lovo cells
Recently Published Documents


TOTAL DOCUMENTS

143
(FIVE YEARS 38)

H-INDEX

22
(FIVE YEARS 3)

Polymers ◽  
2021 ◽  
Vol 14 (1) ◽  
pp. 108
Author(s):  
Bo-Xiang Wang ◽  
Jia Li ◽  
De-Hong Cheng ◽  
Yan-Hua Lu ◽  
Li Liu

Antheraea pernyi silk fibroin (ASF)-based nanofibers have wide potential for biomaterial applications due to superior biocompatibility. It is not clear whether the ASF-based nanofibers scaffold can be used as an in vitro cancer cell culture platform. In the current study, we fabricated novel ASF-based thermoresponsive hydrogel nanofibers by aqueous electrospinning for colon cancer (LoVo) cells culture. ASF was reacted with allyl glycidyl ether (AGE) for the preparation of allyl silk fibroin (ASF-AGE), which provided the possibility of copolymerization with allyl monomer. The investigation of ASF-AGE structure by 1H NMR revealed that reactive allyl groups were successfully linked with ASF. ASF-based thermoresponsive hydrogel nanofibers (p (ASF-AGE-NIPAAm)) were successfully manufactured by aqueous electrospinning with the polymerization of ASF and N-isopropylacrylamide (NIPAAm). The p (ASF-AGE-NIPAAm) spinning solution showed good spinnability with the increase of polymerization time, and uniform nanofibers were formed at the polymerization time of 360 min. The obtained hydrogel nanofibers exhibited good thermoresponsive that the LCST was similar with PNIPAAm at about 32 °C, and good degradability in protease XIV PBS solution. In addition, the cytocompatibility of colon cancer (LoVo) cells cultured in hydrogel nanofibers was assessed. It was demonstrated that LoVo cells grown on hydrogel nanofibers showed improved cell adhesion, proliferation, and viability than those on hydrogel. The results suggest that the p (ASF-AGE-NIPAAm) hydrogel nanofibers have potential application in LoVo cells culture in vitro. This study demonstrates the feasibility of fabricating ASF-based nanofibers to culture LoVo cancer cells that can potentially be used as an in vitro cancer cell culture platform.


2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Tiancheng Zhao ◽  
Yezhou Li ◽  
Kexin Shen ◽  
Quan Wang ◽  
Jiayu Zhang

AbstractChemoresistance is one of the major problems of colon cancer treatment. In tumors, glycolytic metabolism has been identified to promote cell proliferation and chemoresistance. However, the molecular mechanisms underlying glycolytic metabolism and chemoresistance in colon cancer remains enigmatic. Hence, this research was designed to explore the mechanism underlying the OLR1/c-MYC/SULT2B1 axis in the regulation of glycolytic metabolism, to affect colon cancer cell proliferation and chemoresistance. Colon cancer tissues and LoVo cells were attained, where OLR1, c-MYC, and SULT2B1 expression was detected by immunohistochemistry, RT-qPCR, and western blot analysis. Next, ectopic expression and knockdown assays were implemented in LoVo cells. Cell proliferation was detected by MTS assay and clone formation. Extracellular acidification, glucose uptake, lactate production, ATP/ADP ratio, and GLUT1 and LDHA expression were measured to evaluate glycolytic metabolism. Then, the transfected cells were treated with chemotherapeutic agents to assess drug resistance by MTS experiments and P-gp and SMAD4 expression by RT-qPCR. A nude mouse model of colon cancer transplantation was constructed for in vivo verification. The levels of OLR1, c-MYC, and SULT2B1 were upregulated in colon cancer tissues and cells. Mechanistically, OLR1 increased c-MYC expression to upregulate SULT2B1 in colon cancer cells. Moreover, knockdown of OLR1, c-MYC, or SULT2B1 weakened glycolytic metabolism, proliferation, and chemoresistance of colon cancer cells. In vivo experiments authenticated that OLR1 knockdown repressed the tumorigenesis and chemoresistance in nude mice by downregulating c-MYC and SULT2B1. Conclusively, knockdown of OLR1 might diminish SULT2B1 expression by downregulating c-MYC, thereby restraining glycolytic metabolism to inhibit colon cancer cell proliferation and chemoresistance.


Molecules ◽  
2021 ◽  
Vol 26 (19) ◽  
pp. 6056
Author(s):  
Joanna Wezgowiec ◽  
Marta Tsirigotis-Maniecka ◽  
Jolanta Saczko ◽  
Mieszko Wieckiewicz ◽  
Kazimiera A. Wilk

This study aimed to characterize the hydrogel micro- and macro-particles designed to deliver curcumin to human colon cancer cells (LoVo). Six series of vehicles based on sodium alginate (micro- and macro-particles, uncoated, coated with chitosan or gelatin) were synthesized. The uncoated microparticles were fabricated using an emulsion-based technique and the uncoated macroparticles with an extrusion technique, with both coupled with ionotropic gelation. The surface morphology of the particles was examined with scanning electron microscopy and the average size was measured. The encapsulation efficiency, moisture content, and swelling index were calculated. The release of curcumin from the particles was studied in an experiment simulating the conditions of the stomach, intestine, and colon. To evaluate the anticancer properties of such targeted drug delivery systems, the cytotoxicity of both curcumin-loaded and unloaded carriers to human colon cancer cells was assessed. The microparticles encapsulated much less of the payload than the macroparticles and released their content in a more prolonged manner. The unloaded carriers were not cytotoxic to LoVo cells, while the curcumin-loaded vehicles impaired their viability—more significantly after incubation with microparticles compared to macroparticles. Gelatin-coated or uncoated microparticles were the most promising carriers but their potential anticancer activity requires further thorough investigation.


2021 ◽  
Vol 22 (19) ◽  
pp. 10539
Author(s):  
Richard Jennemann ◽  
Martina Volz ◽  
Felix Bestvater ◽  
Claudia Schmidt ◽  
Karsten Richter ◽  
...  

Colorectal cancer (CRC) is one of the most frequently diagnosed cancers in humans. At early stages CRC is treated by surgery and at advanced stages combined with chemotherapy. We examined here the potential effect of glucosylceramide synthase (GCS)-inhibition on CRC biology. GCS is the rate-limiting enzyme in the glycosphingolipid (GSL)-biosynthesis pathway and overexpressed in many human tumors. We suppressed GSL-biosynthesis using the GCS inhibitor Genz-123346 (Genz), NB-DNJ (Miglustat) or by genetic targeting of the GCS-encoding gene UDP-glucose-ceramide-glucosyltransferase- (UGCG). GCS-inhibition or GSL-depletion led to a marked arrest of the cell cycle in Lovo cells. UGCG silencing strongly also inhibited tumor spheroid growth in Lovo cells and moderately in HCT116 cells. MS/MS analysis demonstrated markedly elevated levels of sphingomyelin (SM) and phosphatidylcholine (PC) that occurred in a Genz-concentration dependent manner. Ultrastructural analysis of Genz-treated cells indicated multi-lamellar lipid storage in vesicular compartments. In mice, Genz lowered the incidence of experimentally induced colorectal tumors and in particular the growth of colorectal adenomas. These results highlight the potential for GCS-based inhibition in the treatment of CRC.


2021 ◽  
Author(s):  
Chun Yang ◽  
Bo-Wen Chen ◽  
Kai-Wen Zhang ◽  
Si-Jia Chen ◽  
Xiao-Peng Zhang ◽  
...  

Abstract BackgroundAccumulating evidence indicates that sporamin, the main storage protein in the sweet potato (Ipomoea batatas), can suppress the development of colorectal cancer (CRC), but the changes in the gut microbiome after sporamin intervention and its relationships with the pathogenesis of CRC have not been investigated.MethodsTwelve male athymic BALB/c nude mice were randomly divided into four groups, CG1, CG2, TCG, and TTG. Mice in TCG and TTG were intraperitoneally transplanted with the LoVo cancer cells before the interventions started. CG2 and TTG were intragastrically infused with sporamin (0.5 g/kg BW/ day) for four weeks while CG1 and TCG were infused with the same volume of water during the experiment. Fecal samples were collected after the interventions and then examined for the changes in the microbiota using the 16S ribosomal RNA (rRNA) sequencing technology. The functional capabilities of the gut microbiota were predicted with the PICRUSt pipeline. Transcriptomic profiling of the tumor tissues was carried out for tumor-bearing mice with the RNA-sequencing (RNA-seq) technology and the resultant differentially expressed genes (DEGs) were then analyzed in terms of gene ontology (GO), protein-protein interaction (PPI), transcription factors (TF) prediction, and biological pathway annotations. ResultsSporamin significantly reduced the tumor burden of tumor-bearing mice and brought beneficial changes to the gut microbiome in both kinds of mice. It significantly increased the proportions of Barnesiella and Lactobacillus but reduced that of Bacteroides in tumor-bearing mice. The phenylalanine metabolism pathway, the glyoxylate, dicarboxylate metabolism, the bacterial secretion system, the glycan biosynthesis and metabolism, and the biosynthesis of stilbenoid, diarylheptanoid, and gingerol were favorably modulated by sporamin intervention. Sporamin mainly modulate the expression of the genes involved in the protein processing in the endoplasmic reticulum, the glycosylphosphatidylinositol (GPI)-anchor biosynthesis pathway, and the mineral absorption pathway. ConclusionSporamin could favorably alter the gut microbiome and its metabolome, improving the gut microenvironment and the viability of the gut microbiota and increasing the detoxification and bioactive substance production activities in the large intestine, by which the host’s metabolome may be altered and in turn exerts a suppressing effect on the protein synthesis and growth of tumor tissues.


2021 ◽  
Vol 65 (3) ◽  
Author(s):  
Zhi-bao Liu ◽  
Jing-hua Zhang ◽  
Jing-hua Gao ◽  
Jian Shi

The aim of this study was to explore the effects of the lncRNA ENST00000623984 on colorectal cancer. In this study, the expression levels of ENST000000623984 were first examined in tumor tissue and adjacent normal tissue from 40 patients with colorectal cancer and LoVo cells using quantitative real-time PCR. By siRNA transfection, ENST00000623984 expression was knocked down. Using flow cytometry, cell cycle progression and cell viability were examined in basal and knockdown LoVo cells. The CCK-8 assay was used to assess the cell proliferation rate, and the Transwell assay was used to determine the migration and invasion abilities. The ENST000000623984 expression level was increased in colorectal cancer. Knockdown of ENST000000623984 reduced cell viability, proliferation rate, cell migration and invasion. These results suggested that lncRNA ENST000000623984 may be involved in colorectal cancer development.


Pharmacology ◽  
2021 ◽  
pp. 1-10
Author(s):  
Yu-Xuan Wang ◽  
Cheng Lin ◽  
Lu-Jia Cui ◽  
Wan-He Yang ◽  
Qiu-Min Li ◽  
...  

<b><i>Background:</i></b> Colorectal cancer (CRC) is one of the most frequent digestive tract tumors in the world with an increasing incidence. Currently, surgical resection and chemotherapy are the main therapeutic options; however, their effects are limited by various adverse reactions. <i>Rauwolfia vomitoria</i> extract (Rau) has been shown to repress the progression of multiple human cancers; however, whether Rau plays a role in CRC remains undetermined. <b><i>Methods:</i></b> Influences of Rau treatment on HCT-116 and LoVo cells were estimated via MTT and colony formation experiments. Flow cytometry analysis was adopted to evaluate the apoptosis rate of HCT-116 and LoVo cells. Apoptosis-related proteins (Bcl-2, Bax, and caspase-3) and autophagy-related proteins (LC3 and P62) were assessed by Western blotting. Effects of Rau on autophagy of HCT-116 and LoVo cell were evaluated through GFP-LC3 analysis. In vivo xenograft tumor assay was conducted to further examine the role of Rau in CRC tumor growth. <b><i>Results:</i></b> Rau remarkably repressed HCT-116 and LoVo cell viability and promoted HCT-116 and LoVo cell apoptosis in vitro in a dose-dependent manner. Rau increased the expression of caspase-3 and Bax and decreased the expression of Bcl-2 in HCT-116 and LoVo cells. Moreover, Rau was demonstrated to decrease the LC3||/LC3| ratio and increase the level of P62 in HCT-116 and LoVo cells. In addition, we found that Rau repressed xenograft tumor growth and also repressed autophagy in vivo. <b><i>Conclusion:</i></b> Our findings revealed that Rau repressed CRC cell viability and autophagy in vitro and in vivo, suggesting that Rau might be a potent therapeutic agent of CRC.


2021 ◽  
pp. 096032712110214
Author(s):  
JY Lee ◽  
HM Lim ◽  
CM Lee ◽  
S-H Park ◽  
MJ Nam

Indole-3-carbinol (I3C) is a phytochemical that exhibits growth-inhibitory activity against various cancer cells. However, there are limited studies on the effects of I3C on colon cancer cells. In this study, the growth-inhibitory activity of I3C against the human colorectal carcinoma cell line (LoVo) was examined. The results of the 3-(4,5-dimethylthiazol-2yl)-2,5-diphenyltetrazolium bromide, colony formation, and cell counting assays revealed that I3C suppressed the proliferation of LoVo cells. Microscopy and wound-healing analyses revealed that I3C affected the morphology and inhibited the migration of LoVo cells, respectively. I3C induced apoptosis and DNA fragmentation as evidenced by the results of fluorescein isothiocyanate-conjugated annexin V staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling assay, respectively. Additionally, I3C arrested the cell cycle at the G0/G1 phase and enhanced the reactive oxygen species levels. Western blotting analysis revealed that treatment with I3C resulted in the activation of apoptotic proteins, such as poly(ADP-ribose) polymerase, caspase-3, caspase-7, caspase-9, Bax, Bim, and p53 in LoVo cells. These results indicate that I3C induces apoptosis in LoVo cells by upregulating p53, leading to the activation of Bax and caspases. Taken together, I3C exerts cytotoxic effects on LoVo cells by activating apoptosis.


Molecules ◽  
2021 ◽  
Vol 26 (11) ◽  
pp. 3121
Author(s):  
Nahed N. E. El-Sayed ◽  
Taghreed M. Al-Otaibi ◽  
Mona Alonazi ◽  
Vijay H. Masand ◽  
Assem Barakat ◽  
...  

The pathogenesis of colorectal cancer is a multifactorial process. Dysbiosis and the overexpression of COX‑2 and LDHA are important effectors in the initiation and development of the disease through chromosomal instability, PGE2 biosynthesis, and induction of the Warburg effect, respectively. Herein, we report the in vitro testing of some new quinoxalinone and quinazolinone Schiff’s bases as: antibacterial, COX‑2 and LDHA inhibitors, and anticolorectal agents on HCT-116 and LoVo cells. Moreover, molecular docking and SAR analyses were performed to identify the structural features contributing to the biological activities. Among the synthesized molecules, the most active cytotoxic agent, (6d) was also a COX-2 inhibitor. In silico ADMET studies predicted that (6d) would have high Caco-2 permeability, and %HIA (99.58%), with low BBB permeability, zero hepatotoxicity, and zero risk of sudden cardiac arrest, or mutagenicity. Further, (6d) is not a potential P-gp substrate, instead, it is a possible P-gpI and II inhibitor, therefore, it can prevent or reverse the multidrug resistance of the anticancer drugs. Collectively, (6d) can be considered as a promising lead suitable for further optimization to develop anti-CRC agents or glycoproteins inhibitors.


Sign in / Sign up

Export Citation Format

Share Document