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.
Exp Eye Res ; 236: 109654, 2023 11.
Article in English | MEDLINE | ID: mdl-37734427

ABSTRACT

SMAD3 downregulation is documented in transforming growth factor ß1 (TGF-ß1)-induced corneal fibroblasts differentiation to myofibroblasts ("fibroTOmyoDiff") or corneal wound healing. However, the exact regulatory mechanism of TGF-ß1/SMAD3 pathway in this context remains unclear. Here, we investigated the role and related mechanism of SMAD3 down-regulation in TGF-ß1-induced human corneal fibroTOmyoDiff. By detecting expression changes of SMAD family during this process, we demonstrated that SMAD3 protein expression was dramatically decreased in the process and the decrease occurred mainly in SMAD3 gene transcription. Furthermore, SMAD3 overexpression using lentivirus infection and knockdown using sgRNA lentivirus infection or siRNAs revealed that SMAD3 overexpression enhanced TGF-ß1-induced corneal fibroTOmyoDiff and vice versa. In addition, specific siRNAs and inhibitors targeting particular signaling pathway were used to figure out the intracellular signaling pathway regulating SMAD3, and the result showed that the decease of SMAD3 induced by TGF-ß1 stimulation in human corneal fibroblasts (HCFs) was strikingly prevented by SMAD4 knockdown or p38 signaling inhibitor SB203580 treatment. Collectively, these results demonstrate that, in TGF-ß1 induced corneal fibroTOmyoDiff, down-regulation of SMAD3 expression regulated by SMAD4 and p38 signaling pathways forms a negative feedback loop of TGFß signaling to avoid excessive activation of the signaling, which suggest that SMAD3 may be a key target for corneal fibrosis treatment.


Subject(s)
Lentivirus Infections , Transforming Growth Factor beta1 , Humans , Transforming Growth Factor beta1/metabolism , Myofibroblasts/metabolism , Smad3 Protein/metabolism , Feedback , RNA, Guide, CRISPR-Cas Systems , Cells, Cultured , Fibroblasts/metabolism , Lentivirus Infections/metabolism , Transforming Growth Factor beta/metabolism
2.
Langmuir ; 38(14): 4364-4370, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35360902

ABSTRACT

Poor solubility, low cellular uptake, and poor cell selectivity are the main obstacles hampering the therapeutic potential and clinic application of macromolecules. To overcome these limitations, here we propose a chemical modification strategy of macromolecules based on enzyme-instructed self-assembly (EISA). By using protoporphyrin IX (PpIX) and its metal complex Zn-PpIX as the modification objects, we demonstrated that the integration of enzymatic transformation and molecular self-assembly of macromolecules successfully improved the solubility of macromolecules, enhancing their intracellular uptake selectively against cancer cells. The proposed strategy is potentially applicable as a general tool for the development of macromolecule-based nanomedicine.


Subject(s)
Antineoplastic Agents , Antineoplastic Agents/chemistry , Nanomedicine
3.
Biochem Biophys Res Commun ; 518(2): 396-401, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31427083

ABSTRACT

Nuclear Dbf2-related kinase 2 (NDR2) is a highly conserved kinase that belongs to the NDR/LATS serine-threonine kinase family. NDR2 is involved in many cellular processes as a kinase or a scaffolding protein. As a known kinase, NDR2 requires self-phosphorylation and trans-phosphorylation to become fully active. However, beside phosphorylation, little is known about other posttranslational modifications of NDR2. In this study, we found that NDR2 can be specially acetylated at K463 in cells. In addition, SIRT1 acts as the major deacetylase for NDR2, while p300 and CBP function as specific acetyltransferases for NDR2. Interestingly, in SIRT1 deficient cells HDAC6 and HDAC1/2 can deacetylate NDR2, which provides a novel insight in deacetylation regulation. Our results demonstrate that NDR2 is a reversible acetylated kinase regulated by SIRT1 and p300/CBP.


Subject(s)
E1A-Associated p300 Protein/metabolism , Peptide Fragments/metabolism , Protein Serine-Threonine Kinases/metabolism , Sialoglycoproteins/metabolism , Sirtuin 1/metabolism , Acetylation , Cells, Cultured , HCT116 Cells , HEK293 Cells , Humans
4.
Nat Commun ; 10(1): 774, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755608

ABSTRACT

The original version of this Article contained an error in the Data Availability Statement. The accession code indicated '53V' and should have read '5X3V'. This has been corrected in both PDF and HTML versions of the Article.

5.
Nat Commun ; 9(1): 4468, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367038

ABSTRACT

The conversion of serine and glycine that is accomplished by serine hydroxymethyltransferase 2 (SHMT2) in mitochondria is significantly upregulated in various cancers to support cancer cell proliferation. In this study, we observed that SHMT2 is acetylated at K95 in colorectal cancer (CRC) cells. SIRT3, the major deacetylase in mitochondria, is responsible for SHMT2 deacetylation. SHMT2-K95-Ac disrupts its functional tetramer structure and inhibits its enzymatic activity. SHMT2-K95-Ac also promotes its degradation via the K63-ubiquitin-lysosome pathway in a glucose-dependent manner. TRIM21 acts as an E3 ubiquitin ligase for SHMT2. SHMT2-K95-Ac decreases CRC cell proliferation and tumor growth in vivo through attenuation of serine consumption and reduction in NADPH levels. Finally, SHMT2-K95-Ac is significantly decreased in human CRC samples and is inversely associated with increased SIRT3 expression, which is correlated with poorer postoperative overall survival. Our study reveals the unknown mechanism of SHMT2 regulation by acetylation which is involved in colorectal carcinogenesis.


Subject(s)
Carcinogenesis/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/physiopathology , Glycine Hydroxymethyltransferase/metabolism , Sirtuin 3/metabolism , Acetylation , Animals , Autophagy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line , Cell Proliferation , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Glycine Hydroxymethyltransferase/chemistry , Glycine Hydroxymethyltransferase/genetics , Humans , Male , Mice , Mice, Nude , Protein Multimerization , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Sirtuin 3/genetics , Survival Analysis , Ubiquitin-Protein Ligases/metabolism
6.
Proc Natl Acad Sci U S A ; 114(4): 687-692, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28069943

ABSTRACT

Tyrosyl-tRNA synthetase (TyrRS) is well known for its essential aminoacylation function in protein synthesis. Recently, TyrRS has been shown to translocate to the nucleus and protect against DNA damage due to oxidative stress. However, the mechanism of TyrRS nuclear localization has not yet been determined. Herein, we report that TyrRS becomes highly acetylated in response to oxidative stress, which promotes nuclear translocation. Moreover, p300/CBP-associated factor (PCAF), an acetyltransferase, and sirtuin 1 (SIRT1), a NAD+-dependent deacetylase, regulate the nuclear localization of TyrRS in an acetylation-dependent manner. Oxidative stress increases the level of PCAF and decreases the level of SIRT1 and deacetylase activity, all of which promote the nuclear translocation of hyperacetylated TyrRS. Furthermore, TyrRS is primarily acetylated on the K244 residue near the nuclear localization signal (NLS), and acetylation inhibits the aminoacylation activity of TyrRS. Molecular dynamics simulations have shown that the in silico acetylation of K244 induces conformational changes in TyrRS near the NLS, which may promote the nuclear translocation of acetylated TyrRS. Herein, we show that the acetylated K244 residue of TyrRS protects against DNA damage in mammalian cells and zebrafish by activating DNA repair genes downstream of transcription factor E2F1. Our study reveals a previously unknown mechanism by which acetylation regulates an aminoacyl-tRNA synthetase, thus affecting the repair pathways for damaged DNA.


Subject(s)
Cell Nucleus/metabolism , DNA Damage/physiology , Oxidative Stress/physiology , Tyrosine-tRNA Ligase/metabolism , Acetylation , Animals , Cell Line , Cell Line, Tumor , E2F1 Transcription Factor/metabolism , HEK293 Cells , HeLa Cells , Humans , Protein Transport/physiology , Signal Transduction/physiology , Sirtuin 1/metabolism , Zebrafish , p300-CBP Transcription Factors/metabolism
7.
Aging (Albany NY) ; 8(7): 1442-56, 2016 07.
Article in English | MEDLINE | ID: mdl-27405111

ABSTRACT

The reduction of DNA damage repair capacity in terminally differentiated cells may be involved in sensitivity to cancer chemotherapy drugs; however, the underlying molecular mechanism is still not fully understood. Herein, we evaluated the role of miR-638 in the regulation of DNA damage repair in terminally differentiated cells. Our results show that miR-638 expression was up-regulated during cellular terminal differentiation and involved in mediating DNA damage repair processes. Results from a luciferase reporting experiment show that structural maintenance of chromosomes (SMC)1A was a potential target of miR-638; this was verified by western blot assays during cell differentiation and DNA damage induction. Overexpression of miR-638 enhanced the sensitivity of cancer cells to cisplatin, thus reducing cell viability in response to chemotherapy drug treatment. Furthermore, miR-638 overexpression affected DNA damage repair processes by interfering with the recruitment of the DNA damage repair-related protein, γH2AX, to DNA break sites. These findings indicate that miR-638 might act as a sensitizer in cancer chemotherapy and accompany chemotherapy drugs to enhance chemotherapeutic efficacy and to improve the chance of recovery from cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Damage/physiology , DNA Repair/physiology , MicroRNAs/metabolism , Antineoplastic Agents/pharmacology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Line, Tumor , Cell Survival/drug effects , Chromosomal Proteins, Non-Histone/genetics , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Repair/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Granulocytes/drug effects , Granulocytes/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , Megakaryocytes/drug effects , Megakaryocytes/metabolism , MicroRNAs/genetics
8.
Int J Nanomedicine ; 10: 6435-44, 2015.
Article in English | MEDLINE | ID: mdl-26604740

ABSTRACT

Nanomaterials have an advantage in "personalized" therapy, which is the ultimate goal of tumor treatment. In order to investigate the potential ability of FePt nanoparticles (NPs) in the diagnosis and chemoradiotherapy treatment of malignant tumors, superparamagnetic, monodispersed FePt (~3 nm) alloy NPs were synthesized, using cysteamine as a capping agent. The NPs were characterized by means of X-ray diffraction; transmission electron microscopy, Physical Property Measurement System, and Fourier transform infrared spectroscopy. The cytotoxicity of FePt NPs on Vero cells was assessed using an MTT assay, and tumor cell proliferation inhibited by individual FePt NPs and FePt NPs combined with X-ray beams were also collected using MTT assays; HeLa human cancer cell lines were used as in vitro models. Further confirmation of the combined effect of FePt NPs and X-rays was verified using HeLa cells, after which, the cellular uptake of FePt NPs was captured by transmission electron microscopy. The results indicated that the growth of HeLa cells was significantly inhibited by FePt NPs in a concentration-dependent manner, and the growth was significantly more inhibited by FePt NPs combined with a series of X-ray beam doses; the individual NPs did not display any remarkable cytotoxicity on Vero cells at a concentration <250 µg/mL. Meanwhile, the FePt NPs showed negative/positive contrast enhancement for MRI/CT molecule imaging at the end of the study. Therefore, the combined results implied that FePt NPs might potentially serve as a promising nanoprobe for the integration of tumor diagnosis and chemoradiotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Iron/pharmacology , Metal Nanoparticles/chemistry , Platinum/pharmacology , X-Rays , Animals , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorocebus aethiops , Dose-Response Relationship, Radiation , Endocytosis/drug effects , HeLa Cells , Humans , Magnetic Resonance Imaging , Metal Nanoparticles/ultrastructure , Tomography, X-Ray Computed , Vero Cells , X-Ray Diffraction
9.
Cell Biosci ; 5: 56, 2015.
Article in English | MEDLINE | ID: mdl-26413265

ABSTRACT

BACKGROUND: MicroRNA-720 (miR-720), a nonclassical miRNA, is involved in the initiation and progression of several tumors. In our previous studies, miR-720 was shown to be significantly upregulated in cervical cancer tissues compared with normal cervical tissues. However, the precise biological functions of miR-720, and its molecular mechanisms of action, are still unknown. RESULTS: Microarray expression profiles, luciferase reporter assays, and western blot assays were used to validate Rab35 as a target gene of miR-720 in HEK293T and HeLa cells. The regulation of Rab35 expression by miR-720 was assessed using qRT-PCR and western blot assays, and the effects of exogenous miR-720 and Rab35 on cell migration were evaluated in vitro using Transwell(®) assay, wound healing assay, and real-time analyses in HeLa cells. The influences of exogenous miR-720 on cell proliferation were evaluated in vitro by the MTT assay in HeLa cells. In addition, expression of E-cadherin and vimentin associated with epithelial-mesenchymal transition were also assessed using western blot analyses after transfection of miR-720 mimics and Rab35 expression vectors. The results showed that the small GTPase, Rab35, is a direct functional target of miR-720 in cervical cancer HeLa cells. By targeting Rab35, overexpression of miR-720 resulted in a decrease in E-cadherin expression and an increase in vimentin expression and finally led to promotion of HeLa cell migration. Furthermore, reintroduction of Rab35 3'-UTR(-) markedly reversed the induction of cell migration in miR-720-expressing HeLa cells. CONCLUSIONS: The miR-720 promotes cell migration of HeLa cells by downregulating Rab35. The results show that miR-720 is a novel cell migration-associated gene in cervical cancer cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...