scholarly journals A Relatively Small Gradient of Extracellular pH Directs Migration of MDA-MB-231 Cells In Vitro

2020 ◽  
Vol 21 (7) ◽  
pp. 2565
Author(s):  
Eiji Takahashi ◽  
Daisuke Yamaguchi ◽  
Yoshihisa Yamaoka

Hematogenous tumor metastasis begins with the invasion and spread of primary tumor cells in the local tissue leading to intravasation. We hypothesized that tumor cells might actively migrate toward intratumor vessels with the extracellular metabolic gradient acting as a guiding cue. Here, we determined in vitro whether the extracellular gradient of pH can act as a cue for directional migration in MDA-MB-231 cells. Cell migration was determined by the wound-healing assay under gradients of extracellular pH (~0.2 units/mm) and oxygen concentration (~6% O2/mm) that were produced by a microfluidic device, gap cover glass (GCG). Without GCG, the migration of cells was spatially homogeneous; the same number of cells migrated to the rectangular wound space from the left and right boundaries. In contrast, when GCG generated pH/O2 gradients across the wound space, the number of cells migrating to the wound space from the boundary with higher pH/O2 values was considerably decreased, indicating a preferential movement of cells toward the region of higher pH/O2 in the gradient. The addition of hepes in the extracellular medium abolished both the extracellular pH gradient and the directional cell migration under GCG. We conclude that relatively small gradients of pH in the extracellular medium compared to those found in Na+/H+ exchanger-driven cell migration were sufficient to guide MDA-MB-231 cells. The directional cell migration as guided by the metabolic gradient could effectively elevate the probability of intravasation and, ultimately, hematogenous metastasis.

Author(s):  
Artyom Mylnikov ◽  
Nikita Navolokin ◽  
Dmitry Mudrak ◽  
Natalya Polukonova ◽  
Alla Bucharskaya ◽  
...  

Objective of the study: We used fluorescence imaging methods of apoptosis and necrosis in human renal carcinoma A498 tumor cells in vitro to reveal the indicated forms of cell death under the combined effect of flavonoid-containing extract of Gratiola officinalis and cytostatic (cyclophosphamide). Materials and methods: The dyes were propidium iodide and acridine orange, which were used in the “alive and dead” test. This test helped us to identify the total number of dead cells in the forms of necrosis and apoptosis and the number of cells in which apoptosis had started, it was characterized by the appearance of apoptotic bodies or nucleus pyknosis. Results: We found the most pronounced cytotoxic activity at the ratio of extract of Gratiola officinalis and cyclophosphamide concentrations of 1:1. The number of living cells decreased when exposed to the ratio of extract and cytostatic concentrations of 2:1. When the ratio of concentration of the extract relative to the cytostatic increased to 3:1, the cytostatic activity of the extract began to appear, the total number of tumor cells decreased. The number of cells with nucleus pyknosis and the number of cells with apoptosis signs significantly increased at a 3:1 ratio of extract and cytostatic concentrations, which confirms the presence of pro-apoptotic activity of the studied combination. This trend indicates the dependence of a certain form of cell death (apoptosis, necrosis) on the ratio of extract and cytostatic doses, and it also demonstrates the cytostatic and cytotoxic effects of this combination. Conclusion: Fluorescence methods of investigation in the “alive and dead” test allowed us to visualize the forms of cell death of human kidney carcinoma A498 by combined exposure to the flavonoid-containing extract of Gratiola officinalis and cytostatic (cyclophosphamide) 24 h after exposure. We found that the combination with a concentration ratio of the extract and cyclophosphamide of 3:1 has the greatest effectiveness due to stimulation of the cytostatic effect and cytotoxic effect.


Development ◽  
1982 ◽  
Vol 69 (1) ◽  
pp. 47-59
Author(s):  
A. S. Mendoza ◽  
W. Breipohl ◽  
F. Miragall

The diflferentiation of the olfactory placode in the chick has been studied using light and electron microscopy. Special attention was paid to the appearance of neuronal cells within the placodal ectodermal thickening, the migration of cells out of this tissue and the appearance of the first fila olfactoria in the differentiating olfactory mucosa. Between the third and fifth day of incubation a large number of cells is observed leaving the base of the invaginating olfactory placode, often in contact with thin axon bundles. These cells are characterized by a well-developed Golgi apparatus, a considerable number of mitochondria and dense-core vesicles. The morphology of these migrating cells resembles that of cells observed near the basement membrane within the developing olfactory epithelium and is clearly different from the mesenchymal cells which are filled with polyribosomes. At the sixth day of incubation thick axon bundles can be observed within the epithelium and the underlying lamina propria. The possible fate of the migrated epitheloid cells is discussed.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 604-604
Author(s):  
Sarah E. M. Herman ◽  
Jade Jones ◽  
Rashida Z. Mustafa ◽  
Mohammed Farooqui ◽  
Adrian Wiestner

Abstract The Bruton’s tyrosine kinase inhibitor ibrutinib has recently been shown to be well tolerated, and to induce objective clinical responses in the majority of patients, irrespective of adverse prognostic markers (Byrd et al., NEJM 2013). Despite the demonstrated clinical activity, ibrutinib also leads to a transient lymphocytosis that is thought to reflect a redistribution of cells from tissue compartments into the peripheral blood. The mechanisms contributing to this lymphocytosis are not well understood. To date, two groups have demonstrated that in vitro treatment with ibrutinib inhibits migration of CLL cells in chemokine gradients (de Rooji et al., Blood 2012 and Ponader et al., Blood 2012). Here we sought to assess the in vivo effect of ibrutinib on cellular migration. To validate our assay we first treated CLL cells with 1uM ibrutinib for 1 hour in vitro and measured migration of CLL cells to a mix of SDF-1 (at 200ng/mL) and CCL19 (at 100ng/mL), two chemo-attractants known to induce migration of CLL cells. Migration was assayed in a dual chamber system separated by a membrane with 5µm pores after 3 hours of incubation. Confirming published data we found a significant reduction in the migration index (ratio of migration to chemokines divided by migration to media alone) of ibrutinib treated cells compared to untreated cells (mean reduction 24%; P = 0.04). Next, we analyzed the migration of CLL cells obtained from patients (n = 9) enrolled on a clinical trial with single agent ibrutinib that were sampled pre-treatment and after 4 weeks on drug. We observed highly variable responses; in about half of the patients treated cells showed increased migration, while in the other half there was decreased migration to the SDF-1/CCL19 mix. Interestingly, patients showing a decrease in migration on treatment often had del17p but there was no difference in regards to IGHV mutation status and no correlation to the degree of lymphocytosis observed in the patient. T-cell migration was not affected by ibrutinib. In order to extend the analysis to a mix of chemo-attractants that the tumor cells may encounter in vivo we used the supernatant harvested from bone marrow aspirates and found that it efficiently induced migration of CLL cells in the dual chamber assay (mean fold increase 5.2 compared to control). Comparing CLL cells from patients sampled pre-treatment to those obtained on treatment day 28 we again found the same mixed effects of ibrutinib on the ability of CLL cells to migrate to bone marrow plasma as we had observed with the SDF-1/CCL19 mix. Thus, direct inhibition of CLL cell migration can account for only a subset of patients with treatment-induced lymphocytosis. Given reports that ibrutinib can inhibit cytokine and chemokine secretion from CLL cells and T-cells (Ponader et al., Blood, 2012; Herman et al., Blood, 2011), we hypothesized that ibrutinib treatment might change the content of chemo-attractants in the bone marrow We therefore compared the ability of the bone marrow plasma obtained pre-treatment and after 2 months on ibrutinib to attract CLL cells (these cells were obtained from the peripheral blood pre-treatment from the same patient donating the marrow). We found that in 4/4 patients evaluated there was a significant reduction in the migration of CLL cells to the on-treatment bone marrow plasma compared to the matching pre-treatment sample (mean decrease 20%; P < 0.05). In conclusion, migration of CLL cells from patients on ibrutinib can be inhibited or increased, with most del17p patients showing decreased migration. Intriguingly, these patients tend to have slower resolution of the treatment induced lymphocytosis, raising the question whether inhibition of homing to tissue sites could affect the time to resolution of the lymphocytosis. In addition, we provide evidence that bone marrow plasma on ibrutinib therapy has a reduced capacity to attract CLL cells, suggesting that ibrutinib may alter the composition of the bone marrow microenvironment This work was supported by the Intramural Research Program of NHLBI, NIH. We thank our patients for donating blood and tissue samples to make this research possible. We acknowledge Pharmacyclics for providing study drug. Disclosures: Off Label Use: Ibrutinib in chronic lymphocytic leukemia.


2020 ◽  
Author(s):  
Fan Shi ◽  
Dan Luo ◽  
Zhou Xuexiao ◽  
Qiaozhen Sun ◽  
Pei Shen ◽  
...  

Abstract Objective: Oral squamous cell carcinoma (OSCC) represents one of the main types of head and neck malignant tumors with high incidence and mortality as well extremely poor prognosis. Hyperthermia (HT) shows great promises for tumor therapy. However it can promote autophagy in tumor microenvironment, which is found to serve as a surviving mechanism for cancer cells. Inhibiting autophagy has been considered as an adjuvant anti-cancer strategy. The present study investigated the role of HT-induced autophagy, while attempting to combine chemotherapy and autophagy blocking with HT in OSCC cells under hypoxia and starvation microenvironment.Materials and methods: HIF-1α and Beclin-1 expression in tissues was determined by immunohistochemistry in 80 OSCC sample pairs. The IC50 of CoCl2, YC-1 (an inhibitor of HIF-1α) and 3-MA (an inhibitor of autophagy) was detected by CCK-8. CoCl2 and complete culture medium without serum were used to achieve the hypoxic and nutrient deficient microenvironment, respectively. HT was performed by heating in a 42 ℃ water bath. The role of HT and YC-1,3-MA on autophagy in vitro were assessed by qRT-PCR and Western blot, and the secretion of high mobility group box1 (HMGB1) was determined by ELISA. The migration and apoptosis rates of cells were assessed by wound healing assay and flow cytometry.Results: We observed that HIF-1α and Beclin1 were highly expressed in OSCC tissues, which were correlated with more advanced malignancy features. CoCl2 could establish hypoxia microenvironment, induce HIF-1α expression with dose-dependence as well as promote cell migration in Cal-27 and SCC-15 cells. Notably, hyperthermia and hypoxia could activate the HIF-1α/BNIP3/Beclin1 signaling pathway and promote HMGB1 secretion, which triggered cytoprotective autophagy to counteract the hypoxia and starvation cellular stresses, as indicated by downregulation of p62 and light chain 3-II (LC3 II). Furthermore, we found that hyperthermia combined YC-1 and/or 3-MA suppressed autophagy and cell migration whereas facilitated cell apoptosis.Conclusion: The present study demonstrated that combined use of YC-1 and 3-MA might increase death of tumor cells in physiological and hyperthermia conditions, which could be relevant with the inhibition of autophagy in OSCC tumor cells under hypoxia microenvironment in vitro.


2017 ◽  
Author(s):  
Edwin F. Juarez ◽  
Carolina Garri ◽  
Ahmadreza Ghaffarizadeh ◽  
Paul Macklin ◽  
Kian Kani

AbstractWe describe an integrated experimental-computational pipeline for quantifying cell migration in vitro. This pipeline is robust to image noise, open source, and user friendly. The experimental component uses the Oris cell migration assay (Platypus Technologies) to create migration regions. The computational component of the pipeline creates masks in Matlab (MathWorks) to cell-covered regions, uses a genetic algorithm to automatically select the migration region, and outputs a metric to quantify the migration of cells. In this work we demonstrate the utility of our pipeline by quantifying the effects of a drug (Taxol) and of the secreted Anterior Gradient 2 (sAGR2) protein in the migration of MDA-MB-231 cells (a breast cancer cell line). In particular, we show that blocking sAGR2 reduces migration of MDA-MB-231 cells.


Stroke ◽  
2012 ◽  
Vol 43 (suppl_1) ◽  
Author(s):  
Sahar Rosenblum ◽  
Nancy Wang ◽  
Joshua Chua ◽  
Eric Westbroek ◽  
Tenille Smith ◽  
...  

Intro: Increasing cell migration through upregulation of chemokine and adhesion molecule receptors could improve intravascular cell treatment for stroke and BDNF has been shown to induce these pathways. Therefore we tested whether BDNF cell-pretreatment would improve cell migration and functional recovery in an experimental stroke model. Methods: hES-derived NPCs (5x10 5 in 5µl saline) pre-treated with BDNF for 5 hours and harboring a reporter gene construct containing renilla luciferase and eGFP in serum free media, non-treated hES-derived NPCs (5×10 5 in 5[l saline) in media, and media control with BDNF were delivered to the brain via the ipsilateral carotid artery at 3 days after hypoxic-ischemic stroke in NODSCID mice (n=11/group). Cell engraftment was monitored by in-vivo bioluminescence imaging (BLI). The ladder test was used to assess behavioral recovery throughout a 4 week time course. Brain homogenates from animals at 28 days were analyzed using RT-qPCR for common chemokines, adhesion molecules, and neurotrophins. Mechanisms of cell migration were evaluated by assessing cell receptor expression of chemokines and adhesion molecules on hES-derived NPC and by analyzing the change in expression profile in the mouse brain at 3 days after stroke. Boyden-chamber migration assays were used to evaluate cell migratory potential in vitro . RESULTS: One day after cell transplantation the subset of animals transplanted with BDNF-pretreated cells showed significantly higher BLI signal at 1 (p=0.021) and 7 days after transplantation (p=0.002). Histological analysis also revealed engraftment of hES-derived NPC at 1 week after transplantation. Behavioral assessment revealed significant functional recovery in the BDNF pre-treated group throughout the 28 day time course (ANOVA, p<0.05). BDNF-pretreatment of hES-derived NPCs upregulated CXCR4 expression 12.5 times and in vitro led to significantly greater migration in response to CXCL12 (CXCR4 ligand) compared to untreated cells. At 28 days after transplantation, neurotrophic factors IL6, IL10, Ntrk1 were upregulated 3.3, 3.4, and 3.3 times. Common T-cell and neutrophil cytokine receptors IL8rb, IL8ra and IL1a were all downregulated, while several chemokines that increase migration of inflammatory cells were downregulated including CCL2, CCL5, CCL8, and CCL12. Anti-Angiogenic factor Adamts8 was also downrgulated in the brains of animals transplanted with BDNF pre-treated cells. Lastly, MMP3, MMP8, and MMP9 were downregulated at 28 days after stroke indicating increased blood brain barrier integrity. Conclusion: Intravascular transplantation of BDNF pre-treated hES-derived NPCs elicits functional gains via increased migration of cells, immunomodulation, increased BBB integrity, and by influencing the upregulation of neur0protective factors.


2019 ◽  
Vol 20 (1) ◽  
Author(s):  
Xiaojuan Wang ◽  
Charlotte Caroline Decker ◽  
Laura Zechner ◽  
Sonja Krstin ◽  
Michael Wink

PeerJ ◽  
2017 ◽  
Vol 5 ◽  
pp. e4033 ◽  
Author(s):  
Tove Kirkegaard ◽  
Mikail Gögenur ◽  
Ismail Gögenur

Background The perioperative period is important for patient outcome. Colorectal cancer surgery can lead to metastatic disease due to release of disseminated tumor cells and the induction of surgical stress response. To explore the overall effects on surgically-induced changes in serum composition, in vitro model systems are useful. Methods A systematic search in PubMed and EMBASE was performed to identify studies describing in vitro models used to investigate cancer cell growth/proliferation, cell migration, cell invasion and cell death of serum taken pre- and postoperatively from patients undergoing colorectal tumor resection. Results Two authors (MG and TK) independently reviewed 984 studies and identified five studies, which fulfilled the inclusion criteria. Disagreements were solved by discussion. All studies investigated cell proliferation and cell invasion, whereas three studies investigated cell migration, and only one study investigated cell death/apoptosis. One study investigated postoperative peritoneal infection due to anastomotic leak, one study investigated mode of anesthesia (general anesthesia with volatile or intravenous anesthetics), and one study investigated preoperative intervention with granulocyte macrophage colony stimulating factor (GMCSF). In all studies an increased proliferation, cell migration and invasion was demonstrated after surgery. Anesthetics with propofol and intervention with GMCSF significantly reduced postoperative cell proliferation, whereas peritoneal infection enhanced the invasive capability of tumor cells. Conclusion This study suggests that in vitro cell models are useful and reliable tools to explore the effect of surgery on colorectal cancer cell proliferation and metastatic ability. The models should therefore be considered as additional tests to investigate the effects of perioperative interventions.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e23149-e23149
Author(s):  
Thomas Nelius ◽  
Courtney Jarvis ◽  
Stephanie Filleur

e23149 Background: Despite the approval of several new agents, taxanes remains the main treatment with survival benefits for castration-refractory metastatic prostate cancer/mCRPC. Still their mechanisms of action and therapeutic efficacy remain incompletely characterized. In the present study, we proposed to compare docetaxel/Doc and cabazitaxel/Cab, delivered as monotherapy or in combination with the anti-angiogenic and anti-tumoral Pigment Epithelium-Derived Factor/PEDF, on CRPC cells in vitro and in vivo. Methods: CRPCcells were assessed for cell growth, cell cycle, and apoptosis under taxane/control treatment by cytotoxicity, PI incorporation and TUNEL assays. CL1 cells that express PEDF/control were used in vivo. Tumor cells were injected s.c. into CB17-SCID mice. After two weeks, mice were randomized into groups: 1) Placebo; 2) Doc 5mg/kg i.p. d4; and 3) Cab 5-1-0.5-0.1mg/kg i.p. d4, d1-7, d2, daily. To assess the anti-tumor effect of the combination, tumor cell migration and phagocytosis were measured by Boyden chamber and confocal microscopy analyses of CL1-RAW264.7 macrophages co-cultures. Results: Cab was more cytotoxic than Doc in all the cell lines tested.This effect was concomitant to increased cell death, but was not due to autophagy or necrosis. Inversely, we showed that apoptosis was superior in Cab-treated cells than in Doc treatment. In vivo, while 0.5 and 0.1 mg/kg Cab did not improve PEDF efficacy on tumor growth, 1mg/kg was very toxic. In contrast, PEDF combined with 5mg/kg Cab lead to stabilization of the disease and was also found to be drastically more efficient than PEDF/Doc in delaying the disease. In vitro, PEDF/Cab inhibited tumor cell migration at a significant superior level compared to PEDF/Doc. Finally, tumor cells phagocytosis was induced in PEDF/Cab suggesting that the combination may target macrophages within the tumor microenvironment. Conclusions: Our data demonstrated the greater anti-tumor efficacy of Cab compared to Doc. They also insist on the fact that PEDF/Cab could be used as a novel combined therapy for CRPC, and emphasize on the importance in evaluating the cytotoxicity of the novel combination tested and investigating the role of the tumor microenvironment in the anti-tumor effect.


Sign in / Sign up

Export Citation Format

Share Document