scholarly journals Mutational spectrum of Barrett’s stem cells suggests paths to initiation of a precancerous lesion

2016 ◽  
Vol 7 (1) ◽  
Author(s):  
Yusuke Yamamoto ◽  
Xia Wang ◽  
Denis Bertrand ◽  
Florian Kern ◽  
Ting Zhang ◽  
...  

Abstract The precancerous lesion known as Barrett’s oesophagus can evolve to oesophageal adenocarcinoma in decades-long processes of regenerative growth. Here we report the isolation and propagation of distinct, patient-matched stem cells of Barrett’s, gastric and oesophageal epithelia that yield divergent tumour types following in vitro transformation and xenografting. Genomic analyses reveal a broad mutational spectrum unique to Barrett’s stem cells that likely reflects their risk for oncogenesis. Remarkably, 25% of cases show no cancer-related genomic changes, suggesting that Barrett’s initiates without driver mutations. Most cases, however, sustain patterns of deletions almost identical to adenocarcinoma though tumour-associated gene amplifications were absent. Notably, those suspected of low-grade dysplasia have p53 mutations or undergo amplifications of proto-oncogenes and receptor tyrosine kinases, implicating these events in lethal transitions. Our findings suggest paths for the initiation and progression of Barrett’s and define a discrete stem cell underlying its regenerative growth whose eradication could prevent oesophageal adenocarcinoma.


2020 ◽  
Vol 21 (12) ◽  
pp. 4199
Author(s):  
Metka Novak ◽  
Miha Koprivnikar Krajnc ◽  
Barbara Hrastar ◽  
Barbara Breznik ◽  
Bernarda Majc ◽  
...  

The chemokine CCL5/RANTES is a versatile inflammatory mediator, which interacts with the receptor CCR5, promoting cancer cell interactions within the tumor microenvironment. Glioblastoma is a highly invasive tumor, in which CCL5 expression correlates with shorter patient survival. Using immunohistochemistry, we identified CCL5 and CCR5 in a series of glioblastoma samples and cells, including glioblastoma stem cells. CCL5 and CCR5 gene expression were significantly higher in a cohort of 38 glioblastoma samples, compared to low-grade glioma and non-cancerous tissues. The in vitro invasion of patients-derived primary glioblastoma cells and glioblastoma stem cells was dependent on CCL5-induced CCR5 signaling and is strongly inhibited by the small molecule CCR5 antagonist maraviroc. Invasion of these cells, which was enhanced when co-cultured with mesenchymal stem cells (MSCs), was inhibited by maraviroc, suggesting that MSCs release CCR5 ligands. In support of this model, we detected CCL5 and CCR5 in MSC monocultures and glioblastoma-associated MSC in tissue sections. We also found CCR5 expressing macrophages were in close proximity to glioblastoma cells. In conclusion, autocrine and paracrine cross-talk in glioblastoma and, in particular, glioblastoma stem cells with its stromal microenvironment, involves CCR5 and CCL5, contributing to glioblastoma invasion, suggesting the CCL5/CCR5 axis as a potential therapeutic target that can be targeted with repositioned drug maraviroc.



Blood ◽  
1997 ◽  
Vol 89 (12) ◽  
pp. 4317-4326 ◽  
Author(s):  
Michihiro Yano ◽  
Atsushi Iwama ◽  
Hitoshi Nishio ◽  
Junko Suda ◽  
Goro Takada ◽  
...  

Abstract Two highly related receptor tyrosine kinases, TIE and TEK, comprise a family of endothelial cell-specific kinase. We established monoclonal antibodies against them and performed detailed analyses on their expression and function in murine hematopoietic stem cells (HSCs). TIE and TEK were expressed on 23.7% and 33.3% of lineage marker-negative, c-Kit+ and Sca-1+ (Lin− c-Kit+ Sca-1+) HSCs that contain the majority of day-12 colony-forming units-spleen (CFU-S) and long-term reconstituting cells, but not committed progenitor cells. Lin− c-Kit+ Sca-1+ cells were further divided by the expression of TIE and TEK. TIE+ and TEK+ HSCs as well as each negative counterpart contained high proliferative potential colony-forming cells and differentiated into lymphoid and myeloid progenies both in vitro and in vivo. However, day-12 CFU-S were enriched in TIE+ and TEK+ HSCs. Our findings define TIE and TEK as novel stem cell marker antigens that segregate day-12 CFU-S, and provide evidence of novel signaling pathways that are involved in the functional regulation of HSCs at a specific stage of differentiation, particularly of day-12 CFU-S.





2021 ◽  
Author(s):  
Cassandra Verheul ◽  
Ioannis Ntafoulis ◽  
Trisha Kers ◽  
Youri Hoogstrate ◽  
Pier Mastroberardino ◽  
...  

Background: Mutations of the isocitrate dehydrogenase (IDH) gene occur in over 80% of low-grade gliomas and secondary glioblastomas. Despite considerable efforts, endogenous in vitro IDH-mutated glioma models remain scarce. Availability of these models is key for the development of new therapeutic interventions. Methods: Cell cultures were established from fresh tumor material and expanded in serum-free culture media. D-2-Hydroxyglutarate levels were determined by mass-spectrometry. Genomic and transcriptomic profiling were carried out on the Illumina Novaseq platform, methylation profiling was performed with the Infinium MethylationEpic BeadChip array. Mitochondrial respiration was measured with the Seahorse XF24 Analyzer. Drug screens were performed with an NIH FDA-approved anti-cancer drug set and two IDH-mutant specific inhibitors. Results: A set of twelve patient-derived IDHmt cell cultures were established. We confirmed high concordance in driver mutations, copy number and methylation profiles between the tumors and derived cultures. Homozygous deletion of CDKN2A/B was observed in all cultures. IDH-mutant cultures had lower mitochondrial reserve capacity. IDH-mutant specific inhibitors did not affect cell viability or global gene expression. Screening of 107 FDA-approved anti-cancer drugs identified nine compounds with potent activity against IDHmt gliomas, including three compounds with favorable pharmacokinetic characteristics for CNS penetration: teniposide, omacetaxine mepesuccinate, and marizomib. Conclusions: Our twelve IDH-mutant cell cultures show high similarity to the parental tissues and offer a unique tool to study the biology and drug sensitivities of high-grade IDHmt gliomas in vitro. Our drug screening studies reveal lack of sensitivity to IDHmt inhibitors, but sensitivity to a set of nine available anti-cancer agents.



Author(s):  
Cassandra Verheul ◽  
Ioannis Ntafoulis ◽  
Trisha V Kers ◽  
Youri Hoogstrate ◽  
Pier G Mastroberardino ◽  
...  

Abstract Background Mutations of the isocitrate dehydrogenase (IDH) gene occur in over 80% of low-grade gliomas and secondary glioblastomas. Despite considerable efforts, endogenous in vitro IDH-mutated glioma models remain scarce. Availability of these models is key for the development of new therapeutic interventions. Methods Cell cultures were established from fresh tumor material and expanded in serum-free culture media. D-2-Hydroxyglutarate levels were determined by mass-spectrometry. Genomic and transcriptomic profiling were carried out on the Illumina Novaseq platform, methylation profiling was performed with the Infinium MethylationEpic BeadChip array. Mitochondrial respiration was measured with the Seahorse XF24 Analyzer. Drug screens were performed with an NIH FDA-approved anti-cancer drug set and two IDH-mutant specific inhibitors. Results A set of twelve patient-derived IDHmt cell cultures was established. We confirmed high concordance in driver mutations, copy number and methylation profiles between the tumors and derived cultures. Homozygous deletion of CDKN2A/B was observed in all cultures. IDH-mutant cultures had lower mitochondrial reserve capacity. IDH-mutant specific inhibitors did not affect cell viability or global gene expression. Screening of 107 FDA-approved anti-cancer drugs identified nine compounds with potent activity against IDHmt gliomas, including three compounds with favorable pharmacokinetic characteristics for CNS penetration: teniposide, omacetaxine mepesuccinate, and marizomib. Conclusions Our twelve IDH-mutant cell cultures show high similarity to the parental tissues and offer a unique tool to study the biology and drug sensitivities of high-grade IDHmt gliomas in vitro. Our drug screening studies reveal lack of sensitivity to IDHmt inhibitors, but sensitivity to a set of nine available anti-cancer agents.



2021 ◽  
Author(s):  
Yuelong Jiang ◽  
Long Liu ◽  
Zhifeng Li ◽  
Liying Feng ◽  
Zhijuan Lin ◽  
...  

Abstract Leukemia stem cells (LSCs) remain as the critical barrier to cure of acute myeloid leukemia (AML) due to its chemoresistance. Here, we explore the role anlotinib -a multiple tyrosine kinases inhibitor in killing LSCs and regulating the chemoresistance. We found anlotinib could effectively induce apoptosis of LSC-like cells as well as primary CD34+ AML LSCs while sparing the normal mononuclear cells in vitro. The anti-leukemia activity of anlotinib was also confirmed in the mice model with Kasumi-1 cells; we further found that anlotinib could impair the regeneration capacity of LSCs in the patient-derived leukemia xenograft mouse model. Mechanistically, anlotinib could not only inhibit phosphorylation of c-kit and JAK2 /STAT3 and STAT5 but also downregulate STAT3 and STAT5 expression. In addition, anlotinib downregulated the anti-apoptotic protein Bcl-2 and Bcl-xl and upregulated Bax, thereby enhancing the sensitivity of LSCs to idarubicin in vitro. In conclusion, our results demonstrated anlotinib showed anti-LSCs activity and enhanced the chemosensitivity via inhibiting JAK2/STAT signaling in preclinical study and provided a rational basis for combinatory strategies that invoving anlotinib and idarubicin.



2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii218-ii218
Author(s):  
Sanjay Singh ◽  
Maxime Munyeshyaka ◽  
Joy Gumin ◽  
Jing Yang ◽  
Daniel Ledbetter ◽  
...  

Abstract Glioblastomas (GBM) exhibit high proliferative index, areas of necrosis, high vascularization, and are highly invasive to normal brain tissues. The most common and lethal form of GBMs are primary GBMs, with no prior clinical history. Whereas, secondary GBMs arise from low-grade gliomas and are associated with IDH1 mutation. Pre-clinical studies of GBM largely depend on patient-derived GBM stem-like cells (GSCs) in vitro and in vivo as orthotopic xenografts. Cerebral organoids (COs) derived from induced pluripotent stem cells can serve as allogenic in vitro model systems to study interactions between normal brain and GSCs. COs have been shown to harbor neural stem cells and their differentiated progenies as well as microglia within distinct niches. Here, we co-cultured 45 day-old COs and MDA-GSCs lines representing mesenchymal sub-group (M-MDA-GSC), classical sub-group (C-MDA-GSC), and IDH1 mutant (IDH1R132H-MDA-GSC). MDA-GSCs stably express fluorescent proteins and is used to track GSCs within COs. These GSC bearing COs were fixed, embedded, sectioned, immuno-stained, and imaged by confocal microscope. There was a positive correlation between GSC numbers in allografted niche and invasion into COs as measured from the edge of organoid, M-MDA-GSC (R2=0.99; 0.89μm/cell), C-MDA-GSC (R2=0.92; 0.66μm/cell), and IDH1R132H-MDA-GSC (R2=0.89; 0.5μm/cell). Additionally, M-MDA-GSCs had significantly high percentage of Ki67+ve invasive cells (24%) in comparison to C-MDA-GSCs (5.1%; p=0.0057). As a measure of interaction of MDA-GSC with normal cells, we assessed proximity of IBA1+ve microglia in GSC niche within organoids and show that M-MDA-GSC and IDH1R132H-MDA-GSC highly co-localized with IBA1+ve microglia on day12 of co-culture. In conclusion, our cerebral organoid-based allograft study shows that mesenchymal GSCs (M-MDA-GSC) are most invasive whereas IDH1 mutant GSCs (IDH1R132H-MDA-GSC) are least invasive. C-MDA-GSCs are least proliferative while invading into normal COs. Uniqueness of CO based allograft system is highlighted by observed similarity between M-MDA-GSC and IDH1R132H-MDA-GSC for their potential to attract IBA1+ve microglia.



2020 ◽  
Vol 40 (6) ◽  
Author(s):  
Sunhye Shin ◽  
Asma S. El-Sabbagh ◽  
Brandon E. Lukas ◽  
Skylar J. Tanneberger ◽  
Yuwei Jiang

Abstract Adipose tissue, the storage of excessive energy in the body, secretes various proteins called adipokines, which connect the body’s nutritional status to the regulation of energy balance. Obesity triggers alterations of quantity and quality of various types of cells that reside in adipose tissue, including adipose stem cells (ASCs; referred to as adipose-derived stem/stromal cells in vitro). These alterations in the functionalities and properties of ASCs impair adipose tissue remodeling and adipose tissue function, which induces low-grade systemic inflammation, progressive insulin resistance, and other metabolic disorders. In contrast, the ability of ASCs to recruit new adipocytes when faced with caloric excess leads to healthy adipose tissue expansion, associated with lower amounts of inflammation, fibrosis, and insulin resistance. This review focuses on recent advances in our understanding of the identity of ASCs and their roles in adipose tissue development, homeostasis, expansion, and thermogenesis, and how these roles go awry in obesity. A better understanding of the biology of ASCs and their adipogenesis may lead to novel therapeutic targets for obesity and metabolic disease.



2019 ◽  
Vol 64 (1) ◽  
pp. 5-14 ◽  
Author(s):  
Ю. Семочкина ◽  
Yu. Semochkina ◽  
А. Родина ◽  
A. Rodina ◽  
Е. Москалева ◽  
...  

Purpose: To study the possibility of malignant transformation of control and irradiated mesenchymal stromal stem cells (MSC) from the bone marrow (BM) and brain (BR) and from the adipose tissue (AT) of mice and some cytokines secretion after mixed γ,neutron (γ, n) irradiation and γ-irradiation. Material and methods: MSCs were isolated and cultured according to generally accepted protocols. γ, n-irradiation was carried out by a collimated beam of neutrons and gamma rays at a special station of the nuclear reactor IR-8. MSCs were irradiated at the 29th passage at doses of 0.05; 0.5 and 2 Gy, were cultured for 10 passages and transplanted subcutaneously 1×106 cells to C57BL/6 syngeneic mice. MSCs AT were irradiated at the facility GUT-200M (60Co) at doses 1–6 Gy. The level of cytokines in the culture medium of MSC was measured by an ELISA. Results: A decrease in RBE was observed after radiation dose increasing from 0.5 to 4.0 Gy. The maximum of RBE for all MSCs, equal to 5.5, was observed at a dose of 0.5 Gy. After the dose increasing to 2 Gy an average RBE decreased to 2.5, and at dose 4.0 Gy RBE it was 2.0. Tumors were detected after 5 months after transplantation into syngeneic mice of MSC BM irradiated at doses of 0.05; 0.5 and 2 Gy. After transplantation of control MSCs BM and of control and irradiated MSCs BR and MSC AT, no tumors were detected. After subcutaneous injection of γ-irradiated at doses of 0.1; 1 and 6 Gy MSC AT, unlike MSCs BM, no tumors were detected. Histological examination of tumors revealed signs of a low-grade fibrosarcoma with active proliferation and a high degree of malignancy. Tumors contained inclusions from the derivatives of several tissues of mesenchymal origin – muscular, fatty, cartilaginous and bone. In the case of a tumor that developed after transplantation of MSCs BM exposed to γ,n-radiation at a dose of 0.05 Gy, the contact metastasis was detected in the shoulder with the penetration of tumor cells between the muscle fibers. From the tumors, the mouse fibrosarcoma cell lines were obtained. The highest level of cytokines VEGF, HGF and IL6was found in the culture medium of MSC AT. The level of TGFβ secretion was practically the same in all studied MSCs. After γ,n-irradiation an increase of VEGF secretion in MSC BM, a decrease of IL6 secretion in MSC BM and MSC BR, and an increase in its secretion in MSC AT were detected. Conclusions: The obtained results testify the high sensitivity of MSC BM to malignant transformation after ionizing irradiation and the much higher resistance of mouse MSC BR and MSC AT. The mechanisms of these differences are yet not known. The highest level of cytokines VEGF, HGF and IL6 was found in the culture medium of MSC AT. After the action of γ,n-radiation, as well as after the action of γ-radiation, the secretion profile of the investigated cytokines was changed, depending both on the dose and on the type of radiation.



2019 ◽  
Vol 40 (Supplement_1) ◽  
Author(s):  
M C Vinci ◽  
V Vigorelli ◽  
A Raucci ◽  
S Genovese ◽  
G Pompilio

Abstract Background/Introduction Diabetes is characterized by a chronic low-grade inflammatory status mediated by cellular senescence and alterations of the circulating cytokine profile and of innate immune system cell components. An abnormal expansion of intermediate (CD14++CD16+) and non-classical (CD14+CD16++) monocyte subpopulations with highly inflammatory and senescent-associated secretory phenotype (SASP) have been observed in T2DM patients with cardiovascular (CV) complications. It is unknown whether CD34+ hematopoietic stem cells (HSCs), players of CV prognosis, are involved in this process. Purpose To assess whether hyperglycemia might induce a pro-inflammatory priming of hematopoietic CD34+ HSCs that through senescence and acquisition of SASP skew their myeloid differentiation into more aggressive monocyte populations. Methods CD34+ cells were purified from cord blood of healthy donors and expanded in normal-glucose (NG; with 30 mM mannitol for osmotic control) or high-glucose (HG; 30 mM) serum-free medium plus cytokines. The cell were counted after 10, 20 and 30 days. The expression of p27, p21 RELA/p65, IL6, TNFα genes and telomere length was assessed by qPCR and secreted cytokines by ELISA. Apoptosis, ROS and monocyte subpopulations were evaluated by flow cytometry after Annexin V, CellRox and CD14/CD16 staining respectively. Results CD34+ HSCs cultured in HG (HG-CD34+) displayed a significant proliferation impairment when compared to their osmotic control (NG-CD34+). This loss of glucose tolerance was associated with a significant increase in mitochondrial ROS production (n=6; p≤0.01) without induction of apoptosis as showed by flow cytometry analysis for Annexin V. Moreover, qPCR assay revealed a significant telomere shortening (n=4; p≤0.05) and up-regulation of cyclin-dependent kinase inhibitors p27 and p21 in HG-CD34+cells (n=13; p≤0.05) along with an enhanced expression and secretion of TNFα (n=9; p≤0.05) and IL6 (n=10; p≤0.05) in HG-CD34+ when compared with NG-CD34+. SASP phenotype in HG-CD34+ was associated to a significant up-regulation of RELA/p65 gene in HG-CD34+ when compared with NG-CD34+ (n=8; p≤0.05). Furthermore, we found that in vitro HG-CD34+ differentiation into myeloid lineage generated higher levels of pro-inflammatory intermediate (n=3; p≤0.05) and non-classical (n=3; p≤0.01) monocyte subsets when compared with the normoglycemic counterpart. Conclusion(s) These data suggest that HG exposure primes HSCs myeloid differentiation towards inflammatory and senescent monocyte subpopulations. Acknowledgement/Funding This work was supported by Ricerca Finalizzata, Ministero della Salute [PE-2011-02348537]



Sign in / Sign up

Export Citation Format

Share Document