Abstract P102: Differences in Angiotensin-(1-7)/alamandine-mediated Signaling in Tumoral and Normal Cell Lines

Hypertension ◽  
2016 ◽  
Vol 68 (suppl_1) ◽  
Author(s):  
Walkyria O Sampaio ◽  
Marilene Luzia Oliveira ◽  
Felipe A Silva ◽  
Leticia B Silva ◽  
Gisele M Etelvino ◽  
...  

Previously studies have demonstrated that besides its actions in the cardiovascular system, Angiotensin-(1-7) also plays a role in inhibiting tumoral growth. The role of recently described Alamandine in this field is not clear. The signaling pathways underling anti-tumoral actions of these peptides are also poorly understood. Therefore, the aim of this study was to elucidate the modulatory effect of Ang-(1-7) and Alamandine in the PI3K cascade, a well-known signaling pathway described to be involved in proliferation and cancer. To achieve this goal, we stimulate human pancreatic and lung cancer cell lineage (Miapaca and A549), as well as a control cell lineage (VERO) with Ang-(1-7) and Alamandine. Through western blotting analysis, our data suggest that both Ang-(1-7) and Alamandine activate the phosphatase pTEN (dephosphorylation of S380/T382/T383) (48±4% after 24 hours Ang-(1-7) treatment in miapaca and A549 in comparison of non-treated cells, p<0.05) (60±5 and 48±4% of phosphorylation level after 24 hours Alamandine treatment in miapaca and A549, respectively, in comparison of non-treated cells, p<0.05), which dephosphorylates PI3K, inactivating this kinase. Furthermore, AKT phosphorylation is transient, followed by a significant dephosphorylation when compared to the non-treated cells (30±5% after 24 hours Ang-(1-7) treatment in miapaca in comparison of non-treated cells, p<0.05). Ang-(1-7) also inhibits a PTEN downstream effector kinase, mTOR through dephosphorylation of T246 (70±5% after 24 hours Ang-(1-7) treatment in miapaca in comparison of non-treated cells, p<0.05). These effects were not observed in control non-tumoral cells (VERO cells). As previously demonstrated with Ang-(1-7) stimulation, Alamandine also induces the FOXO1 activation and migration to the nucleus in A549 (122 ± 8 of A.U. of fluorescence at 4 hours after alamandine treatment vs 46±4 A.U. at control, p<0.001) and Miapaca cells (67 ± 5 of A.U. of fluorescence at 4 hours after alamandine treatment vs 16±2 A.U. at control, p<0.001). These results indicate that, in contrast to normal tissues, Ang-(1-7) and Alamandine decreases, through PTEN activation, PI3K/AKT pathway in tumoral cells.

2017 ◽  
Vol 23 (32) ◽  
pp. 4827-4841 ◽  
Author(s):  
Jack Tuszynski ◽  
Tatiana M. Tilli ◽  
Michael Levin

The activities of individual cells must be tightly coordinated in order to build and maintain complex 3- dimensional body structures during embryogenesis and regeneration. Thus, one way to view cancer is within systems biology as a network disorder affecting the ability of cells to properly interact with a morphodynamic field of instructive signals that keeps proliferation and migration orchestrated toward the anatomical needs of the host organism. One layer of this set of instructive microenvironmental cues is bioelectrical. Voltage gradients among all somatic cells (not just excitable nerve and muscle) control cell behavior, and the ionic coupling of cells into networks via electrochemical synapses allows them to implement tissue-level patterning decisions. These gradients have been increasingly implicated in the induction and suppression of tumorigenesis and metastasis, in the emerging links between developmental bioelectricity to the cancer problem. Consistent with the well-known role of neurotransmitter molecules in transducing electrical activity to downstream cascades in the brain, serotonergic signaling has likewise been implicated in cancer. Here, we review these recent data and propose new approaches for manipulating bioelectric and neurotransmitter pathways in cancer biology based on a bioelectric view of cancer. To support this methodology, we present new data on the effects of the SSRI Prozac and its analog (ZINC ID = ZINC06811610) on survival of both cancer (MCF7) and normal (MCF10A) breast cells exposed to these compounds. We found an IC50 concentration (25 µM for Prozac and 100 µM for the Prozac analog) at which these compounds inhibited tumor cell survival and proliferation. Additionally, at these concentrations, we did not observe alterations in a non-tumoral cell line. This constitutes a proof-of-concept demonstration for our hypothesis that the use of both existing and novel drugs as electroceuticals could serve as an alternative to highly toxic chemotherapy strategies replacing or augmenting them with less toxic alternatives. We believe this new approach forms an exciting roadmap for future biomedical advances.


2015 ◽  
Vol 37 (3) ◽  
pp. 1044-1054 ◽  
Author(s):  
Hong-tao Li ◽  
Hui Zhang ◽  
Yong Chen ◽  
Xian-fu Liu ◽  
Jun Qian

Background/Aims: Colorectal cancer (CRC) is one of the leading causes of cancer-related deaths globally, with many oncogenes and tumor suppressors involved. The miRNAs are small non-coding RNAs known to play a vital role in the pathogenesis of CRC. The miR-423-3p was reported to act as an oncogene; however, its role in CRC growth remains unknown. Methods: qPCR assay was used to detect miR-423-3p expression in CRC specimens. Cell proliferation assay and transwell assay were conducted to evaluate CRC cell proliferation and migration. Luciferase reporter assay was to identify the target gene of miR-423-3p. And tumorigenesis model was established to test the role of miR-423-3p in CRC development in vivo. Results: Here, we showed that miR-423-3p was significantly up regulated in CRC tissues and cells compared with normal tissues and cells. Overexpression of miR-423-3p promoted CRC cell proliferation via enhancing the G1/S transition phase of the cell cycle, while inhibition of miR-423-3p repressed cell growth. Further studies showed that p21Cip1/Waf1 mediated the function of miR-423-3p, and overexpression of p21Cip1/Waf1 reversed the augmented effect of miR-423-3p on cell proliferation. Importantly, all these data were validated in the tumorigenesis assay in vivo. Conclusions: In conclusion, our findings demonstrated a critical impact of miR-423-3p on CRC growth.


2013 ◽  
Vol 304 (6) ◽  
pp. G561-G567 ◽  
Author(s):  
Nhi Huynh ◽  
Mildred Yim ◽  
Jonathan Chernoff ◽  
Arthur Shulkes ◽  
Graham S. Baldwin ◽  
...  

Gastrins, including amidated (Gamide) and glycine-extended (Ggly) forms, function as growth factors for the gastrointestinal mucosa. The p-21-activated kinase 1 (PAK1) plays important roles in growth factor signaling networks that control cell motility, proliferation, differentiation, and transformation. PAK1, activated by both Gamide and Ggly, mediates gastrin-stimulated proliferation and migration, and activation of β-catenin, in gastric epithelial cells. The aim of this study was to investigate the role of PAK1 in the regulation by gastrin of proliferation in the normal colorectal mucosa in vivo. Mucosal proliferation was measured in PAK1 knockout (PAK1 KO) mice by immunohistochemistry. The expression of phosphorylated and unphosphorylated forms of the signaling molecules PAK1, extracellular signal-regulated kinase (ERK), and protein kinase B (AKT), and the expression of β-catenin and its downstream targets c-Myc and cyclin D1, were measured in gastrin knockout (Gas KO) and PAK1 KO mice by Western blotting. The expression and activation of PAK1 are decreased in Gas KO mice, and these decreases are associated with reduced activation of ERK, AKT, and β-catenin. Proliferation in the colorectal mucosa of PAK1 KO mice is reduced, and the reduction is associated with reduced activation of ERK, AKT, and β-catenin. In compensation, antral gastrin mRNA and serum gastrin concentrations are increased in PAK1 KO mice. These results indicate that PAK1 mediates the stimulation of colorectal proliferation by gastrins via multiple signaling pathways involving activation of ERK, AKT, and β-catenin.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3123-3123
Author(s):  
Natasha Malik ◽  
Karen Dunn ◽  
Owen Sansom ◽  
Alison M Michie

Abstract Mechanistic target of rapamycin (mTOR) functions within a complex signalling cascade, through its activity in two unique complexes mTORC1 and mTORC2, to promote a multitude of different cellular functions including autophagy, protein synthesis and survival. The exact role of these complexes during leukaemia initiation/maintenance remains to be elucidated. Here, using transgenic knockout (KO) mouse models, we determine the individual roles of mTORC1 (targeting raptor) and mTORC2 (targeting rictor) in normal haemopoiesis and in CLL initiation/maintenance. Our results demonstrate that mice carrying a targeted KO of raptor at the haemopoietic stem cell (HSC) stage (Vav-cre+Raptorfl/fl ) do not survive post birth. This is due to anaemia resulting from a significant decrease in Ter119+ population, a significant decrease in KLF1 and KLF2 gene expression, and a significant increase in the megakaryocyte-erythroid population (MEP), suggesting a block at the MEP stage in Vav-cre+Raptorfl/fl foetal liver. While mTORC1 plays a fundamental role in RBC development, we show that mTORC2 plays a role in RBC regulation, as Rictor-deficient HSPCs exhibit an increase in RBC colony formation ex vivo. Conditional KO (cKO) of Raptor (Mx1-cre+Raptorfl/fl) in adult mice results in splenomegaly accompanied by increased spleen organ cellularity. There is a significant decrease in the B cell lineage, with a block in B cell development at the Lin-Sca-1+CD117+ (LSK) stage in the BM. mTORC2, on the other hand regulates late B cell maintenance as indicated by a significant decrease in transitional B cells (T1/T2), marginal zone progenitor (MZP), and follicular 1 (Fol1) cells in Vav-cre+Rictorfl/fl mice compared to controls. To address the role of mTORC1 and mTORC2 in CLL initiation/maintenance in vitro, BM-derived haemopoietic progenitors isolated from control (Cre-), Raptor-deficient (Mx1-cre+Raptorfl/fl) or Rictor-deficient (Vav-cre+Rictorfl/fl) mice were retrovirally-transduced with a kinase dead PKCα (PKCα-KR) construct to induce an aggressive CLL-like disease. Raptor-deficient BM progenitors exhibited reduced proliferation and failed to generate a CLL-like disease, due to a block in B cell lineage commitment. However, there was an increase in cell cycling and migration in PKCα-KR CLL-like cells with Rictor- deficiency suggesting a role of mTORC2 in disease maintenance. To determine a role for mTORC1 in disease maintenance in vivo, NSG mice were transplanted with PKCα-KR-transduced BM-isolated from either Mx1-cre-Raptorfl/fl or Mx1-cre+Raptorfl/fl. Once disease was established in vivo, cKO was induced and disease load and progression was monitored. Our data indicate a significant decrease in disease load with Raptor cKO, together with a trend towards increased survival. Ongoing experiments with Mx1-cre+Rictorfl/fl mice will give us an insight into the role of mTORC2 in CLL. Taken together, mTORC1 plays an essential role in haemopoiesis, with Raptor-deficiency causing a block in RBC and B cell development at the MEP and LSK stage respectively. In comparison, Rictor-deficiency regulates later B cell lineages and promotes RBC colony formation, potentially through mTORC1 activation. Importantly, CLL-like cells lacking mTORC2 have increased cell cycling and migration whereas mTORC1 deficiency causes a decrease in disease load. Therefore, mTORC1 and mTORC2 play complementary roles in haemopoietic development and leukaemia initiation/progression. These studies provide a strong foundation for further studies testing clinical mTOR inhibitors for CLL in our models. Disclosures No relevant conflicts of interest to declare.


PLoS ONE ◽  
2014 ◽  
Vol 9 (10) ◽  
pp. e110119 ◽  
Author(s):  
Eric Bradley ◽  
Somsankar Dasgupta ◽  
Xue Jiang ◽  
Xiaying Zhao ◽  
Gu Zhu ◽  
...  

PLoS ONE ◽  
2021 ◽  
Vol 16 (3) ◽  
pp. e0247394
Author(s):  
Oleksi Petrenko ◽  
Jinyu Li ◽  
Velasco Cimica ◽  
Patricio Mena-Taboada ◽  
Ha Youn Shin ◽  
...  

The inflammatory cytokine IL-6 is known to play a causal role in the promotion of cancer, although the underlying mechanisms remain to be completely understood. Interplay between endogenous and environmental cues determines the fate of cancer development. The Eμ-myc transgenic mouse expresses elevated levels of c-Myc in the B cell lineage and develops B cell lymphomas with associated mutations in p53 or other genes linked to apoptosis. We generated Eμ-myc mice that either lacked the IL-6 gene, or lacked the STAT3 gene specifically in B cells to determine the role of the IL-6/JAK/STAT3 pathway in tumor development. Using the Eμ-myc lymphoma mouse model, we demonstrate that IL-6 is a critical tumor promoter during early stages of B cell lymphomagenesis. IL-6 is shown to inhibit the expression of tumor suppressors, notably BIM and PTEN, and this may contribute to advancing MYC-driven B cell tumorigenesis. Several miRNAs known to target BIM and PTEN are upregulated by IL-6 and likely lead to the stable suppression of pro-apoptotic pathways early during the tumorigenic process. STAT3, a classical downstream effector of IL-6, appears dispensable for Eμ-myc driven lymphomagenesis. We conclude that the growth-promoting and anti-apoptotic mechanisms activated by IL-6 are critically involved in Eμ-myc driven tumor initiation and progression, but the B cell intrinsic expression of STAT3 is not required.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Itziar Urizar-Arenaza ◽  
Aitor Benedicto ◽  
Arantza Perez-Valle ◽  
Nerea Osinalde ◽  
Vyacheslav Akimov ◽  
...  

AbstractHuman sperm protein associated with the nucleus on the X chromosome (SPANX) genes encode a protein family (SPANX-A, -B, -C and -D), whose expression is limited to the testis and spermatozoa in normal tissues and various tumour cells. SPANX-A/D proteins have been detected in metastatic melanoma cells, but their contribution to cancer development and the underlying molecular mechanisms of skin tumourigenesis remain unknown. Combining functional and proteomic approaches, the present work describes the presence of SPANX-A/D in primary and metastatic human melanoma cells and how it promotes pro-tumoural processes such as cell proliferation, motility and migration. We provide insights into the molecular features of skin tumourigenesis, describing for the first time a multifunctional role of the SPANX-A/D protein family in nuclear function, energy metabolism and cell survival, considered key hallmarks of cancer. A better comprehension of the SPANX-A/D protein subfamily and its molecular mechanisms will help to describe new aspects of tumour cell biology and develop new therapeutic targets and tumour-directed pharmacological drugs for skin tumours.


Circulation ◽  
2008 ◽  
Vol 118 (suppl_18) ◽  
Author(s):  
Ines Chopra ◽  
Huifang Li ◽  
Nanette H Bishopric ◽  
Keith A Webster

INTRODUCTION: Akt (PKB) is the downstream effector of insulin and IGF-1 and regulates multiple targets that control cell survival and growth. Akt is phosphorylated at Thr-308 (activating) and Ser-473 (regulatory) sites by PI3-kinase dependent kinase-1 (PDK1) and TOR2-rictor/PDK2 respectively. When energy is abundant Akt supports cell growth by stimulating TORC1 and inhibiting GSK3β. When energy and insulin are low such as during ischemia, AMPK is activated and depresses TORC1 by activating the repressive TSC1/2 complex. It is not known how AMPK affects TORC2 in this setting. HYPOTHESIS: Under low energy states AMPK promotes survival by differentially regulating TORC1 and 2 thereby maintaining Akt phosphorylation and survival in the absence of insulin. METHODS: Cardiac myocytes (CM) were subjected to glucose and insulin-free incubation for 4h and the activities of insulin signaling components were measured by western blot and co-IP. Survival was measured ± siRNAs by Hoechst/PI staining. RESULTS: After 4h of glucose/insulin deprivation there was increased phosphorylation of Akt-Thr-308 (11.2±2.4; n=6, p<0.001) and Akt-Ser-473 (9.7± 2; n=6, p<0.001). Phosphorylation of GSK-3β (Ser-9) also increased but there was decreased phosphorylation of p70S6-kinase (Thr389) and 4EBP (Ser65) indicating global down-regulation of TORC1. We identified 2 separate pathways for the insulin-independent phosphorylation of Akt at both sites in glucose-depleted CM. Activated AMPK promoted phosphorylation of insulin receptor substrate-1 (IRS-1) on Ser-789. AraA or a Ser-789 decoy peptide blocked this, and IRS-1-P-Ser-789 co-IP’d with PI3-kinase suggesting positive regulation of PDK1. Glucose depletion did not change the levels of rictor or raptor or their relative binding to TOR. However TSC1 and rictor were quantitatively associated with PI3-kinase selectively under glucose/insulin depletion and this correlated with Akt phosphorylation. CM survival under glucose/insulin depletion was impaired by siRNA-mediated knockdown of IRS-1 or rictor (n=3; P<0.05). CONCLUSIONS: AMPK maintains phosphorylation of Akt under low energy states in the absence of insulin. The mechanism involves IRS-1 phosphorylation by AMPK and enhanced binding of rictor and TSC1 to PI3K. This research has received full or partial funding support from the American Heart Association, AHA Greater Southeast Affiliate (Alabama, Florida, Georgia, Louisiana, Mississippi, Puerto Rico & Tennessee).


2021 ◽  
Vol 11 ◽  
Author(s):  
Hongfeng Zhao ◽  
Minping Bi ◽  
Meng Lou ◽  
Xiaowei Yang ◽  
Liwen Sun

ObjectiveLncRNA SOX2-OT is involved in a variety of cancers. This study explored the effect of lncRNA SOX2-OT on hepatocellular carcinoma (HCC) cells.MethodsSOX2-OT expressions were detected in HCC tissues and normal tissues, normal cells, and HCC cells. The relationship between SOX2-OT and prognosis was analyzed by TCGA. After SOX2-OT expression was inhibited using siRNA, HCC cell malignant behaviors were evaluated. The subcellular localization of SOX2-OT in HCC cells was predicted and analyzed. The binding relationships among SOX2-OT, miR-143-3p, and MSI2 were analyzed by bioinformatics website, dual-luciferase assay, and RNA pull-down assay. The effect of miR-143-3p and MSI2 on the regulation of SOX2-OT on biological behaviors of HCC cells was confirmed by functional rescue experiments. The effect of SOX2-OT on the tumorigenicity of HCC was evaluated by subcutaneous tumorigenesis in nude mice.ResultsSOX2-OT was highly expressed in HCC cells and tissues. The prognosis was poor in HCC patients with high SOX2-OT expression. Downregulating SOX2-OT inhibited HCC cell malignant behaviors. SOX2-OT bound to miR-143-3p to promote MSI2 expression. Downregulating miR-143-3p or upregulating MSI2 averted the role of si-SOX2-OT in HCC cells. Nude mouse subcutaneous tumorigenesis showed that SOX2-OT downregulation decreased the tumorigenicity of HCC, and affected the levels of miR-143-3p and MSI2 mRNA in tumor tissues.ConclusionSOX2-OT inhibited the targeted inhibition of miR-143-3p on MSI2 through competitively binding to miR-143-3p, thus promoting MSI2 expression and proliferation, invasion, and migration of HCC cells.


2018 ◽  
Vol 15 (3) ◽  
pp. 389-398
Author(s):  
Ruchi Singh

Rural economies in developing countries are often characterized by credit constraints. Although few attempts have been made to understand the trends and patterns of male out-migration from Uttar Pradesh (UP), there is dearth of literature on the linkage between credit accessibility and male migration in rural Uttar Pradesh. The present study tries to fill this gap. The objective of this study is to assess the role of credit accessibility in determining rural male migration. A primary survey of 370 households was conducted in six villages of Jaunpur district in Uttar Pradesh. Simple statistical tools and a binary logistic regression model were used for analyzing the data. The result of the empirical analysis shows that various sources of credit and accessibility to them play a very important role in male migration in rural Uttar Pradesh. The study also found that the relationship between credit constraints and migration varies across various social groups in UP.


Sign in / Sign up

Export Citation Format

Share Document