes the formation of lung metastasis. Although SOSTDC1 displayed limited inhibition of canonical SMAD-dependent bone morphogenetic proteins (BMP) pathway, it remarkably restrained the c-Jun activation and transcription of c-Jun downstream targets in the noncanonical BMP signaling pathway. Furthermore, c-Jun N-terminal kinase (JNK) blockage attenuated cell proliferative and migrative advantages of SOSTDC1 knockdown cell lines. Our study comprehensively elucidated the role of SOSTDC1 in gastric cancer progression and the results translate into potential therapy for gastric cancer.

11.
Graefes Arch Clin Exp Ophthalmol ; 257(11): 2401-2427, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31529323

ABSTRACT

PURPOSE: This study was conducted in order to study Sostdc1 expression in rat and human developing and adult eyes. METHODS: Using the yeast signal sequence trap screening method, we identified the Sostdc1 cDNA encoding a protein secreted by the adult rat retinal pigment epithelium. We determined by in situ hybridization, RT-PCR, immunohistochemistry, and western blot analysis Sostdc1 gene and protein expression in developing and postnatal rat ocular tissue sections. We also investigated Sostdc1 immunohistolocalization in developing and adult human ocular tissues. RESULTS: We demonstrated a prominent Sostdc1 gene expression in the developing rat central nervous system (CNS) and eyes at early developmental stages from E10.5 days postconception (dpc) to E13 dpc. Specific Sostdc1 immunostaining was also detected in most adult cells of rat ocular tissue sections. We also identified the rat ocular embryonic compartments characterized by a specific Sostdc1 immunohistostaining and specific Pax6, Sox2, Otx2, and Vsx2 immunohistostaining from embryonic stages E10.5 to E13 dpc. Furthermore, we determined the localization of SOSTDC1 immunoreactivity in ocular tissue sections of developing and adult human eyes. Indeed, we detected SOSTDC1 immunostaining in developing and adult human retinal pigment epithelium (RPE) and neural retina (NR) as well as in several developing and adult human ocular compartments, including the walls of choroidal and scleral vessels. Of utmost importance, we observed a strong SOSTDC1 expression in a pathological ocular specimen of type 2 Peters' anomaly complicated by retinal neovascularization as well in the walls ofother pathological extra-ocular vessels.  CONCLUSION: As rat Sostdc1 and human SOSTDC1 are dual antagonists of the Wnt/ß-catenin and BMP signaling pathways, these results underscore the potential crucial roles of these pathways and their antagonists, such as Sostdc1 and SOSTDC1, in developing and adult mammalian normal eyes as well as in syndromic and nonsyndromic congenital eye diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Eye Diseases, Hereditary/genetics , Gene Expression Regulation, Developmental , RNA/genetics , Retinal Pigment Epithelium/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Aged , Animals , Blotting, Western , Child, Preschool , Disease Models, Animal , Eye Diseases, Hereditary/metabolism , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Rats , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/growth & development
12.
Am J Transl Res ; 11(6): 3722-3736, 2019.
Article in English | MEDLINE | ID: mdl-31312383

ABSTRACT

T follicular helper (TFH) cells are crucial for effective humoral immunity by providing the required signals to cognate B cells and promoting germinal center (GC) formation. Many intrinsic and extrinsic factors have been reported to be involved in the multistage, multifactorial differentiation process of TFH cells. By comparing gene expression between TFH cells and TH1 cells based on published GEO data, we found selective and high expression of sclerostin domain-containing protein 1 (SOSTDC1) in TFH cells but not in TH1 cells; however, it is unclear whether SOSTDC1 is important for the differentiation and/or function of TFH cells. Using a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection, we confirmed the selective expression of SOSTDC1 in TFH cells compared to that in TH1 cells, but the ablation of SOSTDC1 did not affect TFH cell differentiation or effector function. Thus, our results indicate that the SOSTDC1 protein is merely a specific marker of TFH cells but does not play a functional role in the differentiation of TFH cells during acute viral infection.

13.
Mol Cell Biochem ; 460(1-2): 165-173, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31313025

ABSTRACT

Nonunion is a serious complication after fracture due to its difficulty of self-healing. MicroRNA-26a (miR-26a) has been known to play a crucial role in bone metabolism. In this study, we established a rat nonunion model by removing periosteum, and found that miR-26a was significantly upregulated. Osteogenic differentiation of mesenchymal stem cells (MSCs) isolated from bone marrow transfected with miR-26a mimics was significantly enhanced, evidenced by increased calcium deposition and expression levels of alkaline phosphatase (ALP) and osteocalcin. Bioinformatics analysis suggested that sclerostin domain-containing 1 (SOSTDC1) may be a target of miR-26a, which was confirmed by dual-luciferase assay and western blot. Besides, miR-26a was used for nonunion rats. Delightfully, radiographs of nonunion rats with miR-26a mimics administration showed obvious new bone formation compared with nonhealing control. Hematoxylin-eosin and Masson staining assays revealed that osteogenesis capacity was greatly enhanced by miR-26a mimics' administration. In addition, miR-26a mimics could promote osteogenic differentiation in nonunion rats, evidenced by increased protein levels of ALP and osteocalcin, while SOSTDC1 was suppressed. The injection of miR-26a mimics also gave rise to phosphorylation of GSK3ß and nuclear accumulation of ß-catenin, which indicated the activation of canonical Wnt/ß-catenin signaling. In conclusion, we demonstrated that miR-26a promoted fracture healing of rats with nonunion in vivo and osteogenic differentiation of MSCs in vitro, possibly by targeting SOSTDC1, and that Wnt/ß-catenin signaling pathway was involved in this process.


Subject(s)
Fracture Healing/genetics , Fractures, Ununited/genetics , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Wnt Signaling Pathway , Animals , Base Sequence , Cell Differentiation , Disease Models, Animal , Down-Regulation , Mesenchymal Stem Cells/metabolism , Osteogenesis , Rats, Sprague-Dawley
14.
BMC Oral Health ; 19(1): 90, 2019 05 27.
Article in English | MEDLINE | ID: mdl-31133012

ABSTRACT

BACKGROUND: The purpose of this study was to analyze the association between the genetic polymorphism of genes (PAX6, SOSTDC1and FAM20B) and the susceptibility to mesiodens. METHODS: This study was carried out on 50 patients with mesiodens and 50 controls. The family history of each patient with mesiodens were recorded. Genomic DNA was extracted from saliva samples, and single nucleotide polymorphisms were detected in all exons and exon/intron boundaries of PAX6, SOSTDC1 and FAM20B using Sanger sequencing. The data were analyzed using pearson chi-square test with theoretical frequency ≥ 5. For theoretical frequency less than 5 but at least 1 (≤20% cell), the data were analyzed by continuity correction. For the rest, Fisher's Exact test was used. A P-value< 0.05 was considered statistically significant. The Odds ratio (OR) and confidence intervals (CI) were recorded. RESULTS: Three polymorphisms were detected in PAX6. Two polymorphisms were detected in SOSTDC1. Twenty-nine polymorphisms were detected in FAM20B. Although, the T allele of FAM20B (rs3766626) appears to be associated with mesiodens (P = 0.051), there were no significant differences of PAX6/SOSTDC1/FAM20B gene polymorphisms between the two groups. The T allele of FAM20B (rs3766626) was associated with susceptibility to two mesiodens (P < 0.001; OR = 8.333; CI = 2.516-27.600). CONCLUSIONS: Lack of association between PAX6/SOSTDC1/FAM20B gene polymorphisms and mesiodens in the population studied was detected. Further studies with large samples on T allele of FAM20B (rs3766626) are needed.


Subject(s)
PAX6 Transcription Factor/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proteins/genetics , Tooth, Supernumerary/genetics , Adaptor Proteins, Signal Transducing , Alleles , Case-Control Studies , Exons , Gene Frequency , Genetic Predisposition to Disease , Humans , Intracellular Signaling Peptides and Proteins , Odds Ratio , Polymorphism, Single Nucleotide
15.
Bone ; 122: 82-92, 2019 05.
Article in English | MEDLINE | ID: mdl-30776499

ABSTRACT

Multiple myeloma (MM) is characterised by destructive lytic bone disease, caused by induction of bone resorption and impaired bone formation. Our understanding of the molecular mechanisms responsible for osteoblast suppression, are limited. Using the 5T2MM murine model of MM we have previously shown that suppression of the activity of a known inhibitor of bone formation Dikkopf-1 (Dkk1) prevents the development of lytic bone disease. Here we have demonstrated that another potential inhibitor of bone formation, sclerostin domain containing 1 (Sostdc1) is expressed at low levels in MM and osteoblast lineage cells when these cells are grown separately in cell culture but its expression is significantly induced in both cell types when these cells are in contact. The distribution of Sostdc1 staining in bones infiltrated with 5TGM1 myeloma cells in vivo suggested its presence in both myeloma and osteoblast lineage populations when in close proximity. We have also shown that recombinant Sostdc1 inhibits both bone morphogenic proteins (BMP2 and 7) and Wnt signalling in primary osteoblasts and suppresses differentiation of these cells. Together, these findings suggest that Sostdc1 expression in 5TGM1-infiltrated bones as a result of the interaction between myeloma and osteoblast lineage populations, could result in suppression of osteoblast differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Communication , Cell Lineage , Multiple Myeloma/pathology , Osteoblasts/pathology , Wnt Proteins/antagonists & inhibitors , Animals , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Down-Regulation/drug effects , Male , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Solubility , Stem Cells/drug effects , Stem Cells/metabolism , Tibia/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
J Dent Res ; 98(1): 98-106, 2019 01.
Article in English | MEDLINE | ID: mdl-30325689

ABSTRACT

Crown shapes in mammalian teeth vary considerably from species to species, and morphological characters in crown shape have been used to identify species. Cusp pattern is one of the characters in crown shape. In the processes governing the formation of cusp pattern, the Shh pathway has been implicated as an important player. Suppression of Shh signaling activity in vitro in explant assays appears to induce supernumerary cusp formation in wild-type tooth germs. However, the in vivo role of Shh signaling in cusp pattern formation and the molecular mechanisms by which Shh regulates cusp patterning are not clear. Here, through in vivo phenotypic analyses of mice in which Shh activity was suppressed and compared with wild-type mice, we characterized differences in the location, number, incidence, and shape of supernumerary cusps in molars at embryonic day 15.5. We found that the distances between cusps were reduced in molars of Shh activity-suppressed mice in vivo. These findings confirm and extend the previous idea that Shh acts as an inhibitor in the reaction-diffusion model for cusp pattern formation by negatively regulating the intercuspal distance. We uncovered a significant reduction of expression level of Sostdc1, which encodes a secreted modulator of Wnt signaling, after suppression of Shh activity. The supernumerary cusp formation in Sostdc1-/- mice and compound Sostdc1 and Lrp mutant mice indicates a strong association between Wnt and Shh signaling pathways in cusp patterning. In further support of this idea, there is a high degree of similarity in the supernumerary cusp patterns of mice lacking Sostdc1 or Shh at embryonic day 15.5. These results suggest that Shh plays an inhibitory role in cusp pattern formation by modulating Wnt signaling through the positive regulation of Sostdc1.


Subject(s)
Body Patterning/genetics , Bone Morphogenetic Proteins/physiology , Hedgehog Proteins/physiology , Tooth/embryology , Wnt Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Body Patterning/physiology , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice , Molar , Signal Transduction , Tooth/metabolism , Tooth Crown , Tooth Germ , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway
17.
BMC Cancer ; 18(1): 405, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29642861

ABSTRACT

BACKGROUND: Robust and precise molecular prognostic predictors for luminal breast cancer are required. This study aimed to identify key methylation sites in luminal breast cancer, as well as precise molecular tools for predicting prognosis. METHODS: We compared methylation levels of normal and luminal breast cancer samples from The Cancer Genome Atlas dataset. The relationships among differentially methylated sites, corresponding mRNA expression levels and prognosis were further analysed. Differentially expressed genes in normal and cancerous samples were analysed, followed by the identification of prognostic signature genes. Samples were divided into low- and high-risk groups based on the signature genes. Prognoses of low- and high-risk groups were compared. The Gene Expression Omnibus dataset were used to validate signature genes for prognosis prediction. Prognosis of low- and high-risk groups in Luminal A and Luminal B samples from the TCGA and the Metabric cohort dataset were analyzed. We also analysed the correlation between clinical features of low- and high- risk groups as well as their differences in gene expression. RESULTS: Fourteen methylation sites were considered to be related to luminal breast cancer prognosis because their methylation levels, mRNA expression and prognoses were closely related to each other. The methylation level of SOSTDC1 was used to divide samples into hypo- and hyper-methylation groups. We also identified an mRNA signature, comprising eight transcripts, ESCO2, PACSIN1, CDCA2, PIGR, PTN, RGMA, KLK4 and CENPA, which was used to divide samples into low- and high-risk groups. The low-risk group showed significantly better prognosis than the high-risk group. A correlation analysis revealed that the risk score was an independent prognostic factor. Low- and high- risk groups significantly correlated with the survival ratio in Luminal A samples, but not in Luminal B samples on the basis of the TCGA and the Metabric cohort dataset. Further functional annotation demonstrated that the differentially expressed genes were mainly involved in cell cycle and cancer progression. CONCLUSIONS: We identified several key methylation sites and an mRNA signature for predicting luminal breast cancer prognosis. The signature exhibited effective and precise prediction of prognosis and may serve as a prognostic and diagnostic marker for luminal breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/mortality , DNA Methylation , Epigenesis, Genetic , Adaptor Proteins, Signal Transducing , Adult , Aged , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Kaplan-Meier Estimate , Middle Aged , Prognosis , Proportional Hazards Models , Proteins/genetics , Reproducibility of Results
18.
Development ; 144(15): 2824-2836, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28694256

ABSTRACT

During development and homeostasis, precise control of Wnt/ß-catenin signaling is in part achieved by secreted and membrane proteins that negatively control activity of the Wnt co-receptors Lrp5 and Lrp6. Lrp4 is related to Lrp5/6 and is implicated in modulation of Wnt/ß-catenin signaling, presumably through its ability to bind to the Wise (Sostdc1)/sclerostin (Sost) family of Wnt antagonists. To gain insights into the molecular mechanisms of Lrp4 function in modulating Wnt signaling, we performed an array of genetic analyses in murine tooth development, where Lrp4 and Wise play important roles. We provide genetic evidence that Lrp4 mediates the Wnt inhibitory function of Wise and also modulates Wnt/ß-catenin signaling independently of Wise. Chimeric receptor analyses raise the possibility that the Lrp4 extracellular domain interacts with Wnt ligands, as well as the Wnt antagonists. Diverse modes of Lrp4 function are supported by severe tooth phenotypes of mice carrying a human mutation known to abolish Lrp4 binding to Sost. Our data suggest a model whereby Lrp4 modulates Wnt/ß-catenin signaling via interaction with Wnt ligands and antagonists in a context-dependent manner.


Subject(s)
Receptors, LDL/metabolism , Tooth/embryology , Tooth/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Mice, Mutant Strains , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tooth/pathology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics
19.
J Dent Res ; 96(11): 1273-1281, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28692808

ABSTRACT

Cleft palate is a common birth defect caused by disruption of palatogenesis during embryonic development. Although mutations disrupting components of the Wnt signaling pathway have been associated with cleft lip and palate in humans and mice, the mechanisms involving canonical Wnt signaling and its regulation in secondary palate development are not well understood. Here, we report that canonical Wnt signaling plays an important role in Pax9-mediated regulation of secondary palate development. We found that cleft palate pathogenesis in Pax9-deficient embryos is accompanied by significantly reduced expression of Axin2, an endogenous target of canonical Wnt signaling, in the developing palatal mesenchyme, particularly in the posterior regions of the palatal shelves. We found that expression of Dkk2, encoding a secreted Wnt antagonist, is significantly increased whereas the levels of active ß-catenin protein, the essential transcriptional coactivator of canonical Wnt signaling, is significantly decreased in the posterior regions of the palatal shelves in embryonic day 13.5 Pax9-deficent embryos in comparison with control littermates. We show that small molecule-mediated inhibition of Dickkopf (DKK) activity in utero during palatal shelf morphogenesis partly rescued secondary palate development in Pax9-deficient embryos. Moreover, we found that genetic inactivation of Wise, which is expressed in the developing palatal shelves and encodes another secreted antagonist of canonical Wnt signaling, also rescued palate morphogenesis in Pax9-deficient mice. Furthermore, whereas Pax9del/del embryos exhibit defects in palatal shelf elevation/reorientation and significant reduction in accumulation of hyaluronic acid-a high molecular extracellular matrix glycosaminoglycan implicated in playing an important role in palatal shelf elevation-80% of Pax9del/del;Wise-/- double-mutant mouse embryos exhibit rescued palatal shelf elevation/reorientation, accompanied by restored hyaluronic acid accumulation in the palatal mesenchyme. Together, these data identify a crucial role for canonical Wnt signaling in acting downstream of Pax9 to regulate palate morphogenesis.


Subject(s)
Cleft Palate/embryology , Cleft Palate/genetics , Paired Box Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Embryonic Development , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Mice , Morphogenesis , PAX9 Transcription Factor , Palate/embryology , Signal Transduction/genetics , Transcription Factors/genetics , Wnt Proteins/genetics , beta Catenin/genetics
20.
FEBS Lett ; 591(11): 1584-1600, 2017 06.
Article in English | MEDLINE | ID: mdl-28471520

ABSTRACT

The process of cell reprogramming has been characterized considerably since the successful generation of induced pluripotent stem cells. However, the importance of cell-cell communications for cellular reprogramming remains largely unknown. Secreted factors, which are expressed and secreted during reprogramming, may influence the reprogramming efficiency. Here, we have identified Sostdc1, Glb1l2, Fetub, Dpp4, Gdf3, Trh, and Tdgf1 as prominently upregulated secreted factors during reprogramming. Our detailed analysis reveals that these seven factors may be categorized into four groups based on their expression patterns in relation to the reprogramming stages. Remarkably, knockdown of Sostdc1, which is the most prominently upregulated factor and which is expressed earlier than the other six factors, results in reduced reprogramming efficiency, suggesting its involvement in the reprogramming process.


Subject(s)
Cellular Reprogramming/genetics , Gene Expression Regulation , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cells, Cultured , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fetuin-B/genetics , Fetuin-B/metabolism , Fibroblasts/metabolism , Flow Cytometry , Growth Differentiation Factor 3/genetics , Growth Differentiation Factor 3/metabolism , Immunoblotting , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Microarray Analysis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL