scholarly journals Development of a reliable automated screening system to identify small molecules and biologics that promote human β-cell regeneration

2016 ◽  
Vol 311 (5) ◽  
pp. E859-E868 ◽  
Author(s):  
Kristie I. Aamodt ◽  
Radhika Aramandla ◽  
Judy J. Brown ◽  
Nathalie Fiaschi-Taesch ◽  
Peng Wang ◽  
...  

Numerous compounds stimulate rodent β-cell proliferation; however, translating these findings to human β-cells remains a challenge. To examine human β-cell proliferation in response to such compounds, we developed a medium-throughput in vitro method of quantifying adult human β-cell proliferation markers. This method is based on high-content imaging of dispersed islet cells seeded in 384-well plates and automated cell counting that identifies fluorescently labeled β-cells with high specificity using both nuclear and cytoplasmic markers. β-Cells from each donor were assessed for their function and ability to enter the cell cycle by cotransduction with adenoviruses encoding cell cycle regulators cdk6 and cyclin D3. Using this approach, we tested 12 previously identified mitogens, including neurotransmitters, hormones, growth factors, and molecules, involved in adenosine and Tgf-1β signaling. Each compound was tested in a wide concentration range either in the presence of basal (5 mM) or high (11 mM) glucose. Treatment with the control compound harmine, a Dyrk1a inhibitor, led to a significant increase in Ki-67+ β-cells, whereas treatment with other compounds had limited to no effect on human β-cell proliferation. This new scalable approach reduces the time and effort required for sensitive and specific evaluation of human β-cell proliferation, thus allowing for increased testing of candidate human β-cell mitogens.

2019 ◽  
Vol 9 (7) ◽  
pp. 982-987
Author(s):  
Xiaoying Wang ◽  
Yanke Hao

Vascular smooth muscle cell (VSMC) abnormal proliferation is related to hypertension. P27 can arrest cell cycle and its downregulation is associated with hypertension. miR-155 plays a regulatory role in VSMC proliferation, while its relationship with hypertension is still unclear. Bioinformatics analysis reveals a relationship between p27 mRNA and miR-155. The present study explores miR-155's role in p27 expression, VSMC proliferation and apoptosis, as well as in the pathogenesis of hypertension. Dual luciferase assay verified the relationship between miR-155 and p27. miR155, p27, α-SMA, and Ki-67 expressions in the thoracic aorta media of rat hypertension model were detected. VSMCs were cultured in vitro and grouped into, anti-miR-NC, anti-miR-155, pIRES2-blank, pIRES2-p27, and anti-miR-155 + pIRES2-p27 groups followed by analysis of cell cycle by flow cytometry and cell proliferation by EdU staining. Hypertension rats were randomly divided into antagomir-155 and antagomir-control. Caudal artery systolic and diastolic pressures were measured. miR-155 suppressed p27 expression. miR-155 and Ki-67 expressions were significantly enhanced, while p27 and α-SMA levels were reduced in the tunica media from hypertension rats compared with control. Downregulation of miR-155 and/or upregulation of p27 obviously declined cell proliferation and arrested cell cycle in G1 phase. Antagomir-155 injection significantly decreased systolic and diastolic pressures, elevated p27 and α-SMA expressions in media, and reduced the thickness of tunica media. miR-155 enhances VSMC proliferation via regulating p27. miR-155 enhancement was related to hypertension. miR-155 plays a therapeutic effect in hypertension.


2020 ◽  
Author(s):  
Yeting Hong ◽  
Wei He ◽  
Jianbin Zhang ◽  
Lu Shen ◽  
Chong Yu ◽  
...  

Abstract Background: Cyclin D3-CDK6 complex is a component of the core cell cycle machinery that regulates cell proliferation. By using Human Protein Atlas database, a higher expression level of this complex was found in gastric cancer. However, the function of this complex in gastric cancer remain poorly understood. This study aims to determine the expression pattern of this complex in gastric cancer and to investigate its biological role during tumorigenesis.Methods: To demonstrate that Cyclin D3-CDK6 regulate the c-Myc/miR-15a/16 axis in a feedback loop in gastric cancer, a series of methods were conducted both in vitro and in vivo experiments, including qRT-PCR, western blot analysis, EdU assay, flow cytometry, luciferase reporter assay and immunohistochemical staining. SPSS and Graphpad prism software were used for data analysis.Results: In this study, we found that Cyclin D3 and CDK6 were significantly upregulated in gastric cancer and correlated with poorer overall survival. Further study proved that this complex significantly promoted cell proliferation and cell cycle progression in vitro and accelerated xenografted tumor growth in vivo. Furthermore, we explored the molecular mechanisms through which the complex mediated Rb phosphorylation and then promoted c-Myc expression in vitro, we also found c-Myc could suppress miR-15a/16 expression in gastric cancer cell. Finally, we found that miR-15a/16 can simultaneously regulate Cyclin D3 and CDK6 expression as direct target genes.Conclusions: Our findings uncover the Cyclin D3-CDK6/c-Myc/miR-15a/16 feedback loop axis as a pivotal role in the regulation of gastric cancer tumorigenesis, and this regulating axis may provide a potential therapeutic target for gastric cancer treatment.


2019 ◽  
Author(s):  
Shinsuke Tokumoto ◽  
Daisuke Yabe ◽  
Hisato Tatsuoka ◽  
Ryota Usui ◽  
Muhammad Fauzi ◽  
...  

SummaryInducing β-cell proliferation could inhibit diabetes progression. Many factors have been suggested as potential β-cell mitogens, but their impact on β-cell replication has not been confirmed due to the lack of a standardized β-cell proliferation assay. In this study, we developed a novel method that specifically labels replicating β cells and yields more reproducible results than current immunohistochemical assays. We established a mouse line expressing the fluorescent ubiquitination-based cell cycle indicator (Fucci2a) reporter only in β cells through Cre-mediated recombination under the control of the rat insulin promoter (RIP-Cre;Fucci2aR). Three-dimensional imaging of optically cleared pancreas tissue from these mice enabled the quantification of replicating β cells in islets and morphometric analysis of islets following mitogen treatment. Intravital imaging of RIP-Cre;Fucci2aR mice revealed cell cycle progression of β cells. Thus, this novel mouse line is a powerful tool for spatiotemporal analysis of β-cell proliferation in response to mitogen stimulation.


2017 ◽  
Vol 69 (1) ◽  
pp. 71-81
Author(s):  
Qian Xu ◽  
Dong-zhi Yuan ◽  
Sheng Zhang ◽  
Ting Qu ◽  
Shi-mao Zhang ◽  
...  

Uterine stromal cell decidualization is a dynamic physiological process in which cell proliferation, differentiation and apoptosis are orchestrated and occur in a temporal and cell-specific manner. This process is important for successful embryo implantation. Many cell-cycle regulators are involved in decidualization. The protein cyclin G1 is a unique regulator of the cell cycle with dual functions in cell proliferation. It was reported that cyclin G1 is expressed in mouse uterine stromal cells during the period of peri-implantation. To prove the function of cyclin G1 in mouse uterine stromal cells during this period, immunohistochemistry was used to stain mouse uterine tissues on days 4-8 of pregnancy. The results showed obvious spatial and temporal expression of cyclin G1 in uterine stromal cells, and that it is expressed in the cells of the primary decidual zone (PDZ) on day 5 and secondary decidual zone (SDZ) on days 6 and 7, when the stromal cells experienced active proliferation and differentiation was initiated. Applying the decidualization model of cultured primary stromal cells in vitro, we further revealed that the expression of cyclin G1 is associated with decidualization of stromal cells induced by medroxyprogesterone acetate (MPA) and estradiol-17? (E2). RNA interference was used for the knockdown of cyclin G1 in the induced decidual cells. Flow cytometry analysis indicated that the proportion of cells in the S stage was increased, and decreased in the G2/M phase. Our study indicates that cyclin G1, as a negative regulator of the cell cycle, plays an important role in the process of decidualization in mouse uterine stromal cells by inhibiting cell-cycle progression.


2010 ◽  
Vol 298 (2) ◽  
pp. E245-E256 ◽  
Author(s):  
Florence Figeac ◽  
Benjamin Uzan ◽  
Monique Faro ◽  
Noura Chelali ◽  
Bernard Portha ◽  
...  

Wnt/β-catenin signaling is critical for a variety of fundamental cellular processes. Here, we investigated the implication of the Wnt/β-catenin signaling in the in vivo regulation of β-cell growth and regeneration in normal and diabetic rats. To this aim, TCF7L2, the distal effector of the canonical Wnt pathway, was knocked down in groups of normal and diabetic rats by the use of specific antisense morpholino-oligonucleotides. In other groups of diabetic rats, the Wnt/β-catenin pathway was activated by the inhibition of its negative regulator GSK-3β. GSK-3β was inactivated by either LiCl or anti-GSK-3β oligonucleotides. The β-cell mass was evaluated by morphometry. β-cell proliferation was assessed in vivo and in vitro by BrdU incorporation method. In vivo β-cell neogenesis was estimated by the evaluation of PDX1-positive ductal cells and GLUT2-positive ductal cells and the number of β cells budding from the ducts. We showed that the in vivo disruption of the canonical Wnt pathway resulted in the alteration of normal and compensatory growth of β-cells mainly through the inhibition of β-cell proliferation. Conversely, activation of the Wnt pathway through the inhibition of GSK-3β had a significant stimulatory effect on β-cell regeneration in diabetic rats. In vitro, GSK-3β inactivation resulted in the stimulation of β-cell proliferation. This was mediated by the stabilization of β-catenin and the induction of cyclin D. Taken together, our results demonstrate the involvement of the canonical Wnt signaling in the neonatal regulation of normal and regenerative growth of pancreatic β-cells. Moreover, we provide evidence that activation of this pathway by pharmacological maneuvers can efficiently improve β-cell regeneration in diabetic rats. These findings might have potential clinical applications in the regenerative therapy of diabetes.


Author(s):  
Xiaoming Li ◽  
Chengcheng Liu ◽  
Wenwen Qi ◽  
Qiu Meng ◽  
Hui Zhao ◽  
...  

Background: The hypoxia-induced pro-proliferative and anti-apoptotic characteristics of pulmonary arterial endothelial cells (PAECs) play critical roles in pulmonary vascular remodeling and contribute to hypoxic pulmonary arterial hypertension (PAH) pathogenesis. However, the mechanism underlying this hypoxic disease has not been fully elucidated.Methods: Bioinformatics was adopted to screen out the key hypoxia-related genes in PAH. Gain- and loss-function assays were then performed to test the identified hypoxic pathways in vitro. Human PAECs were cultured under hypoxic (3% O2) or normoxic (21% O2) conditions. Hypoxia-induced changes in apoptosis and proliferation were determined by flow cytometry and Ki-67 immunofluorescence staining, respectively. Survival of the hypoxic cells was estimated by cell counting kit-8 assay. Expression alterations of the target hypoxia-related genes, cell cycle regulators, and apoptosis factors were investigated by Western blot.Results: According to the Gene Expression Omnibus dataset (GSE84538), differentiated embryo chondrocyte expressed gene 1-peroxisome proliferative-activated receptor-γ (Dec1-PPARγ) axis was defined as a key hypoxia-related signaling in PAH. A negative correlation was observed between Dec1 and PPARγ expression in patients with hypoxic PAH. In vitro observations revealed an increased proliferation and a decreased apoptosis in PAECs under hypoxia. Furthermore, hypoxic PAECs exhibited remarkable upregulation of Dec1 and downregulation of PPARγ. Dec1 was confirmed to be crucial for the imbalance of proliferation and apoptosis in hypoxic PAECs. Furthermore, the pro-surviving effect of hypoxic Dec1 was mediated through PPARγ inhibition.Conclusion: For the first time, Dec1-PPARγ axis was identified as a key determinant hypoxia-modifying signaling that is necessary for the imbalance between proliferation and apoptosis of PAECs. These novel endothelial signal transduction events may offer new diagnostic and therapeutic options for patients with hypoxic PAH.


2018 ◽  
Vol 2018 ◽  
pp. 1-8 ◽  
Author(s):  
He Hu ◽  
Shanshan Li ◽  
Jianqiao Li ◽  
Chao Huang ◽  
Fang Zhou ◽  
...  

Purpose. Recent research has provided novel insight into the function of fibromodulin (FMOD) in wound healing and angiogenesis. The role of FMOD in initiation of proliferative vitreoretinopathy (PVR) has not been studied. This study investigated the effect of FMOD on human retinal pigment epithelial (RPE) cell, which plays an essential role in the progression of PVR, and the possible mechanisms. Methods. Small interfering (si) RNA-based gene transfer technology was used to decrease FMOD expression and to study its effects on RPEs in vitro. Cell Counting Kit-8 assays, transwells, and flow cytometry analysis were used to measure cell proliferation, migration, cell cycle, and apoptosis. Western blot was used to measure expression of vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), extracellular signal-related kinase 1/2 (ERK1/2), and phosphoinositide 3 kinase (PI3K/AKT). Results. After transfection of RPEs with a FMOD-specific siRNA, cell proliferation and migration were inhibited to the percentage of 65% ± 5% and 39% ± 10%, respectively, compared to the control group. Depletion of FMOD induced cell cycle arrest and apoptosis in RPE cells. Downregulation of VEGF, VEGFR2, and phosphorylated AKT (p-AKT) were detected in transfected RPEs. Conclusion. Depletion of FMOD selectively downregulated the expression of VEGF and VEGFR2 and inhibited the signaling pathway of AKT phosphorylation, which consequently inhibited the proliferation and migration of RPE Cell.


2008 ◽  
Vol 197 (3) ◽  
pp. 543-552 ◽  
Author(s):  
Hannah J Welters ◽  
Alina Oknianska ◽  
Kai S Erdmann ◽  
Gerhart U Ryffel ◽  
Noel G Morgan

In pancreatic β-cells, increased expression of the MODY5 gene product, HNF1β, leads to enhanced rates of apoptosis and altered regulation of the cell cycle, suggesting that control of HNF1β expression may be important for the control of β-cell proliferation and viability. It is unclear how these effects of HNF1β are mediated, but previously we have identified a protein tyrosine phosphatase, (PTP)-BL, as an HNF1β-regulated protein in β-cells and have now studied the role of this protein in INS-1 β-cells. Stably transfected cells were generated, which express either wild-type (WT) or a phosphatase-deficient mutant (PTP-BL-CS) of PTP-BL conditionally under the control of a tetracycline-regulated promoter. Enhanced expression of WT PTP-BL inhibited INS-1 cell growth dose dependently, but this effect was not observed when PTP-BL-CS was expressed. Neither construct altered the rate of apoptosis. PTP-BL has been reported to interact with components of the Wnt signalling pathway, and we observed that addition of exogenous Wnt3a resulted in an increase in cell proliferation and a rise in β-catenin levels, consistent with the operation of this pathway in INS-1 cells. Up-regulation of WT PTP-BL antagonised these responses but PTP-BL-CS failed to inhibit Wnt3a-induced proliferation. The rise in β-catenin caused by Wnt3a was also suppressed by over-expression of HNF1β, suggesting that HNF1β may interact with the Wnt signalling pathway via an increase in PTP-BL levels. We conclude that PTP-BL plays an important role in the regulation of cell cycle progression in pancreatic β-cells, and that it interacts functionally with components of the Wnt signalling pathway.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4149-4149
Author(s):  
Florencia Palacios ◽  
Xiao-Jie Yan ◽  
Jaqueline C. Barrientos ◽  
Jonathan E. Kolitz ◽  
Steven L. Allen ◽  
...  

Abstract Chronic Lymphocytic Leukemia (CLL) is an incurable disease in which most of the tumor cells in the blood are arrested in G0/G1 stages of the cell cycle with only a minimal number displaying proliferative activity. In this regard, our group has found by gene expression profiling (GEP) that the proliferative fraction (PF) of CLL cells is enriched in the intraclonal subset marked by CXCR4dim CD5brite expression. Indeed, this subset differs by more than 1000 genes from the CXCR4brite CD5dim resting fraction (RF). The genes over-expressed in the PF relate to replication and migration as well as regulation of gene expression. One of these genes is Musashi 2 (MSI2). Of note, MSI2 is expressed at the highest levels in IGHV unmutated CLL (U-CLL) clones and their PFs. Normally, MSI2 binds mRNA and blocks translation of proteins, playing an important role in post-transcriptional regulation. In addition, MSI2 has been linked to proliferation of normal and malignant stem cells, tumorigenesis, and poor prognosis. In CLL, high MSI2 mRNA expression has been identified in patients with worse prognosis. Nevertheless, nothing is known about the function of MSI2 in CLL cells. Therefore, we have begun to study the biological role of MSI2 in B-CLL cells and its possible association with B-cell proliferation and CLL disease progression. Fist, we studied MSI2 protein expression by flow cytometry in CD19+ B cells from healthy donors (HD) and CD5+CD19+ cells from CLL patients, observing an up-regulation in CLL compared to HD. Also, we documented higher MSI2 expression in U-CLL compared to IGHV-mutated (M-CLL) CLL as well as HD. Within the leukemic clone, we observed that MSI2 expression was highest in the PF, lower in the intermediate (INT) fraction (defined as CXCR4int CD5int), and much lower in the RF (PF>INT>RF). The PF expressed 40% more MSI2 than the RF, suggesting that the highest amounts of MSI2 protein is in dividing and recently-divided cells of the clone. Since CLL B-cell proliferation occurs in the microenvironment of lymphoid organs, presumably delivered by external signals, we tested whether such signals could stimulate MSI2 expression. After stimulating CLL cells with TLR9 agonist + IL15 + IL2 in vitro MSI2 protein was up-regulated form 0.3 to 2.5 fold. In addition, the increase in MSI2 protein was associated with an enhancement in Ki-67+ cells and in phosphorylation of MAPK/ERK and AKT signaling components, measured by flow cytometry. These results suggest that signals from the microenvironment that induce cell growth and proliferation lead to MSI2 synthesis in CLL B cells. In order to study a possible association between MSI2 expression and cell division, we labeled CLL PBMCs with a dilutable cell tracer, CFSE, and then stimulated them in vitro with TLR9 agonist + IL15 + IL2. These studies indicated that MSI2 protein synthesis was increased in the activated cells and that MSI2 protein levels increased with each cell division. However, it was also clear that this increase was not directly associated to the extent of cell replication as CLL B cells from only 10% of the patients underwent 4 cycles of cell division. Since we observed an increase in MSI2 and Ki-67 expression after stimulation in all patients' clones but did not detect replication of CLL cells in all patients, we studied the effects of in vitro stimulation on cell cycle entry and completion and how this related to MSI2 expression. Experiments using propidium iodide to evaluate DNA content of PBMCs showed that in vitro stimulation increased the percentage of cells in S phase (5-25%) compared to control cells without activation (<5%), whereas only a small fraction of cells entered the M/G2 phases, with or without activation (<1% and <0.5%, respectably) suggesting that only a small portion of the cells completed the cell cycle and divided. Hence, MSI2 synthesis corresponds with DNA replication and not cell division, suggesting that MSI2 could be an important molecule involved in entry into and/or in the early phases of the cell cycle. These results, and the facts that MSI2 plays an important role in post-transcriptional regulation and is associated with cell proliferation and poor prognosis in cancer, suggest that a better understanding of the role of MSI2 in CLL patients will provide clues to understanding the birth and growth of CLL B cells and to identifying and designing new therapeutic strategies for the disease. Disclosures No relevant conflicts of interest to declare.


2018 ◽  
Vol 399 (6) ◽  
pp. 565-576 ◽  
Author(s):  
Brian Lu ◽  
Miguel Munoz-Gomez ◽  
Yasuhiro Ikeda

Abstract Glucokinase (GCK) is crucial to regulating glucose metabolism in the liver and in pancreatic β-cells. There are two major GCK isoforms, hepatic and pancreatic GCKs, which differ only in exon 1. However, the functional differences between the two GCK isoforms remain poorly understood. Here, we used a β-cell-targeted gene transfer vector to determine the impact of isoform-specific GCK overexpression on β-cells in vitro and in vivo. We showed that pancreatic GCK had a nuclear localization signal unique to the pancreatic isoform, facilitating its nuclear distribution in β-cells. Despite the difference in subcellular distribution, overexpression of GCK isoforms similarly enhanced glucose uptake and β-cell proliferation in vitro. Overexpression of hepatic or pancreatic GCK also similarly enhanced β-cell proliferation in normal diet mice without affecting fasting glucose and intraperitoneal glucose tolerance tests (IPGTT). Our further study on human GCK sequences identified disproportional GCK amino acid variants in exon 1, while mutations linked to maturity onset diabetes of the young type 2 (MODY2) were disproportionally found in exons 2 through 10. Our results therefore indicate functional conservation between the two major GCK isoforms despite their distinct subcellular distribution.


Sign in / Sign up

Export Citation Format

Share Document