Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
J Endocrinol ; 261(2)2024 May 01.
Article in English | MEDLINE | ID: mdl-38513352

ABSTRACT

The impaired endometrial receptivity is a major factor contributing to infertility in patients with endometriosis (EM), but the underlying mechanism remains unclear. Our study aimed to investigate the role of Kruppel-like factor 15 (KLF15) in endometrial receptivity and its regulation in EM. We observed a significant decrease in KLF15 expression in the mid-secretory epithelial endometrial cells of EM patients compared to normal females without EM. To confirm the role of KLF15 in endometrial receptivity, we found a significantly reduced KLF15 expression and a significant decrease in embryo implantation number in the rat model via uterine horn infection with siRNA. This highlights the importance of KLF15 as a regulator receptivity. Furthermore, through ChIP-qPCR, we discovered that the progesterone receptor (PR) directly binds to KLF15 promoter regions, indicating that progesterone resistance may mediate the decrease in KLF15 expression in EM patients. Additionally, we found that the mid-secretory endometrium of EM patients exhibited impaired epithelial-mesenchymal transition (EMT). Knockdown of KLF15 upregulated E-cadherin and downregulated vimentin expression, leading to inhibited invasiveness and migration of Ishikawa cells. Overexpression KLF15 promotes EMT, invasiveness, and migration ability, and increases the attachment rate of JAR cells to Ishikawa cells. Through RNA-seq analysis, we identified TWIST2 as a downstream gene of KLF15. We confirmed that KLF15 directly binds to the promoter region of TWIST2 via ChIP-qPCR, promoting epithelial cell EMT during the establishment of endometrial receptivity. Our study reveals the involvement of KLF15 in the regulation of endometrial receptivity and its downstream effects on EMT. These findings provide valuable insights into potential therapeutic approaches for treating non-receptive endometrium in patients with EM.


Subject(s)
Endometriosis , Epithelial-Mesenchymal Transition , Kruppel-Like Transcription Factors , Animals , Female , Humans , Rats , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , Embryo Implantation/physiology , Endometriosis/genetics , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Epithelial Cells , Epithelial-Mesenchymal Transition/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism
2.
ACS Nano ; 18(11): 8229-8247, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38427686

ABSTRACT

Endothelial-mesenchymal transition (EndoMT) of vascular endothelial cells has recently been considered as a key player in the early progression of a variety of vascular and nonvascular diseases, including atherosclerosis, cancer, and organ fibrosis. However, current strategies attempting to identify pharmacological inhibitors to block the regulatory pathways of EndoMT suffer from poor selectivity, unwanted side effects, and a heterogeneous response from endothelial cells with different origins. Furthermore, EndoMT inhibitors focus on preventing EndoMT, leaving the endothelial cells that have already undergone EndoMT unresolved. Here, we report the design of a simple but powerful nanoparticle system (i.e., N-cadherin targeted melanin nanoparticles) to convert cytokine-activated, mesenchymal-like endothelial cells back to their original endothelial phenotype. We term this process "Reversed EndoMT" (R-EndoMT). R-EndoMT allows the impaired endothelial barriers to recover their quiescence and intactness, with significantly reduced leukocyte and cancer cell adhesion and transmigration, which could potentially stop atheromatous plaque formation and cancer metastasis in the early stages. R-EndoMT is achieved on different endothelial cell types originating from arteries, veins, and capillaries, independent of activating cytokines. We reveal that N-cadherin targeted melanin nanoparticles reverse EndoMT by downregulating an N-cadherin dependent RhoA activation pathway. Overall, this approach offers a different prospect to treat multiple EndoMT-associated diseases by designing nanoparticles to reverse the phenotypical transition of endothelial cells.


Subject(s)
Atherosclerosis , Neoplasms , Humans , Endothelial Cells/metabolism , Melanins , Endothelial-Mesenchymal Transition , Cadherins/metabolism , Cadherins/pharmacology , Cytokines/metabolism , Atherosclerosis/metabolism , Neoplasms/pathology
3.
Sci Total Environ ; 918: 170773, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38336054

ABSTRACT

Cadmium (Cd) exposure is known to enhance breast cancer (BC) progression. Cd promotes epithelial-mesenchymal transition (EMT) in BC cells, facilitating BC cell aggressiveness and invasion, but the underlying molecular mechanisms are unclear. Hence, transgenic MMTV-Erbb2 mice (6 weeks) were orally administered Cd (3.6 mg/L, approximately equal to 19.64 µΜ) for 23 weeks, and BC cells (BT474 cells) were exposed to Cd (0, 0.1, 1 or 10 µΜ) for 72 h to investigate the effect of Cd exposure on EMT in BC cells. Chronic Cd exposure dramatically expedited tumor metastasis to multiple organs; decreased E-cadherin density; and increased Vimentin, N-cadherin, ZEB1, and Twist density in the tumor tissues of MMTV-Erbb2 mice. Notably, transcriptomic analysis of BC tumors revealed cytochrome P450 1B1 (CYP1B1) as a key factor that regulates EMT progression in Cd-treated MMTV-Erbb2 mice. Moreover, Cd increased CYP1B1 expression in MMTV-Erbb2 mouse BC tumors and in BT474 cells, and CYP1B1 inhibition decreased Cd-induced BC cell malignancy and EMT in BT474 cells. Importantly, the promotion of EMT by CYP1B1 in Cd-treated BC cells was presumably controlled by glutamine metabolism. This study offers novel perspectives into the effect of environmental Cd exposure on driving BC progression and metastasis, and this study provides important guidance for comprehensively assessing the ecological and health risks of Cd.


Subject(s)
Cadmium , Neoplasms , Mice , Animals , Cadmium/pharmacology , Cell Line, Tumor , Glutamine/metabolism , Glutamine/pharmacology , Metabolic Reprogramming , Epithelial-Mesenchymal Transition , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology
4.
BMC Complement Med Ther ; 24(1): 68, 2024 Jan 31.
Article in English | MEDLINE | ID: mdl-38297301

ABSTRACT

BACKGROUND: Small cell lung cancer (SCLC) is the most malignant lung cancer type. Due to the high rates of metastasis and drug resistance, effective therapeutic strategies remain lacking. Tanshinone IIA (Tan IIA) has been reported to exhibit anti-tumor activity. Therefore, this study investigated the ability and underlying mechanism of Tan IIA to inhibit the metastasis and proliferation of SCLC. METHODS: H1688 and H446 cells were treated in vitro with Tan IIA (0, 1, 2 and 4 µM) or LY294002 (10 µM) for 24, 48, 72 h. H1688 and H446 cell migration was evaluated in wound healing and transwell migration assays. RNA-sequencing helped assess gene expression. BALB/c nude mice were injected with H1688 cells and treated with the Tan IIA group (10 mg/kg/day) or a control. Expression of E-cadherin, vimentin and PI3K/Akt signaling pathway proteins in tumors and H1688 was investigated by immunohistochemical analysis and western blot. RESULTS: Tan IIA inhibited H1688 and H446 cell proliferation without inducing apoptosis and suppressed H1688 and H446 cell migration. E-cadherin expression was increased, while vimentin expression was reduced after administration of Tan IIA. RNA-sequencing revealed that some genes related with the PI3K/Akt signaling pathway were altered using Tan IIA treatment. Furthermore, western blot helped detect PI3K and p-Akt expression was also reduced by Tan IIA treatment. Tan IIA inhibited tumor growth in vivo. Moreover, Tan IIA increased tumoral expression of E-cadherin accompanied by PI3K and p-Akt downregulation. CONCLUSION: Tan IIA suppresses SCLC proliferation and metastasis by inhibiting the PI3K/Akt signaling pathway, thereby highlighting the potential of Tan IIA as a new and relatively safe drug candidate to treat SCLC.


Subject(s)
Abietanes , Lung Neoplasms , Small Cell Lung Carcinoma , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Vimentin/metabolism , Small Cell Lung Carcinoma/drug therapy , Down-Regulation , Lung Neoplasms/drug therapy , Mice, Nude , Cell Proliferation , Cadherins/pharmacology , RNA/pharmacology
5.
Discov Med ; 36(180): 199-208, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38273760

ABSTRACT

BACKGROUND: Lenvatinib is an important molecular target drug for the treatment of advanced hepatocellular carcinoma (HCC). However, the application of molecular targeted therapies for HCC also faces some challenges. Cumulative evidence has also shown that curcumol is a potential anti-HCC drug. Curcumol can be used as a chemosensitizer to enhance the antitumor effect of chemotherapeutic drugs. The purpose of our study is to explore the effect of curcumol combined with lenvatinib on HCC. METHODS: The antitumor effects of curcumol or/and lenvatinib on Huh 7 cells of the HCC cell line were examined using the cell counting kit-8 (CCK-8) assay, colony formation assay, and transwell assay. For in vivo investigation, the effect on subcutaneous growth was also determined in nude mice. Changes in autophagy were determined by transmission electron microscope (TEM). Protein levels of apoptotic-related factors, epithelial mesenchymal transition (EMT)-related factors, autophagy factors, and N-cadherin and janus tyrosine kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3) were examined by Western blot. RESULTS: In this study, we found that curcumol or lenvatinib could promote HCC cell apoptosis in vitro and inhibit the growth of HCC tumors in vivo (curcumol or lenvatinib group compared with control group, p < 0.05). While combination with curcumol treatment could improve the effect of lenvatinib on promoting cell apoptosis of HCC in vitro and inhibiting the growth of HCC tumors in vivo (combination group compared with lenvatinib group, p < 0.05). Curcumol combined with lenvatinib could induce more autolysosome formation detected by TEM. Mechanically, curcumol or lenvatinib could increase the expression of Bcl-2-associated X protein (Bax), E-cadherin, UNC-51-like kinase 1 (ULK), and microtubule-associated protein 1 light chain 3 (LC3B) II/I, whereas it reduced the expression of B-cell lymphoma-2 (Bcl-2), JAK2/STAT3 (curcumol or lenvatinib group compared with control group, p < 0.05). Furthermore, combined with curcumol treatment could increase the expression of Bax, E-cadherin, ULK, and LC3B II/I, whereas it reduced the expression of Bcl-2, N-cadherin, and JAK2/STAT3 (combination group compared with lenvatinib group, p < 0.05). These findings suggest that curcumol enhances the antitumor effect of lenvatinib on hepatocellular carcinoma cells. CONCLUSION: Curcumol enhances the antitumor effect of lenvatinib on hepatocellular carcinoma cells.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Phenylurea Compounds , Quinolines , Sesquiterpenes , Mice , Animals , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Cell Line, Tumor , bcl-2-Associated X Protein/pharmacology , bcl-2-Associated X Protein/therapeutic use , Mice, Nude , Apoptosis , Cadherins/pharmacology , Cadherins/therapeutic use , Cell Proliferation
6.
J Cell Physiol ; 239(1): 212-226, 2024 01.
Article in English | MEDLINE | ID: mdl-38149479

ABSTRACT

Our study was conducted to investigate whether cadherin-5 (CDH5), a vascular endothelial cell adhesion glycoprotein, could facilitate the differentiation of human induced pluripotent stem cells (hiPSCs) into sinoatrial node-like pacemaker cells (SANLPCs), following previous findings of silk-fibroin hydrogel-induced direct conversion of quiescent cardiomyocytes into pacemaker cells in rats through the activation of CDH5. In this study, the differentiating hiPSCs were treated with CDH5 (40 ng/mL) between Day 5 and 7 during cardiomyocytes differentiation. The findings in the present study demonstrated that CDH5 stimulated the expression of pacemaker-specific markers while suppressing markers associated with working cardiomyocytes, resulting in an increased proportion of SANLPCs among hiPSCs-derived cardiomyocytes (hiPSC-CMs) population. Moreover, CDH5 induced typical electrophysiological characteristics resembling cardiac pacemaker cells in hiPSC-CMs. Further mechanistic investigations revealed that the enriched differentiation of hiPSCs into SANLPCs induced by CDH5 was partially reversed by iCRT14, an inhibitor of ß-catenin. Therefore, based on the aforementioned findings, it could be inferred that the regulation of ß-catenin by CDH5 played a crucial role in promoting the enriched differentiation of hiPSCs into SANLPCs, which presents a novel avenue for the construction of biological pacemakers in forthcoming research.


Subject(s)
Cadherins , Induced Pluripotent Stem Cells , Myocytes, Cardiac , beta Catenin , Animals , Humans , Rats , Antigens, CD , beta Catenin/metabolism , Cadherins/pharmacology , Cell Differentiation , Myocytes, Cardiac/metabolism , Sinoatrial Node
7.
Discov Med ; 35(179): 1035-1042, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38058068

ABSTRACT

BACKGROUND: Approximately 50% of hepatocellular carcinoma (HCC) arises due to the infection by hepatitis B virus X protein (HBx). Sorafenib, a unique targeted oral kinase inhibitor, is the therapeutic agent of choice for advanced HCC. The mechanism of HBx in drug resistance of sorafenib-resistant HCC cells was evaluated in this study. METHODS: Employing a stepwise increase of the sorafenib content, Hep3B and HepG2 cells were iteratively induced to establish drug-resistant cell lines (Hep3B/R and HepG2/R). The survival rate of Hep3B, Hep3B/R, HepG2, and HepG2/R cells was estimated using the cell counting kit-8 (CCK-8) assay. The IC50 values of sorafenib were calculated, exploring its effects under varying concentrations. The HBx content was quantified via quantitative reverse transcription PCR (RT-qPCR) and Western Blot. HBx overexpression and interfering virus vectors were constructed and transfected into Hep3B/R and HepG2/R cells. Cell viability and metastasis were assessed by colony formation, wound healing, and transwell assays. E-cadherin, N-cadherin, Vimentin, Slug, and Snail content was evaluated via Western Blot. RESULTS: HBx content was significantly elevated in Hep3B/R and HepG2/R subgroups compared to Hep3B and HepG2 subgroups. The proliferation, clonogenicity, invasiveness, and migratory abilities of Hep3B/R and HepG2/R cells in the HBx subgroup were markedly enhanced; E-cadherin content was significantly reduced, whereas the content of N-cadherin, Vimentin, Slug, and Snail was significantly elevated in the HBx subgroup. Conversely, in the sh-HBx subgroup, the proliferation, clonogenicity, invasion, and migration of Hep3B/R and HepG2/R cells were significantly reduced, E-cadherin content was markedly increased, and N-cadherin, Vimentin, Slug, and Snail content was significantly reduced, compared to the sh-negative control (NC) subgroup. CONCLUSIONS: HBx knockout may affect the development of HCC by reducing the proliferation, invasion, and migration of Hep3B/R and HepG2/R cells through the inhibition of Epithelial-Mesenchymal Transition (EMT).


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Sorafenib/pharmacology , Sorafenib/therapeutic use , Liver Neoplasms/genetics , Vimentin/metabolism , Vimentin/pharmacology , Vimentin/therapeutic use , Cell Line, Tumor , Hepatitis B virus , Drug Resistance , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , Cell Proliferation , Cell Movement , Gene Expression Regulation, Neoplastic
8.
J Physiol Pharmacol ; 74(4)2023 Aug.
Article in English | MEDLINE | ID: mdl-37865957

ABSTRACT

Cisplatin is the leading chemotherapy agent for advanced liver cancer. However, the resistance to cisplatin in liver cancer reduces its efficacy. A potential strategy to increase its effectiveness and reduce toxicity is to combine cisplatin with 1,3,8-trihydroxy-6-methylanthraquinone (emodin). In this study, we examined the effects of emodin combined with cisplatin on the invasion and migration of HepG2 cells and analyzed the role of emodin. The effects of cisplatin, emodin and their combination were assessed in HepG2 cells. Proliferation, invasion and migration of HepG2 cells were examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), scar and Transwell assays. The gelatinase spectrum and an ELISA detected the expression of matrix metallopeptidase 2 (MMP-2) and matrix metallopeptidase 9 (MMP-9). The expression of E-cadherin and vimentin was detected by immunofluorescence and Western blots. Emodin inhibited cell invasion and migration in HepG2 hepatoma cells, increased E-cadherin expression, decreased vimentin, MMP-2, and MMP-9 expression. The combination of emodin and cisplatin-induced a more significant effect in a dose-dependent manner. In this study, we found that emodin inhibited hepatocellular carcinoma (HCC) metastasis. Compared with either cisplatin or emodin alone, the combination of both showed a more significant synergistic effect. Emodin can enhance the sensitivity of HepG2 HCC cells to cisplatin by inhibiting epithelial-mesenchymal transition, and thus, play a role in preventing recurrence and metastasis in HCC.


Subject(s)
Carcinoma, Hepatocellular , Emodin , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Hep G2 Cells , Cisplatin/pharmacology , Cisplatin/therapeutic use , Emodin/pharmacology , Emodin/therapeutic use , Matrix Metalloproteinase 9 , Vimentin/pharmacology , Matrix Metalloproteinase 2 , Cell Line, Tumor , Cadherins/pharmacology , Cell Movement , Epithelial-Mesenchymal Transition , Cell Proliferation
9.
Clin Lab ; 69(8)2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37560863

ABSTRACT

BACKGROUND: Diabetic nephropathy (hereinafter referred to as DN) is one of the important causes of chronic renal failure, with great harm. We aimed to elucidate the role of transgelin-2, a key early detection for diabetic ne-phropathy. METHOD: The serum samples of 12 DN patients and 12 normal volunteers were collected for this experiment. Mice of the model group were injected intraperitoneally with streptozotocin following a high fat diet. Mouse podocyte (MPC5) cells were induced with 20 mmol/L d-glucose. RESULT: Transgelin-2 was highly expressed in DN patients with diabetic nephropathy both at the expression levels of mRNA and protein. Transgelin-2 expression was correlated with blood sugar in patients with DN. Transgelin-2 gene up-regulation enhanced inflammation and periostin levels, and reduced E-cadherin activity level in mice with DN. Over-expression of transgelin-2 increased inflammation and periostin levels, and reduced E-cadherin activity level in the in vitro model. Down-regulation of Transgelin-2 reduced inflammation and periostin levels and induced E-cadherin activity level in the in vitro model. Transgelin-2 induced ANXA2/ STAT3 signaling in a mouse model or an in vitro model. ANXA2 was one of the regulatory factors for the effects of transgelin-2 with inflammation, periostin, and E-cadherin in a model of DN. CONCLUSIONS: Taken together, these findings demonstrated that transgelin-2 promoted inflammation and periostin levels, and suppressed E-cadherin levels in DN by STAT3 signaling through ANXA2.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Podocytes , Mice , Animals , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , Signal Transduction , Inflammation/metabolism , Podocytes/metabolism
10.
Acta Biomater ; 168: 400-415, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37479156

ABSTRACT

After spinal cord injury (SCI), endogenous neural stem cells (NSCs) near the damaged site are activated, but few NSCs migrate to the injury epicenter and differentiate into neurons because of the harsh microenvironment. It has demonstrated that implantation of hydrogel scaffold loaded with multiple cues can enhance the function of endogenous NSCs. However, programming different cues on request remains a great challenge. Herein, a time-programmed linear hierarchical structure scaffold is developed for spinal cord injury recovery. The scaffold is obtained through coaxial 3D printing by encapsulating a dual-network hydrogel (composed of hyaluronic acid derivatives and N-cadherin modified sodium alginate, inner layer) into a temperature responsive gelatin/cellulose nanofiber hydrogel (Gel/CNF, outer layer). The reactive species scavenger, metalloporphyrin, loaded in the outer layer is released rapidly by the degradation of Gel/CNF, inhibiting the initial oxidative stress at lesion site to protect endogenous NSCs; while the inner hydrogel with appropriate mechanical support, linear topology structure and bioactive cues facilitates the migration and neuronal differentiation of NSCs at the later stage of SCI treatment, thereby promoting motor functional restorations in SCI rats. This study offers an innovative strategy for fabrication of multifunctional nerve regeneration scaffold, which has potential for clinical treatment of SCI. STATEMENT OF SIGNIFICANCE: Two major challenges facing the recovery from spinal cord injury (SCI) are the low viability of endogenous neural stem cells (NSCs) within the damaged microenvironment, as well as the difficulty of neuronal regeneration at the injured site. To address these issues, a spinal cord-like coaxial scaffold was fabricated with free radical scavenging agent metalloporphyrin Mn (III) tetrakis (4-benzoic acid) porphyrin and chemokine N-cadherin. The scaffold was constructed by 3D bioprinting for time-programmed protection and modulation of NSCs to effectively repair SCI. This 3D coaxially bioprinted biomimetic construct enables multi-factor on-demand repair and may be a promising therapeutic strategy for SCI.


Subject(s)
Metalloporphyrins , Spinal Cord Injuries , Spinal Cord Regeneration , Rats , Animals , Cell Differentiation , Hydrogels/pharmacology , Metalloporphyrins/pharmacology , Tissue Scaffolds/chemistry , Collagen/chemistry , Spinal Cord/pathology , Cadherins/pharmacology
11.
Endocr Relat Cancer ; 30(10)2023 10 01.
Article in English | MEDLINE | ID: mdl-37490874

ABSTRACT

Despite decades of research presenting insulin-like growth factor-1 receptor (IGF1R) as an attractive target for cancer therapy, IGF1R inhibitors ultimately failed in clinical trials. This was surprising due to the known cancer-promoting functions of IGF1R, including stimulation of cell invasion, proliferation, and survival. Discourse in the literature has acknowledged that a lack of patient stratification may have impacted the success of IGF1R-inhibitor trials. This argument alludes to the possibility that IGF1R function may be contingent on tumor type and cellular composition. Looking into the known roles of IGF1R, it becomes clear that this receptor interacts with a multitude of different proteins and even has tumor-suppressing functions. IGF1R is implicated in both cell-cell and cell-surface adhesion dynamics, and the effects of either IGF1R downregulation or pharmacological inhibition on cellular adhesion remain poorly understood. In turn, adhesion receptors modulate IGF1R signaling. In addition, our understanding of IGF1R function in tumor-associated immune and stromal cells is lacking, which could contribute to the overwhelming failure of IGF1R inhibitors in the clinic. In this review, we re-investigate clinical trial data to make connections between the failure of these drugs in human cancer patients and the understudied facets of IGF1R function. We describe lesser-known and potentially tumor-suppressive functions of IGF1R that include promoting cell-cell adhesion through E-cadherin, augmenting a pro-inflammatory macrophage phenotype, and stimulating B cells to produce immunoglobulins. We also highlight the important role of adhesion receptors in regulating IGF1R function, and we use this information to infer stratification criteria for selecting patients that might benefit from IGF1R inhibitors.


Subject(s)
Insulin-Like Growth Factor I , Neoplasms , Receptor, IGF Type 1 , Humans , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , Cell Line, Tumor , Cell Proliferation , Insulin-Like Growth Factor I/metabolism , Integrins , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Signal Transduction
12.
Theriogenology ; 209: 60-75, 2023 Oct 01.
Article in English | MEDLINE | ID: mdl-37356280

ABSTRACT

Histone methylation plays an essential role in oocyte growth and preimplantation embryonic development. The modification relies on histone methyl-transferases and demethylases, and one of these, lysine-specific demethylase 2a (Kdm2a), is responsible for modulating histone methylation during oocyte and early embryonic development. The mechanism of how Kdm2a deficiency disrupts early embryonic development and fertility remains elusive. To determine if maternally deposited Kdm2a is required for preimplantation embryonic development, the expression profile of Kdm2a during early embryos was detected via immunofluorescence staining and RT-qPCR. The Kdm2a gene in oocytes was specifically deleted with the Zp3-Cre/LoxP system and the effects of maternal Kdm2a loss were studied through a comprehensive range of female reproductive parameters including fertilization, embryo development, and the number of births. RNA transcriptome sequencing was performed to determine differential mRNA expression, and the interaction between Kdm2a and the PI3K/Akt pathway was studied with a specific inhibitor and activator. Our results revealed that Kdm2a was continuously expressed in preimplantation embryos and loss of maternal Kdm2a suppressed the morula-to-blastocyst transition, which may have been responsible for female subfertility. After the deletion of Kdm2a, the global H3K36me2 methylation in mutant embryos was markedly increased, but the expression of E-cadherin decreased significantly in morula embryos compared to controls. Mechanistically, RNA-seq analysis revealed that deficiency of maternal Kdm2a altered the mRNA expression profile, especially in the PI3K/Akt signaling pathway. Interestingly, the addition of a PI3K/Akt inhibitor (LY294002) to the culture medium blocked embryo development at the stage of morula; however, the developmental block caused by maternal Kdm2a loss was partially rescued with a PI3K/Akt activator (SC79). In summary, our results indicate that loss of Kdm2a influences the transcriptome profile and disrupts the PI3K/Akt signaling pathway during the development of preimplantation embryo. This can result in embryo block at the morula stage and female subfertility, which suggests that maternal Kdm2a is a potential partial redundancy with other genes encoding enzymes in the dynamics of early embryonic development. Our results provide further insight into the role of histone modification, especially on Kdm2a, in preimplantation embryonic development in mice.


Subject(s)
Infertility, Female , Animals , Female , Mice , Pregnancy , Blastocyst , Cadherins/metabolism , Cadherins/pharmacology , Embryonic Development , Gene Expression Regulation, Developmental , Histones/metabolism , Infertility, Female/veterinary , Morula , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction
13.
Acta Physiol (Oxf) ; 238(4): e14006, 2023 08.
Article in English | MEDLINE | ID: mdl-37243909

ABSTRACT

Regulation of cadherin-mediated cell adhesion is crucial not only for maintaining tissue integrity and barrier function in the endothelium and epithelium but also for electromechanical coupling within the myocardium. Therefore, loss of cadherin-mediated adhesion causes various disorders, including vascular inflammation and desmosome-related diseases such as the autoimmune blistering skin dermatosis pemphigus and arrhythmogenic cardiomyopathy. Mechanisms regulating cadherin-mediated binding contribute to the pathogenesis of diseases and may also be used as therapeutic targets. Over the last 30 years, cyclic adenosine 3',5'-monophosphate (cAMP) has emerged as one of the master regulators of cell adhesion in endothelium and, more recently, also in epithelial cells as well as in cardiomyocytes. A broad spectrum of experimental models from vascular physiology and cell biology applied by different generations of researchers provided evidence that not only cadherins of endothelial adherens junctions (AJ) but also desmosomal contacts in keratinocytes and the cardiomyocyte intercalated discs are central targets in this scenario. The molecular mechanisms involve protein kinase A- and exchange protein directly activated by cAMP-mediated regulation of Rho family GTPases and S665 phosphorylation of the AJ and desmosome adaptor protein plakoglobin. In line with this, phosphodiesterase 4 inhibitors such as apremilast have been proposed as a therapeutic strategy to stabilize cadherin-mediated adhesion in pemphigus and may also be effective to treat other disorders where cadherin-mediated binding is compromised.


Subject(s)
Pemphigus , Humans , Pemphigus/metabolism , Pemphigus/pathology , Desmosomes/metabolism , Cell Adhesion/physiology , Cadherins/metabolism , Cadherins/pharmacology , Myocardium/metabolism , Epithelium/metabolism , Endothelium/metabolism
14.
Funct Integr Genomics ; 23(2): 154, 2023 May 10.
Article in English | MEDLINE | ID: mdl-37162618

ABSTRACT

Kinesin family member 23 (KIF23) serves as a tumor-promoting gene with prognostic values in various tumors. However, the role of KIF23 in esophageal carcinoma (ESCA) progression is largely unknown. The overlapping differentially expressed genes (DEGs) in GSE12452, GSE17351, and GSE20347 datasets were identified via GEO2R tool and Venn diagram software. KIF23 expression was analyzed using GSE12452, GSE17351, and GSE20347 datasets, GEPIA database, and qRT-PCR. Cell proliferation was assessed by CCK-8 and EdU incorporation assays. Gene set enrichment analysis (GSEA) analysis was performed to investigate the pathways associated with the regulatory mechanisms of KIF23 in ESCA. The expression of E-cadherin, vimentin, N-cadherin, and matrix metalloproteinase-9 (MMP-9) and alternation of Wnt/ß-catenin pathway were detected by western blot analysis. We identified two overlapping upregulated DEGs, among which KIF23 was selected for subsequent experiments. KIF23 was overexpressed in ESCA samples and cells, and knockdown of KIF23 retarded cell proliferation in ESCA cells. Besides, KIF23 knockdown suppressed epithelial-mesenchymal transition (EMT) process in ESCA cells, as evidenced by the increase of E-cadherin expression and the reduction of vimentin, N-cadherin, and MMP-9 expression. GSEA analysis suggested that Wnt signaling pathway was the significant pathway related to KIF23. Moreover, we demonstrated that KIF23 silencing inhibited the Wnt/ß-catenin pathway in ESCA cells. Activation of Wnt/ß-catenin pathway by SKL2001 reversed the effects of KIF23 silencing on cell proliferation and EMT in ESCA cells. In conclusion, KIF23 knockdown inhibited the proliferation and EMT in ESCA cells through blockage of Wnt/ß-catenin pathway.


Subject(s)
Carcinoma , Esophageal Neoplasms , Humans , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , Carcinoma/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Esophageal Neoplasms/genetics , Family , Gene Expression Regulation, Neoplastic , Kinesins/genetics , Kinesins/metabolism , Kinesins/pharmacology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Vimentin/genetics , Vimentin/metabolism , Vimentin/pharmacology , Wnt Signaling Pathway/genetics
15.
Integr Cancer Ther ; 22: 15347354231172732, 2023.
Article in English | MEDLINE | ID: mdl-37157810

ABSTRACT

6-Shogaol from ginger has anti-inflammatory, anti-oxidation and anti-cancer effects. Aim of the Study: To study the effects and possible mechanisms of 6-Shogaol on inhibiting the migration of colon cancer cells Caco2 and HCT116 and prove the effects on proliferation and apoptosis. Materials and methods: The cells were treated with 6-Shogaol at the concentrations of 20, 40, 60, 80, and 100 µM, the cytotoxicity was tested by Colony formation assays and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), and the Western blot was used to evaluate IKKß/NF-κB/Snail pathway and EMT-related proteins. In addition, in order to eliminate the interference of proliferation inhibition on the experiment, Caco2 cells were treated with 6-Shogaol at the concentrations of 0, 40, and 80 µM, HCT116 cells were treated with 6-Shogaol at the concentrations of 0, 20, and 40 µM, apoptosis was measured by Annex V/PI staining, and migration was measured by Wound healing assays and Transwell test. Results: 6-Shogaol significantly inhibited the growth of cells. The maximum inhibitory concentration of half of them was 86.63 µM in Caco2 cells and 45.25 µM in HCT116 cells. At 80 µM and 40 µM concentrations, 6-Shogaol significantly promoted apoptosis of colon cancer Caco2 cells and HCT116 cells, and also significantly inhibited cell migration (P < .05). In addition, Western blot analysis showed that at 80 µM dose of 6-Shogaol significantly reduced MMP-2, N-cadherin, IKKß, P-NF-κB and Snail expression in Caco2 cells (P < .05). 40 µM dose of 6-Shogaol significantly reduced VEGF, IKKß, and P-NF-κB expression, and MMP-2, N-cadherin and Snail was significantly decreased at 60 µM of 6-Shogaol in HCT116 cells(P < .05). However, there was no significant change in E-cadherin in Caco2 cells, and the expression of E-cadherin protein in HCT116 cells decreased. Conclusion: This study proposes and confirms that 6-Shogaol can significantly inhibit the migration of colon cancer cells Caco2 and HCT116, and its mechanism may be produced by inhibiting EMT through IKKß/NF-κB/Snail signaling pathway. It was also confirmed that 6-Shogaol inhibited the proliferation and promoted apoptosis of Caco2 and HCT116 cells.


Subject(s)
Colonic Neoplasms , NF-kappa B , Humans , NF-kappa B/metabolism , I-kappa B Kinase/pharmacology , I-kappa B Kinase/therapeutic use , Matrix Metalloproteinase 2 , Caco-2 Cells , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Cadherins/metabolism , Cadherins/pharmacology , Cadherins/therapeutic use , Cell Movement , Cell Proliferation , Cell Line, Tumor , Epithelial-Mesenchymal Transition
16.
Perit Dial Int ; 43(6): 448-456, 2023 11.
Article in English | MEDLINE | ID: mdl-36998201

ABSTRACT

BACKGROUND: The roles of tight junction (TJ) proteins in peritoneal membrane transport and peritoneal dialysis (PD) require further characterisation. Dipeptidyl peptidase-4 is expressed in mesothelial cells, and its activity may affect peritoneal membrane function and morphology. METHODS: Human peritoneal mesothelial cells (HPMCs) were isolated and cultured from omentum obtained during abdominal surgery, and paracellular transport functions were evaluated by measuring transmesothelial electrical resistance (TMER) and dextran flux. Sprague-Dawley rats were infused daily with 4.25% peritoneal dialysate with and without sitagliptin administration for 8 weeks. At the end of this period, rat peritoneal mesothelial cells (RPMCs) were isolated to evaluate TJ protein expression. RESULTS: In HPMCs, the protein expression of claudin-1, claudin-15, occludin and E-cadherin was decreased by TGF-ß treatment but reversed by sitagliptin co-treatment. TMER was decreased by TGF-ß treatment but improved by sitagliptin co-treatment. Consistent with this, dextran flux was increased by TGF-ß treatment and reversed by sitagliptin co-treatment. In the animal experiment, sitagliptin-treated rats had a lower D2/D0 glucose ratio and a higher D2/P2 creatinine ratio than PD controls during the peritoneal equilibration test. Protein expression of claudin-1, claudin-15 and E-cadherin decreased in RPMCs from PD controls but was not affected in those from sitagliptin-treated rats. Peritoneal fibrosis was induced in PD controls but ameliorated in sitagliptin-treated rats. CONCLUSION: The expression of TJ proteins including claudin-1 and claudin-15 was associated with transport function both in HPMCs and in a rat model of PD. Sitagliptin prevents peritoneal fibrosis in PD and can potentially restore peritoneal mesothelial cell TJ proteins.


Subject(s)
Peritoneal Dialysis , Peritoneal Fibrosis , Humans , Rats , Animals , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/metabolism , Tight Junction Proteins/metabolism , Claudin-1/genetics , Claudin-1/metabolism , Dextrans/metabolism , Dextrans/pharmacology , Rats, Sprague-Dawley , Peritoneum/metabolism , Transforming Growth Factor beta/metabolism , Cadherins/metabolism , Cadherins/pharmacology
17.
J Complement Integr Med ; 20(2): 425-430, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-36480470

ABSTRACT

OBJECTIVES: To study the inhibitory effect of ß-elemene on invasion and metastasis of colorectal cancer cells and its possible mechanism. METHODS: Human colon cancer HCT116 cells were treated with different concentrations of ß-elemene. The proliferation inhibition rate of the cells was detected by MTT assay, cell migration rate was detected by scratched assay, and cell invasion rate was evaluated by Transwell cell invasion assay. The expressions of Vimentin, E-cadherin, N-cadherin, and ß-catenin were detected by Western blotting. The mRNA expressions of Vimentin, E-cadherin, N-cadherin, and ß-catenin were detected by real-time PCR. RESULTS: Compared with the control group, the expressions of migration rate, invasion rate, scratch healing rate, N-cadherin, and Vimentin protein of HCT116 cells were decreased after ß-elemene treatment, while the expression of E-cadherin protein was increased, and the inhibition rate of cell proliferation was increased (p<0.05). CONCLUSIONS: ß-Elemene may inhibit cell proliferation and invasion and metastasis by inhibiting EMT signaling pathway in human colon cancer cell line HCT116.


Subject(s)
Colonic Neoplasms , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , beta Catenin/pharmacology , Epithelial-Mesenchymal Transition/genetics , Vimentin/genetics , Vimentin/pharmacology , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , Colonic Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation
18.
Gut Liver ; 17(5): 766-776, 2023 09 15.
Article in English | MEDLINE | ID: mdl-36167345

ABSTRACT

Background/Aims: The purpose of the current study was to examine the anti-inflammatory effects of CKD-506, a novel histone deacetylase 6 inhibitor, on human peripheral blood mononuclear cells (PBMCs) and CD4+ T cells and to explore the relationship between CKD-506 and gut epithelial barrier function. Methods: Lipopolysaccharide-stimulated human PBMCs from inflammatory bowel disease (IBD) patients were treated with CKD-506, and tumor necrosis factor (TNF)-α expression was measured using an enzyme-linked immunosorbent assay. The proliferation of CD4+ T cells from IBD patients was evaluated using flow cytometric analysis. The effects of CKD-506 on gut barrier function in a cell line and colon organoids, based on examinations of mRNA production, goblet cell differentiation, and E-cadherin recovery, were investigated using quantitative reverse transcription polymerase chain reaction, immunofluorescence, and a fluorescein isothiocyanate-dextran permeability assay. Results: Secretion of TNF-α, a pivotal pro-inflammatory mediator in IBD, by lipopolysaccharide-triggered PBMCs was markedly decreased by CKD-506 treatment in a dose-dependent manner and to a greater extent than by tofacitinib or tubastatin A treatment. E-cadherin mRNA expression and goblet cell differentiation increased significantly and dose-dependently in HT-29 cells in response to CKD-506, and inhibition of E-cadherin loss after TNF-α stimulation was significantly reduced both in HT-29 cells and gut organoids. Caco-2 cells treated with CKD-506 showed a significant reduction in barrier permeability in a dose-dependent manner. Conclusions: The present study demonstrated that CKD-506 has anti-inflammatory effects on PBMCs and CD4 T cells and improves gut barrier function, suggesting its potential as a small-molecule therapeutic option for IBD.


Subject(s)
Inflammatory Bowel Diseases , Tumor Necrosis Factor-alpha , Humans , Caco-2 Cells , Histone Deacetylase 6/metabolism , Histone Deacetylase 6/pharmacology , Histone Deacetylase 6/therapeutic use , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Lipopolysaccharides/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Cadherins/metabolism , Cadherins/pharmacology , Cadherins/therapeutic use , RNA, Messenger/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use
19.
Zhonghua Nan Ke Xue ; 29(3): 210-217, 2023 Mar.
Article in Chinese | MEDLINE | ID: mdl-38597701

ABSTRACT

OBJECTIVE: To explore the effects of lutein on the adhesion, invasiveness and metastasis of human prostate cancer PC-3M cells and its action mechanism. METHODS: We divided human prostate cancer PC-3M cells into a control, a low-dose lutein, a medium-dose lutein and a high-dose lutein group, and treated them with 0, 10, 20 and 40 µmol/L lutein, respectively. Then we examined the adhesion of the cells to matrix by cell adhesion assay and the changes in cell pseudopodia by Phalloidin staining, detected the expressions of paxillin, matrix metalloproteinase 2 (MMP-2), MMP-9, recombinant tissue inhibitors of metalloproteinase 1 (TIMP-1), E-cadherin, N-cadherin and vimentin by Western blot, determined the invasiveness and migration of the cells by scratch and Transwell assays, and observed their dynamic movement by high-intension imaging. RESULTS: Compared with the control, the lutein intervention groups showed significant reduction in the number of the cells adhered to matrix, the number of cell pseudopodia, the expressions of paxillin, MMP-2, MMP-9, N-cadherin and vimentin, the rates of migration, invasion and metastasis, and the distances of displacement and movement of the cells. However, the expressions of TIMP-1 and epithelial-mesenchymal transition-related E-cadherin were upregulated significantly. CONCLUSION: Lutein can inhibit cell adhesion, reduce the expressions of MMPs, and suppress cell invasion and migration by inhibiting the process of epithelial-mesenchymal transition.


Subject(s)
Matrix Metalloproteinase 2 , Prostatic Neoplasms , Male , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/pharmacology , Paxillin/metabolism , Paxillin/pharmacology , Lutein/metabolism , Lutein/pharmacology , Lutein/therapeutic use , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Matrix Metalloproteinase 9/therapeutic use , Vimentin/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-1/therapeutic use , Cell Movement , Cell Line, Tumor , Cadherins/metabolism , Cadherins/pharmacology , Cadherins/therapeutic use , Prostatic Neoplasms/pathology , Neoplasm Invasiveness , Epithelial-Mesenchymal Transition
20.
Nanotoxicology ; 16(6-8): 695-712, 2022.
Article in English | MEDLINE | ID: mdl-36345150

ABSTRACT

We and others have previously demonstrated that exposure to nickel nanoparticles (Nano-Ni) caused fibrogenic and carcinogenic effects; however, the underlying mechanisms are still not fully understood. This study aimed to investigate the effects of Nano-Ni on epithelial-mesenchymal transition (EMT) in human bronchial epithelial cells (BEAS-2B) and its underlying mechanisms since EMT is involved in both cancer pathogenesis and tissue fibrosis. Our results showed that exposure to Nano-Ni, compared to the control Nano-TiO2, caused a remarkable decrease in the expression of E-cadherin and an increase in the expression of vimentin and α-SMA, indicating an inducible role of Nano-Ni in EMT development in human bronchial epithelial cells. HIF-1α nuclear accumulation, HDAC3 upregulation, and decreased histone acetylation were also observed in the cells exposed to Nano-Ni, but not in those exposed to Nano-TiO2. Pretreatment of the cells with a specific HIF-1α inhibitor, CAY10585, or HIF-1α-specific siRNA transfection prior to Nano-Ni exposure resulted in the restoration of E-cadherin and abolished Nano-Ni-induced upregulation of vimentin and α-SMA, suggesting a crucial role of HIF-1α in Nano-Ni-induced EMT development. CAY10585 pretreatment also attenuated the HDAC3 upregulation and increased histone acetylation. Inhibition of HDAC3 with specific siRNA significantly restrained Nano-Ni-induced reduction in histone acetylation and restored EMT-related protein expression to near control levels. In summary, our findings suggest that exposure to Nano-Ni promotes the development of EMT in human bronchial epithelial cells by decreasing histone acetylation through HIF-1α-mediated HDAC3 upregulation. Our findings may provide information for further understanding of the molecular mechanisms of Nano-Ni-induced fibrosis and carcinogenesis.


Subject(s)
Nanoparticles , Nickel , Humans , Nickel/toxicity , Nickel/metabolism , Vimentin/metabolism , Vimentin/pharmacology , Epithelial-Mesenchymal Transition/genetics , Histones , Epithelial Cells , Cadherins/genetics , Cadherins/metabolism , Cadherins/pharmacology , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Fibrosis , Hypoxia-Inducible Factor 1, alpha Subunit , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL