Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
In Vitro Cell Dev Biol Anim ; 54(2): 111-119, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29247399

ABSTRACT

Kidney mainly arises from the induction of metanephric mesenchymal cells (MM cells) and the ureteric bud (UB). Transmembrane protein-100 (Tmem100) consists of two transmembrane regions with strong temporal and spatial expression characteristics during renal development. However, the function of Tmem100 in mouse embryonic kidney-derived cells remained unclear. We provided qPCR to verify the relationship between Tmem100 and the BMP signal pathway. To clarify the role of Tmem100 in cell proliferation and apoptosis, we carry out EdU incorporation, annexin V- fluorescein isothiocyanate (FITC) apoptosis assay. Here, we find that the knockdown of Tmem100 increases the proliferation and apoptosis of mouse embryonic kidney-derived cells, and this promotion can be inhibited by knockdown of BMP7 at the same time; these results suggest that BMP7 plays a crucial role in Tmem100-regulated cell proliferation and apoptosis. qRT-PCR results further demonstrate that the deficiency of Tmem100 leads to BMP7 upregulation and overexpression could get opposite results. In BMP7-depleted MK3 cells, Tmem100 is highly upregulated and BMPR-II is downregulated. And in BMP7-overexpressed MK3 cells, the expression of Tmem100 is decreased. In BMPR-II-depleted MK3 cells, Tmem100 is downregulated and BMP7 expression remains still. These findings indicate that both BMP7 and BMPR-II can regulate Tmem100 and vice versa, and BMPR-II expression is regulated by BMP7. However, BMP7 has no association with BMPR-II in MK3 cells. Our data demonstrated the significant role of BMP7 in Tmem100-regulated cell proliferation and apoptosis and revealed the complicated regulation network among Tmem100, BMP7, and BMPR-II in mouse embryonic kidney-derived cells.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Kidney/cytology , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Animals , Apoptosis/genetics , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cell Line , Cell Proliferation/genetics , Gene Knockdown Techniques , Kidney/embryology , Membrane Proteins/genetics , Mesenchymal Stem Cells/physiology , Mice
2.
Gene Expr Patterns ; 22(2): 37-45, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27826126

ABSTRACT

Tetratricopeptide repeat domain 36 (Ttc36), whose coding protein belongs to tetratricopeptide repeat (TPR) motif family, has not been studied extensively. We for the first time showed that Ttc36 is evolutionarily conserved across mammals by bioinformatics. Rabbit anti-mouse Ttc36 polyclonal antibody was generated by injecting synthetic full-length peptides through "antigen intersection" strategy. Subsequently, we characterized Ttc36 expression profile in mouse, showing its expression in liver and kidney both from embryonic day 15.5 (E15.5) until adult, as well as in testis. Immunofluorescence staining showed that Ttc36 is diffusely expressed in liver, however, specifically in kidney cortex. Thus, we further compare Ttc36 with proximal tubules (PT) marker Lotus Tetragonolobus Lectin (LTL) and distal tubules (DT) marker Calbindin-D28k respectively by double immunofluorescence staining. Results showed the co-localization of Ttc36 with LTL rather than Calbindin-D28k. Collectively, on the basis of the expression pattern, Ttc36 is specifically expressed in proximal distal tubules.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Protein Domains/genetics , Amino Acid Motifs/genetics , Animals , Calbindin 1/biosynthesis , Calbindin 1/genetics , Carrier Proteins/biosynthesis , Kidney Tubules, Proximal/metabolism , Lectins/biosynthesis , Lectins/genetics , Mice , Rabbits , Repetitive Sequences, Amino Acid/genetics
3.
Mol Med Rep ; 14(5): 4315-4320, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27667021

ABSTRACT

To seek out the potential microRNAs (miRNAs) that target Wilms' tumor suppressor 1 (WT1), a transcription factor required for progenitor proliferation as well as normal development of the kidney, and to clarify the effects of the miRNAs on WT1, the 3'-untranslated region (3'­UTR) of WT1 was initially analyzed and miR­743a, a seldom­reported miRNA, was identified. In the present paper, luciferase reporter assays were performed to confirm that miR­743a is able to directly target the 3'­UTR of WT1. Subsequently, reverse transcription­quantitative polymerase chain reaction, combined with western blotting analyses, were performed, and the results revealed a significant inhibition of WT1 at the mRNA and the protein levels. Furthermore, a 5­ethynyl­2'­deoxyuridine (EdU) cell proliferation assay, coupled with a WT1 rescue strategy, demonstrated that miR­743a inhibited the proliferation of metanephric mesenchymal (MM) cells, in part by targeting WT1. In conclusion, by targeting WT1, miR­743a suppresses the proliferation of MM cells in vitro, and probably possesses vital functions in kidney development and kidney­associated diseases.


Subject(s)
Kidney/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Repressor Proteins/biosynthesis , 3' Untranslated Regions , Animals , Cell Line , Cell Proliferation/genetics , Humans , Kidney/growth & development , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , MicroRNAs/biosynthesis , RNA, Messenger/biosynthesis , Repressor Proteins/genetics , WT1 Proteins
4.
Int J Mol Sci ; 17(9)2016 Sep 08.
Article in English | MEDLINE | ID: mdl-27618015

ABSTRACT

The metanephric mesenchyme (MM) cells are a subset of kidney progenitor cells and play an essential role in mesenchymal-epithelial transition (MET), the key step of nephron generation. Six2, a biological marker related to Wnt signaling pathway, promotes the proliferation, inhibits the apoptosis and maintains the un-differentiation of MM cells. Besides, LiCl is an activator of Wnt signaling pathway. However, the role of LiCl in cellular regulation of MM cells remains unclear, and the relationship between LiCl and Six2 in this process is also little known. Here, we performed EdU assay and flow cytometry assay to, respectively, detect the proliferation and apoptosis of MM cells treated with LiCl of increasing dosages. In addition, reverse transcription-PCR (RT-PCR) and Western-blot were conducted to measure the expression of Six2 and some maker genes of Wnt and bone-morphogenetic-protein (BMP) signaling pathway. Furthermore, luciferase assay was also carried out to detect the transcriptional regulation of Six2. Then we found LiCl promoted MM cell proliferation at low-concentration (10, 20, 30, and 40 mM). The expression of Six2 was dose-dependently increased in low-concentration (10, 20, 30, and 40 mM) at both mRNA and protein level. In addition, both of cell proliferation and Six2 expression in MM cells declined when dosage reached high-concentration (50 mM). However, Six2 knock-down converted the proliferation reduction at 50 mM. Furthermore, Six2 deficiency increased the apoptosis of MM cells, compared with negative control cells at relative LiCl concentration. However, the abnormal rise of apoptosis at 30 mM of LiCl concentration implies that it might be the reduction of GSK3ß that increased cell apoptosis. Together, these demonstrate that LiCl can induce the proliferation and apoptosis of MM cells coordinating with Six2.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Homeodomain Proteins/metabolism , Lithium Chloride/pharmacology , Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Mice , Transcription Factors/genetics , Wnt Signaling Pathway
5.
Int J Mol Sci ; 17(8)2016 Aug 06.
Article in English | MEDLINE | ID: mdl-27509493

ABSTRACT

Nephron progenitor cells surround around the ureteric bud tips (UB) and inductively interact with the UB to originate nephrons, the basic units of renal function. This process is determined by the internal balance between self-renewal and consumption of the nephron progenitor cells, which is depending on the complicated regulation networks. It has been reported that Zeb1 regulates the proliferation of mesenchymal cells in mouse embryos. However, the role of Zeb1 in nephrons generation is not clear, especially in metanephric mesenchyme (MM). Here, we detected cell proliferation, apoptosis and migration in MM cells by EdU assay, flow cytometry assay and wound healing assay, respectively. Meanwhile, Western and RT-PCR were used to measure the expression level of Zeb1 and Six2 in MM cells and developing kidney. Besides, the dual-luciferase assay was conducted to study the molecular relationship between Zeb1 and Six2. We found that knock-down of Zeb1 decreased cell proliferation, migration and promoted cell apoptosis in MM cells and Zeb1 overexpression leaded to the opposite data. Western-blot and RT-PCR results showed that knock-down of Zeb1 decreased the expression of Six2 in MM cells and Zeb1 overexpression contributed to the opposite results. Similarly, Zeb1 promoted Six2 promoter reporter activity in luciferase assays. However, double knock-down of Zeb1 and Six2 did not enhance the apoptosis of MM cells compared with control cells. Nevertheless, double silence of Zeb1 and Six2 repressed cell proliferation. In addition, we also found that Zeb1 and Six2 had an identical pattern in distinct developing phases of embryonic kidney. These results indicated that there may exist a complicated regulation network between Six2 and Zeb1. Together, we demonstrate Zeb1 promotes proliferation and apoptosis and inhibits the migration of MM cells, in association with Six2.


Subject(s)
Apoptosis , Cell Movement , Cell Proliferation , Homeodomain Proteins/genetics , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/physiology , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Conserved Sequence , Gene Expression , Gene Expression Regulation, Developmental , HEK293 Cells , Homeodomain Proteins/metabolism , Humans , Kidney/growth & development , Mesoderm/cytology , Mice , Promoter Regions, Genetic , Protein Binding , Transcription Factors/metabolism , Transcriptional Activation
6.
Int J Mol Sci ; 17(6)2016 May 25.
Article in English | MEDLINE | ID: mdl-27231908

ABSTRACT

Apobec-1 complementation factor (A1CF) is a heterogeneous nuclear ribonuceloprotein (hnRNP) and mediates apolipoprotein-B mRNA editing. A1CF can promote the regeneration of the liver by post-transcriptionally stabilizing Interleukin-6 (IL-6) mRNA. It also contains two transcriptional variants-A1CF64 and A1CF65, distinguished by the appearance of a 24-nucleotide motif which contributes to the corresponding eight-amino acid motif of EIYMNVPV. For the first time, we demonstrated that the EIYMNVPV motif was essential for A1CF nucleus localization, A1CF deficient of the EIYMNVPV motif, A1CF (-8aa) showed cytoplasm distribution. More importantly, we found that A1CF (-8aa), but not its full-length counterpart, can promote proliferation of MDA-MB-231 cells accompanied with increased level of IL-6 mRNA. Furthermore, silencing of IL-6 attenuated A1CF (-8aa)-induced proliferation in MDA-MB-231 cells. In conclusion, notably, these findings suggest that A1CF (-8aa) promoted proliferation of MDA-MB-231 cells in vitro viewing IL-6 as a target. Thus, the EIYMNVPV motif could be developed as a potential target for basal-like breast cancer therapy.


Subject(s)
Cell Nucleus/metabolism , Interleukin-6/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Up-Regulation , Amino Acid Motifs , Animals , Cell Line, Tumor , Cell Proliferation , Cytoplasm/metabolism , Dogs , Humans , Madin Darby Canine Kidney Cells , RNA-Binding Proteins/genetics
7.
Int J Mol Sci ; 17(2)2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26848653

ABSTRACT

Apobec-1 complementation factor (A1CF) is a member of the heterogeneous nuclear ribonucleoproteins (hnRNP) family, which participates in site-specific posttranscriptional RNA editing of apolipoprotein B (apoB) transcript. The posttranscriptional editing of apoB mRNA by A1CF in the small intestine is required for lipid absorption. Apart from the intestine, A1CF mRNA is also reported to be highly expressed in the kidneys. However, it is remained unknown about the functions of A1CF in the kidneys. The aim of this paper is to explore the potential functions of A1CF in the kidneys. Our results demonstrated that in C57BL/6 mice A1CF was weakly expressed in embryonic kidneys from E15.5dpc while strongly expressed in mature kidneys after birth, and it mainly existed in the tubules of inner cortex. More importantly, we identified A1CF negatively regulated the process of epithelial-mesenchymal transition (EMT) in kidney tubular epithelial cells. Our results found ectopic expression of A1CF up-regulated the epithelial markers E-cadherin, and down-regulated the mesenchymal markers vimentin and α-smooth muscle actin (α-SMA) in NRK52e cells. In addition, knockdown of A1CF enhanced EMT contrary to the overexpression effect. Notably, the two A1CF variants led to the similar trend in the EMT process. Taken together, these data suggest that A1CF may be an antagonistic factor to the EMT process of kidney tubular epithelial cells.


Subject(s)
Cell Movement , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Kidney Tubules, Proximal/metabolism , Actins/genetics , Actins/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line , Epithelial Cells/physiology , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/growth & development , Mice , Mice, Inbred C57BL , Rats
8.
Int J Mol Sci ; 16(11): 27945-55, 2015 Nov 24.
Article in English | MEDLINE | ID: mdl-26610487

ABSTRACT

Accumulating evidence demonstrated that miRNAs are highly involved in kidney fibrosis and Epithelial-Eesenchymal Transition (EMT), however, the mechanisms of miRNAs in kidney fibrosis are poorly understood. In this work, we identified that miR542-3p could promote EMT through down-regulating bone morphogenetic protein 7 (BMP7) expression by targeting BMP7 3'UTR. Firstly, real-time PCR results showed that miR542-3p was significantly up-regulated in kidney fibrosis in vitro and in vivo. Moreover, Western blot results demonstrated that miR542-3p may promote EMT in the NRK52e cell line. In addition, we confirmed that BMP7, which played a crucial role in anti-kidney fibrosis and suppressed the progression of EMT, was a target of miR542-3p through Dual-Luciferase reporter assay, as did Western blot analysis. The effects of miR542-3p on regulating EMT could also be suppressed by transiently overexpressing BMP7 in NRK52e cells. Taken together, miR542-3p may be a critical mediator of the induction of EMT via directly targeting BMP7.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , RNA Interference , Animals , Binding Sites , Bone Morphogenetic Protein 7/chemistry , Cell Line , Disease Models, Animal , Fibrosis , Gene Expression Regulation , Humans , Kidney Diseases/genetics , Kidney Diseases/pathology , Mice , MicroRNAs/chemistry , RNA, Messenger/chemistry , RNA, Messenger/genetics , Transforming Growth Factor beta1/metabolism
9.
Int J Mol Sci ; 16(10): 24032-47, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26473838

ABSTRACT

MicroRNAs (miRNAs) possess an important regulating effect among numerous renal diseases, while their functions in the process of epithelial-to-mesenchymal transition (EMT) after podocyte injury remain unclear. The purpose of our study is to identify the potential functions of miR-30a in EMT of podocytes and explore the underlying mechanisms of miR-30a in the impaired podocytes. The results revealed that downregulation of miR-30a in podocyte injury animal models and patients, highly induced the mesenchymal markers of EMT including Collagen I, Fibronectin and Snail. Furthermore, overexpression of miR-30a enhances epithelial markers (E-cadherin) but diminished mesenchymal markers (Collagen I, Fibronectin and Snail) in podocytes. In addition, we established miR-30a target NFATc3, an important transcription factor of Non-canonical Wnt signaling pathway. More importantly, our findings demonstrated that the augmentation of miR-30a level in podocytes inhibits the nuclear translocation of NFATc3 to protect cytoskeleton disorder or rearrangement. In summary, we uncovered the protective function of miR30a targeting NFATc3 in the regulation of podocyte injury response to EMT.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , MicroRNAs/biosynthesis , NFATC Transcription Factors/biosynthesis , Podocytes/metabolism , Active Transport, Cell Nucleus/genetics , Animals , Cadherins/biosynthesis , Cell Line , Collagen Type I/biosynthesis , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Fibronectins/biosynthesis , Gene Expression Regulation, Neoplastic , Glomerulosclerosis, Focal Segmental/pathology , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , NFATC Transcription Factors/metabolism , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Wnt Signaling Pathway/physiology
SELECTION OF CITATIONS
SEARCH DETAIL