P–284 Changes in protein expression due to metformin treatment and hyperinsulinemia in a human endometrial cancer cell line

2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
C Lange ◽  
A Machad. Weber ◽  
R Schmidt ◽  
C Schroeder ◽  
T Strowitzki ◽  
...  

Abstract Study question The aim of the study was to identify new target proteins/pathways that are affected by metformin treatment in endometrial cancer cells in a proteomic approach. Summary answer The expression of 1,300 different proteins were investigated, of which 80 proteins with the most prominent changes were presented and some discussed in detail. What is known already The incidence of endometrial cancer (EC) has increased over the past years. Metabolic diseases such as obesity, type II diabetes mellitus (T2DM), and associated conditions (i.e. polycystic ovary syndrome (PCOS), insulin resistance) lead to elevated levels of circulating estrogens, which promote EC development and progression. Metformin, an insulin-sensitizing biguanide drug, commonly used in the treatment of T2DM, especially in obese patients, displayed anti-cancer effects in various cancer types, including EC. Different proteins and pathways have been suggested as potential targets, but the underlying mechanism of action of metformin’s anti-cancer activity is still not completely understood. Study design, size, duration In the present in vitro study, EC cells were cultured in 5.5 mmol/L glucose medium (supplemented with 10 nmol/L ß-estradiol (E2)) and treated with metformin (0.5 mmol/L), insulin (100 ng/mL), or remained untreated for 7 d. The expression of 1,300 different proteins was detected in cellular extracts in an affinity proteomic approach and compared between the treatment groups in order to identify potential target proteins and pathways that contribute to the anti-cancer effects of metformin. Participants/materials, setting, methods The study was carried out with the EC cell line HEC–1A that represents a postmenopausal model with low E2 sensitivity. Proteins were extracted, quantified with the BCA assay, and protein expression was analyzed using the scioDiscover antibody microarray. Differences in protein abundance between samples were presented as log2-fold changes (log2FC) with significance for samples that displayed |log2FC| ≥ 0.5 and adjusted p ≤ 0.05. Pathway analysis was carried out with the STRING and DAVID databases. Main results and the role of chance The data revealed that metformin and insulin targeted similar pathways in the present study and mostly acted on proteins related to proliferation, migration and tumor immune response. These pathways may be affected in a tumor-promoting as well as a tumor-suppressing way by either metformin treatment or insulin supplementation. Results for the 80 most affected proteins were presented and the consequences for the cells resulting from the detected expression changes were discussed in detail for several proteins. The presented data helps identify potential target proteins and pathways affected by metformin treatment in EC and allows for a better understanding of the mechanism of action of the biguanide drug’s anti-cancer activity. However, further investigations are necessary to confirm the observations and conclusions drawn from the presented data after metformin administration, especially for proteins that were regulated in a favorable way, i.e. AKT3, CCND2, CD63, CD81, GFAP, IL5, IL17A, IRF4, PI3, and VTCN1. Further proteins might be of interest, where metformin counteracted unfavorable effects that have been induced by hyperinsulinemia. Limitations, reasons for caution The results were obtained from an in vitro study with human cancer cell lines, and thus cannot be easily extrapolated to patients. Wider implications of the findings: In the context of a hyperinsulinemic environment, further proteins might be of interest, i.e. AMFR, CCND2, CD63, ERBB3, EZR, GFAP, IRF4, PI3, PLCG2, SORL1, VEGFA, VTCN1, SPP1, and TM9SF2. Here, a metformin-induced insulin-sensitization might be able to counteract unfavorable effects on protein expression profile that have been induced by hyperinsulinemia. Trial registration number Not applicable

2018 ◽  
Vol 8 (1) ◽  
Author(s):  
Alexander J. Stevenson ◽  
Eleanor I. Ager ◽  
Martina A. Proctor ◽  
Dubravka Škalamera ◽  
Andrew Heaton ◽  
...  

PLoS ONE ◽  
2021 ◽  
Vol 16 (3) ◽  
pp. e0248103
Author(s):  
Carsten Lange ◽  
Amanda Machado Weber ◽  
Ronny Schmidt ◽  
Christoph Schroeder ◽  
Thomas Strowitzki ◽  
...  

The incidence of endometrial cancer (EC) has increased over the past years and mainly affects women above the age of 45 years. Metabolic diseases such as obesity and type II diabetes mellitus as well as associated conditions like polycystic ovary syndrome (PCOS), insulin resistance and hyperinsulinemia lead to elevated levels of circulating estrogens. Increased estrogen concentrations, in turn, further trigger the proliferation of endometrial cells and thus promote EC development and progression, especially in the absence of progesterone as seen in postmenopausal women. Elevated blood glucose levels in diabetic patients further contribute to the risk of EC development. Metformin is an insulin-sensitizing biguanide drug, commonly used in the treatment of type II diabetes mellitus, especially in obese patients. Besides its effects on glucose metabolism, metformin displayed anti-cancer effects in various cancer types, including EC. Direct anti-cancer effects of metformin target signaling pathways that are involved in cellular growth and proliferation, e.g. the AKT/PKB/mTOR pathway. Further proteins and pathways have been suggested as potential targets, but the underlying mechanism of action of metformin’s anti-cancer activity is still not completely understood. In the present study, the effects of metformin on protein expression were investigated in the human EC cell line HEC-1A using an affinity proteomic approach. Cells were treated with 0.5 mmol/L metformin over a period of 7 days and changes in the expression pattern of 1,300 different proteins were compared to the expression in untreated control cells as well as insulin-treated cells. Insulin treatment (100 ng/mL) was incorporated into the study in order to implement a model for insulin resistance and associated hyperinsulinemia, conditions that are often observed in obese and diabetic patients. Furthermore, the culture medium was supplemented with 10 nmol/L ß-estradiol (E2) during treatments to mimic increased estrogen levels, a common risk factor for EC development. Based on the most prominent and significant changes in expression, a set of 80 proteins was selected and subjected to a more detailed analysis. The data revealed that metformin and insulin targeted similar pathways in the present study and mostly acted on proteins related to proliferation, migration and tumor immune response. These pathways may be affected in a tumor-promoting as well as a tumor-suppressing way by either metformin treatment or insulin supplementation. The consequences for the cells resulting from the detected expression changes were discussed in detail for several proteins. The presented data helps identify potential targets affected by metformin treatment in EC and allows for a better understanding of the mechanism of action of the biguanide drug’s anti-cancer activity. However, further investigations are necessary to confirm the observations and conclusions drawn from the presented data after metformin administration, especially for proteins that were regulated in a favorable way, i.e. AKT3, CCND2, CD63, CD81, GFAP, IL5, IL17A, IRF4, PI3, and VTCN1. Further proteins might be of interest, where metformin counteracted unfavorable effects that have been induced by hyperinsulinemia.


Author(s):  
S. V. Kirthanashri ◽  
N. Ramesh Kumar ◽  
S. Chitra

The Garcinia mangostana Linn, a medicinal plant commonly used in Southeast Asia. The crude extract and isolated metabolites were used to evaluate its potential anti-cancer activity which was compared to methotrexate (MTX) in cervical cancer cell line. The present study involved in using crude mangosteen (CM), γ-mangostin (γ-M), isopropyl mangostin (IPM) and Di-O-methyl mangostin (DMM) against the standard anti-neoplastic drug MTX. Cell viability and cytotoxicity by lactate dehydrogenase (LDH) were assessed. Analysis of DNA fragmentation and clonogenic assay further supports the anti-cancer activity of the drugs in HeLa cells. The IC50 value for CM, γ-M, DMM, IPM and MTX were 13.4µg, 34.84µM, 15.57µM 5.3µM and 16.05µM respectively observed in the present study. This study suggests that the G. mangostana and its derivatives have potential anti-cancer activity an in vitro study in cervical cancer.


2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
T Thüner ◽  
C Lange ◽  
J Jauckus ◽  
T Strowitzki ◽  
A Germeyer

Abstract Study question Does metformin treatment lead to temporal expression changes of specific genes and proteins in endometrial cancer cell lines? Summary answer The expression of three different genes and proteins was investigated, of which all displayed changes over time. What is known already Endometrial cancer (EC) is one of the most common malignancies among postmenopausal women. A long-term estrogen effect on the endometrium often seen in women with obesity or the polycystic ovary syndrome (PCOS), as well as type II diabetes mellitus (T2DM) are well-known risk factors for the development and progression of EC. Metformin is a biguanide used in the treatment of T2DM patients and off label in women with PCOS. Moreover, metformin displays anti-tumor and anti-proliferative effects in various cancer types, including EC. In that regards BCL2L11, CDH1 and CDKN1A play an important role in apoptotic pathways, proliferation and invasion processes. Study design, size, duration The EC cells were cultured in normal (5.5 mmol/L) or high (17 mmol/L) glucose medium supplemented with 10 nmol/L ß-estradiol. The cells were treated with low dose metformin (1.0 mmol/L) for 2, 6, 24, 48 and 168 h (7 d). In addition, EC cells were treated with a combination of metformin and insulin (100 ng/mL) or remained untreated. Five independent experiments were fulfilled and untreated cells served as controls. Participants/materials, setting, methods The study was accomplished using two different human EC cell lines. HEC–1A represents an estrogen-independent EC, whereas Ishikawa represents the more common, estrogen-dependent EC. Proteins were extracted, quantified with a BCA assay, and the protein expression of BCL2L11, CDH1 and CDKN1A was analyzed by western blots. Furthermore, total RNA was extracted, transcribed to cDNA and Taqman real-time PCR was carried out to measure the expression of the associated genes, using fold change (FC) as parameter. Main results and the role of chance The expression of the selected genes, analyzed by RT-PCR, changed in both cell lines over time as follows: After 6 h, metformin induced a decrease in the expression of BCL2L11 (FC = 0.7) and CDH1 (FC = 0.75), whereas the expression of CDKN1A slightly increased (FC = 0.95–1.35). After 24 h, BCL2L11 expression increased in normal glucose groups (FC = 1.3, high glucose: FC = 0.93) and CDH1 expression decreased in combination with metformin and high glucose (FC = 0.7, normal glucose: FC = 1.1). CDKN1A expression was increased by metformin in both cell lines after 24 h (FC = 1.2–1.8). After 48 h of metformin treatment, expression for all three genes was only slightly changed (FC = 0.9–1.0). After 7 d it was observed that the combination of high glucose and metformin (i.e. like obese T2DM patients) led to an increased expression of BCL2L11, CDH1 and CDKN1A (FC = 1.4–2.9) in the presence and absence of insulin, whereas metformin induced a decreased expression of CDH1 and CDKN1A (FC = 0.5–0.75) in normal glucose medium. BCL2L11, CDH1 and CDKN1A expression was investigated at the protein level as well. Limitations, reasons for caution The results cannot be directly transferred to metformin treatment of patients, since the study was carried out in vitro. Additionally, further studies including more timepoints would indicate a more precisely gene and protein expression over time. Wider implications of the findings: This is the first in vitro study showing the temporal changes of BCL2L11, CDH1 and CDKN1A expression, genes related to tumorigenesis due to low dose metformin over time, suggesting differentially pathways in long term metformin treatment using physiologically achievable metformin levels. Trial registration number Not applicable


2009 ◽  
Vol 1209 ◽  
Author(s):  
Phong Anh Tran ◽  
Erik Taylor ◽  
Love Sarin ◽  
Robert H. Hurt ◽  
Thomas J Webster

AbstractTwo common problems with implantation after cancerous tumor resection are cancer recurrence and bacteria infection at the implant site. Tumor resection surgery sometimes can not remove all the cancerous cells, thus, cancer can return after implantation. In addition, bacteria infection is one of the leading causes of implant failure. Therefore, it is desirable to have anti-cancer and anti-bacterial molecules which both rapidly (for anti-infection purposes) and continuously (for anti-cancer purposes) are available at the implant site following implantation. Therefore, the objective of the present in vitro study was to create a multi-functional coating for anti-cancer and anti-bacterial orthopedic implant applications. Elemental selenium was chosen as the biologically active agent in this effort because of its known chemopreventive and anti-bacterial properties. To achieve that objective, titanium (Ti), a conventional orthopedic implant material was coated with selenium (Se) nanoclusters. Different coating densities were achieved by varying Se concentration in the reaction mixture. Titanium substrates coated with Se nanoclusters were shown to enhance healthy osteoblast (bone-forming cell) and inhibit cancerous osteoblast proliferation in co-culture experiments. Functions of S. epidermidis (one of the leading bacteria that infect implants) were inhibited on Ti coated with Se-nanoclusters compared to uncoated materials. Thus, this study provided for the first time a coating material (selenium nanoclusters) to the biomaterials’ community to promote healthy bone cells’ functions, inhibit cancer growth and prevent bacteria infection.


RSC Advances ◽  
2017 ◽  
Vol 7 (6) ◽  
pp. 3408-3412 ◽  
Author(s):  
Long Ma ◽  
Haiyan Liu ◽  
Lingpei Meng ◽  
Ping Qin ◽  
Botao Zhang ◽  
...  

Triterpenoidal saponins fraction isolated from a traditional Chinese medicine Conyza blinii H. Lév. demonstrates anti-cancer activity both in vitro and in vivo.


2021 ◽  
Vol 25 (11) ◽  
pp. 104-109
Author(s):  
Gullapelli Kumaraswamy ◽  
Ravichandar Maroju ◽  
Srinivas Bandari ◽  
Gouthami Dasari ◽  
Gullapelli Sadanandam

A novel series of 2-(1-((1-substitutedphenyl-1H-1,2,3- triazol-4-yl)methoxy)ethyl)-1-((1-substituted phenyl- 1H-1,2,3-triazol-4-yl)methyl)-1H-benzo[d]imidazole (3a-j)derivatives was synthesized in moderate to high yields. The structures of all the synthesized compounds were characterized by 1HNMR, 13CNMR and Mass spectroscopic methods. The title compounds were screened for their anti-oxidant activity and anti-cancer activity. The cancer activity results reveal that the compounds 3j, 3b and 3f are showing promising activity and remaining compounds exhibited moderate activity against all the tested cancer cell lines. The anti-oxidant activity also shows that the compounds 3c and 3d have shown excellent activity and remaining compounds were also found to exhibit moderate activity against the test organisms employed.


2021 ◽  
Vol 25 ◽  
Author(s):  
Evgenia S. Veligina ◽  
Nataliya V. Obernikhina ◽  
Stepan G. Pilyo ◽  
Oleksiy D. Kachkovsky ◽  
Volodymyr S. Brovarets

: Background: Synthesis of a series of 2-(dichloromethyl)pyrazolo[1,5- a][1,3,5]triazines was carried out and evaluated in vitro for their anticancer activity against a panel of 60 cell lines derived from nine cancer types. The joint quantum-chemical and experimental study of the influence of the extended πconjugated phenyl substituents on the electron structure of the pyrazolo[1,5-a][1,3,5]triazines as Pharmacophores were performed. It is shown that the decrease in the barriers to the rotation of phenyl substituents in compounds 1-7 possibly leads to an increase in the anti-cancer activity, which is in agreement with the change in the parameter biological affinity ϕ0. Analysis of the S0 → S1 electronic transitions (π→π*) of the pyrazolo[1,5-a][1,3,5]triazines shows that an increase in their intensity correlates with anti-cancer activity. Thus, the introduction of phenyl substituents increases the likelihood of investigated pyrazolo[1,5-a][1,3,5]triazines interacting with protein molecules (Biomolecule) by the π stacking mechanism. In both methyl and phenyl derivatives of pyrazolo[1,5-a][1,3,5]triazines, the second electronic transition includes the n-MO (the level of the lone electron pair in two-coordinated nitrogen atoms). The highest intensity of the η→π* electronic transition is observed in pyrazolo[1,5-a][1,3,5]triazine with pyridine residue, which does not exhibit anti-cancer activity, but exhibits antiviral activity [13]. It can be assumed that the possibility of the formation of [Pharmacophore-Biomolecule] complex by hydrogen bonding ([H-B]) mechanism with protein molecules increases.


Sign in / Sign up

Export Citation Format

Share Document