Proliferative Effect of Tilapia Fish (Oreochromis niloticus) Lectin on BALB/c Mice Splenocytes

2019 ◽  
Vol 26 (12) ◽  
pp. 887-892
Author(s):  
Cynarha Daysy Cardoso da Silva ◽  
Cristiane Moutinho Lagos de Melo ◽  
Elba Verônica Matoso Maciel Carvalho ◽  
Mércia Andréa Lino da Silva ◽  
Rosiely Félix Bezerra ◽  
...  

Background: Lectins have been studied in recent years due to their immunomodulatory activities. Objective: We purified a lectin named OniL from tilapia fish (Oreochromis niloticus) and here we analyzed the cell proliferation and cytokine production in Balb/c mice splenocytes. Methods: Cells were stimulated in vitro in 24, 48, 72 hours and 6 days with different concentrations of OniL and Con A. Evaluation of cell proliferation was performed through [3H]-thymidine incorporation, cytokines were investigated using ELISA assay and cell viability assay was performed by investigation of damage through signals of apoptosis and necrosis. Results: OniL did not promote significant cell death, induced high mitogenic activity in relation to control and Con A and stimulated the cells to release high IL-2 and IL-6 cytokines. Conclusion: These findings suggest that, like Con A, OniL lectin can be used as a mitogenic agent in immunostimulatory assays.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1526-1526
Author(s):  
Justin M Watts ◽  
Aymee Perez ◽  
Fernando Vargas ◽  
Yao-Shan Fan ◽  
Alejandra Weisman ◽  
...  

Abstract Introduction: We describe for the first time, a novel chromosomal translocation in AML. The cells bearing this translocation proliferated rapidly following stimulation with ATRA in vitro. A 30-year old woman presented with monocytic AML. Karyotype revealed: 46,XX,t(9;11)(p22;q23)[13]/46,XX[7]. Mutational analysis showed an activating mutation in NRAS (c.34G>A; p.G12S). She achieved remission with standard induction therapy followed by cytarabine consolidation in the absence of an optimal donor. Following a disease free interval of 8 months, she relapsed. Karyotype at relapse: 46,XX,t(4;15)(q31;q22),t(9;11)(p22;q23)[20] (Figure 1). Mutational analysis revealed no new mutations. In the setting of chemotherapy refractory disease, she was enrolled on a phase I clinical trial combining escalating doses of TCP (tranylcypromine, Parnate¨) with fixed doses of ATRA (NCT02273102). The patient died from rapid disease progression shortly after her first cycle of therapy. Unexpectedly, we observed a rapid proliferation in the patient's blasts following treatment with ATRA in vitro. We hypothesized that the novel t(4;15) translocation was involved in the regulation of retinoic acid (RA) signaling, and may have contributed to the rapid disease progression observed in this patient when she was treated with ATRA. To this end, we fully characterized the fusion gene and created a cell line bearing the translocation. Using these cells, we will further elucidate the mechanisms accounting for a rare and potentially clinically relevant effect of ATRA, in vitro. Methods: Cytogenetics and molecular-cytogenetic techniques, next generation sequencing (WES), RNA-seq, RT-PCR, and direct Sanger sequencing were used to map the chromosomal translocation. Cell proliferation and Annexin V apoptosis assays were used to test the effects of ATRA and various retinoic acid receptor (RAR) agonists and antagonists in vitro. Cells were treated with ATRA, RARα, and RARγ agonists and antagonists for 72 h and cell proliferation and apoptosis were measured with CellTiter-Glow (Promega)¨ luminescence viability assay and Annexin V/PI staining (FACS), respectively. Results: We mapped the t(4;15)(q31;q22) at the single nucleotide level, and discovered a novel fusion of TMEM154 (4q31.3) and RASGRF1 (15q24.2) genes at both the DNA and RNA level. The fusion protein included exons 1-6 from TMEM154 and exons 15-24 from RASGRF1. RASGRF1 (Ras protein specific nucleotide releasing factor 1) activates Ras by catalyzing the exchange of Ras-bound GDP for GTP. We determined that TMEM154 (transmembrane protein 154) is regulated by RA based on gene expression profiling. This particular translocation appears to result in a proliferative advantage in AML cells treated with RAR agonists such as ATRA, even at physiological levels (10 nM) (Figure 2). In MLL-rearranged cell lines without t(4;15), ATRA has a neutral or anti-proliferative effect, suggesting that the t(4;15) product interacts with the RA pathway to induce proliferation of AML blasts. We hypothesize that by increasing expression of TMEM154-RASGRF1, ATRA drives proliferation through Ras signaling. Of note, RAR antagonists inhibited proliferation and induced cell death (Figure 2). Also, treating the cells with MAP Kinase and Ras inhibitors consistently diminished the proliferative effect of ATRA. Updated studies shedding light on the mechanism of interaction between RA signaling and the TMEM154-RASGRF1 fusion protein will be presented. Conclusions: Our study for the first time identifies t(4;15) as a novel and pathogenic translocation in AML. This rare event may confer a proliferative advantage in the presence of ATRA and caution is advised should patients with this lesion be considered for treatment with ATRA. Figure 1 Conventional karyotyping showing 46,XX,t(4;15)(q31;q22),t(9;11)(p22;q23).Chromosome analysis was performed on 20 G-banded metaphase cells from multiple unstimulated cultures. Both translocations were present in all cells examined. Figure 1. Conventional karyotyping showing 46,XX,t(4;15)(q31;q22),t(9;11)(p22;q23).Chromosome analysis was performed on 20 G-banded metaphase cells from multiple unstimulated cultures. Both translocations were present in all cells examined. Figure 2 ATRA and other RAR agonists increased proliferation in t(4;15) AML cells. Primary cells from a patient with t(4;15) AML were treated with ATRA, RARα (AM80 and 195183), and RARγ (205327) agonists, and RARα (196996), RARγ (205728) and dual (194310) antagonists. Cell proliferation was determined by CellTiter-Glo¨ luminescent cell viability assay (Promega) after 72 h of treatment. Figure 2. ATRA and other RAR agonists increased proliferation in t(4;15) AML cells. Primary cells from a patient with t(4;15) AML were treated with ATRA, RARα (AM80 and 195183), and RARγ (205327) agonists, and RARα (196996), RARγ (205728) and dual (194310) antagonists. Cell proliferation was determined by CellTiter-Glo¨ luminescent cell viability assay (Promega) after 72 h of treatment. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 6 (1) ◽  
Author(s):  
Jun Liu ◽  
Jipeng Li ◽  
Ke Wang ◽  
Haiming Liu ◽  
Jianyong Sun ◽  
...  

AbstractFork-head box protein M1 (FoxM1) is a transcriptional factor which plays critical roles in cancer development and progression. However, the general regulatory mechanism of FoxM1 is still limited. STMN1 is a microtubule-binding protein which can inhibit the assembly of microtubule dimer or promote depolymerization of microtubules. It was reported as a major responsive factor of paclitaxel resistance for clinical chemotherapy of tumor patients. But the function of abnormally high level of STMN1 and its regulation mechanism in cancer cells remain unclear. In this study, we used public database and tissue microarrays to analyze the expression pattern of FoxM1 and STMN1 and found a strong positive correlation between FoxM1 and STMN1 in multiple types of cancer. Lentivirus-mediated FoxM1/STMN1-knockdown cell lines were established to study the function of FoxM1/STMN1 by performing cell viability assay, plate clone formation assay, soft agar assay in vitro and xenograft mouse model in vivo. Our results showed that FoxM1 promotes cell proliferation by upregulating STMN1. Further ChIP assay showed that FoxM1 upregulates STMN1 in a transcriptional level. Prognostic analysis showed that a high level of FoxM1 and STMN1 is related to poor prognosis in solid tumors. Moreover, a high co-expression of FoxM1 and STMN1 has a more significant correlation with poor prognosis. Our findings suggest that a general FoxM1-STMN1 axis contributes to cell proliferation and tumorigenesis in hepatocellular carcinoma, gastric cancer and colorectal cancer. The combination of FoxM1 and STMN1 can be a more precise biomarker for prognostic prediction.


Cells ◽  
2021 ◽  
Vol 10 (7) ◽  
pp. 1606
Author(s):  
Peter Seiringer ◽  
Stefanie Eyerich ◽  
Kilian Eyerich ◽  
Daniela Dittlein ◽  
Anna Caroline Pilz ◽  
...  

Whilst the importance of keratinocytes as a first-line defense has been widely investigated, little is known about their interactions with non-resident immune cells. In this study, the impact of human keratinocytes on T cell effector functions was analyzed in an antigen-specific in vitro model of allergic contact dermatitis (ACD) to nickel sulfate. Keratinocytes partially inhibited T cell proliferation and cytokine production. This effect was dependent on the keratinocyte/T cell ratio and was partially reversible by increasing the number of autologous dendritic cells. The inhibition of T cell proliferation by keratinocytes was independent of the T cell subtype and antigen presentation by different professional antigen-presenting cells. Autologous and heterologous keratinocytes showed comparable effects, while the fixation of keratinocytes with paraformaldehyde abrogated the immunosuppressive effect. The separation of keratinocytes and T cells by a transwell chamber, as well as a cell-free keratinocyte supernatant, inhibited T cell effector functions to the same amount as directly co-cultured keratinocytes, thus proving that soluble factor/s account for the observed suppressive effects. In conclusion, keratinocytes critically control the threshold of inflammatory processes in the skin by inhibiting T cell proliferation and cytokine production.


2021 ◽  
Vol 22 (13) ◽  
pp. 7063
Author(s):  
Sharon Mordechay ◽  
Shaun Smullen ◽  
Paul Evans ◽  
Olga Genin ◽  
Mark Pines ◽  
...  

Progressive loss of muscle and muscle function is associated with significant fibrosis in Duchenne muscular dystrophy (DMD) patients. Halofuginone, an analog of febrifugine, prevents fibrosis in various animal models, including those of muscular dystrophies. Effects of (+)/(−)-halofuginone enantiomers on motor coordination and diaphragm histopathology in mdx mice, the mouse model for DMD, were examined. Four-week-old male mice were treated with racemic halofuginone, or its separate enantiomers, for 10 weeks. Controls were treated with saline. Racemic halofuginone-treated mice demonstrated better motor coordination and balance than controls. However, (+)-halofuginone surpassed the racemic form’s effect. No effect was observed for (−)-halofuginone, which behaved like the control. A significant reduction in collagen content and degenerative areas, and an increase in utrophin levels were observed in diaphragms of mice treated with racemic halofuginone. Again, (+)-halofuginone was more effective than the racemic form, whereas (−)-halofuginone had no effect. Both racemic and (+)-halofuginone increased diaphragm myofiber diameters, with no effect for (−)-halofuginone. No effects were observed for any of the compounds tested in an in-vitro cell viability assay. These results, demonstrating a differential effect of the halofuginone enantiomers and superiority of (+)-halofuginone, are of great importance for future use of (+)-halofuginone as a DMD antifibrotic therapy.


2020 ◽  
Vol 11 (1) ◽  
pp. 319-327
Author(s):  
Chenlin Xu ◽  
Zijian Xiao ◽  
Heng Wu ◽  
Guijuan Zhou ◽  
Duanqun He ◽  
...  

AbstractBackgroundAlzheimer’s disease (AD) is a common neurodegenerative disorder without any satisfactory therapeutic approaches. AD is mainly characterized by the deposition of β-amyloid protein (Aβ) and extensive neuronal cell death. Curcumin, with anti-oxidative stress (OS) and cell apoptosis properties, plays essential roles in AD. However, whether bisdemethoxycurcumin (BDMC), a derivative of curcumin, can exert a neuroprotective effect in AD remains to be elucidated.MethodsIn this study, SK-N-SH cells were used to establish an in vitro model to investigate the effects of BDMC on the Aβ1–42-induced neurotoxicity. SK-N-SH cells were pretreated with BDMC and with or without compound C and EX527 for 30 min after co-incubation with rotenone for 24 h. Subsequently, western blotting, cell viability assay and SOD and GSH activity measurement were performed.ResultsBDMC increased the cell survival, anti-OS ability, AMPK phosphorylation levels and SIRT1 in SK-N-SH cells treated with Aβ1–42. However, after treatment with compound C, an AMPK inhibitor, and EX527, an SIRT1inhibitor, the neuroprotective roles of BDMC on SK-N-SH cells treated with Aβ1–42 were inhibited.ConclusionThese results suggest that BDMC exerts a neuroprotective role on SK-N-SH cells in vitro via AMPK/SIRT1 signaling, laying the foundation for the application of BDMC in the treatment of neurodegenerative diseases related to AMPK/SIRT1 signaling.


Toxins ◽  
2018 ◽  
Vol 10 (12) ◽  
pp. 508 ◽  
Author(s):  
Daniela Luz ◽  
Maria Amaral ◽  
Flavia Sacerdoti ◽  
Alan Bernal ◽  
Wagner Quintilio ◽  
...  

Shiga toxin (Stx) producing Escherichia coli (STEC) is responsible for causing hemolytic uremic syndrome (HUS), a life-threatening thrombotic microangiopathy characterized by thrombocytopenia, hemolytic anemia, and acute renal failure after bacterially induced hemorrhagic diarrhea. Until now, there has been neither an effective treatment nor method of prevention for the deleterious effects caused by Stx intoxication. Antibodies are well recognized as affinity components of therapeutic drugs; thus, a previously obtained recombinant human FabC11:Stx2 fragment was used to neutralize Stx2 in vitro in a Vero cell viability assay. Herein, we demonstrated that this fragment neutralized, in a dose-dependent manner, the cytotoxic effects of Stx2 on human glomerular endothelial cells, on human proximal tubular epithelial cells, and prevented the morphological alterations induced by Stx2. FabC11:Stx2 protected mice from a lethal dose of Stx2 by toxin-antibody pre-incubation. Altogether, our results show the ability of a new encouraging molecule to prevent Stx-intoxication symptoms during STEC infection.


1994 ◽  
Vol 53 (1) ◽  
pp. 44-49 ◽  
Author(s):  
Michael Untch ◽  
Bernd-Uwe Sevin ◽  
James P. Perras ◽  
Roberto Angioli ◽  
Andrea Untch ◽  
...  

2020 ◽  
Vol 17 (36) ◽  
pp. 871-883
Author(s):  
Moath Kahtan BASHIR ◽  
Yasser Fakri MUSTAFA ◽  
Mahmood Khudhayer OGLAH

Cancer constitutes one of the most severe public health menaces worldwide. It is imperative to synthesize new compounds and explore their antitumor activity to find a potential resolution to this health problem. Synthesis of new scaffolds and evaluating their antitumor activity is a relevant approach for combating cancer development. Coumarins can exhibit diverse biological activities, and one of these is the antitumor activity. This study aimed to synthesize new coumarins by grafting their precursors to the aromatic amines via Schiff base formation and evaluating their introductory antitumor activity. New multifunctional coumarins (MC1-MC9) were prepared by integrating a functionalized coumarin with different toluidine derivatives via a Schiff-base linkage. Spectral characterization inspired by FTIR, 1H- and 13C- NMR spectroscopies has established the chemical structures of the synthesized products. The antitumor activity was explored in vitro versus four dominant human cancer lines, including HeLa, SKG, MCF-7, and AMN3. The outcomes acquired from the cell viability assay inspected by applying MTT dye have revealed that the synthesized multifunctional coumarins, particularly MC3, have a hopeful activity. It can be concluded that a similar trend of activity against the test cell lines was observed for the synthesized coumarins, with the best action being versus MCF-7 and the least one versus AMN3. This study not only affords a new scaffold of a significant antitumor activity but also provides some insights into its structureactivity relationship.


2010 ◽  
Vol 138 (2) ◽  
pp. 671-681.e2 ◽  
Author(s):  
Chihiro Morishima ◽  
Margaret C. Shuhart ◽  
Chia C. Wang ◽  
Denise M. Paschal ◽  
Minjun C. Apodaca ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document