Phenotypic plasticity: The emergence of cancer stem cells, collective cell migration, and the impact on immune surveillance

Author(s):  
Caterina A.M. La Porta
Author(s):  
Saurav Panicker ◽  
Sivaramakrishnan Venkatabalasubramanian ◽  
Surajit Pathak ◽  
Satish Ramalingam

2021 ◽  
Author(s):  
Ivana Pajic-Lijakovic ◽  
Milan Milivojevic

Although collective cell migration (CCM) is a highly coordinated migratory mode, perturbations in the form of jamming state transitions and vice versa often occur even in 2D. These perturbations are involved in various biological processes, such as embryogenesis, wound healing and cancer invasion. CCM induces accumulation of cell residual stress which has a feedback impact to cell packing density. Density-mediated change of cell mobility influences the state of viscoelasticity of multicellular systems and on that base the jamming state transition. Although a good comprehension of how cells collectively migrate by following molecular rules has been generated, the impact of cellular rearrangements on cell viscoelasticity remains less understood. Thus, considering the density driven evolution of viscoelasticity caused by reduction of cell mobility could result in a powerful tool in order to address the contribution of cell jamming state transition in CCM and help to understand this important but still controversial topic. In addition, five viscoelastic states gained within three regimes: (1) convective regime, (2) conductive regime, and (3) damped-conductive regime was discussed based on the modeling consideration with special emphasis of jamming and unjamming states.


2019 ◽  
Vol 2019 ◽  
pp. 1-16 ◽  
Author(s):  
Qing Xia ◽  
Tao Han ◽  
Pinghua Yang ◽  
Ruoyu Wang ◽  
Hengyu Li ◽  
...  

Background. MicroRNAs (miRNAs) play a critical role in the regulation of cancer stem cells (CSCs). However, the role of miRNAs in liver CSCs has not been fully elucidated. Methods. Real-time PCR was used to detect the expression of miR-miR-28-5p in liver cancer stem cells (CSCs). The impact of miR-28-5p on liver CSC expansion was investigated both in vivo and in vitro. The correlation between miR-28-5p expression and sorafenib benefits in HCC was further evaluated in patient-derived xenografts (PDXs). Results. Our data showed that miR-28-5p was downregulated in sorted EpCAM- and CD24-positive liver CSCs. Biofunctional investigations revealed that knockdown miR-28-5p promoted liver CSC self-renewal and tumorigenesis. Consistently, miR-28-5p overexpression inhibited liver CSC’s self-renewal and tumorigenesis. Mechanistically, we found that insulin-like growth factor-1 (IGF-1) was a direct target of miR-28-5p in liver CSCs, and the effects of miR-28-5p on liver CSC’s self-renewal and tumorigenesis were dependent on IGF-1. The correlation between miR-28-5p and IGF-1 was confirmed in human HCC tissues. Furthermore, the miR-28-5p knockdown HCC cells were more sensitive to sorafenib treatment. Analysis of patient-derived xenografts (PDXs) further demonstrated that the miR-28-5p may predict sorafenib benefits in HCC patients. Conclusion. Our findings revealed the crucial role of the miR-28-5p in liver CSC expansion and sorafenib response, rendering miR-28-5p an optimal therapeutic target for HCC.


2020 ◽  
Vol 11 (2) ◽  
Author(s):  
Josephine Volovetz ◽  
Artem D. Berezovsky ◽  
Tyler Alban ◽  
Yujun Chen ◽  
Adam Lauko ◽  
...  

2020 ◽  
Vol 14 (1) ◽  
Author(s):  
Kandasamy Ashokachakkaravarthy ◽  
Biju Pottakkat

Hepatocellular carcinoma represents one of the most aggressive cancers with high recurrence rates. The high recurrence is a major problem in the management of this disease. Cancer stem cells (CSCs) are often regarded as the basis of cancer recurrence. The anti-proliferative therapy kills the proliferating cells but induces mitotic quiescence in CSCs which remain as residual dormant CSCs. Later on, withdrawal of treatment reactivates the residual CSCs from dormancy to produce new cancer cells. The proliferation of these newly formed cancer cells initiates new tumor formation in the liver leading to tumor recurrence. HCC cells evade the immune surveillance via modulating the key immune cells by alpha feto-protein (AFP) secreted from CSCs or hepatic progenitor cells. This AFP mediated immune evasion assists in establishing new tumors by cancer cells in the liver. In this review, we will summarise the CSC mechanisms of recurrence, mitotic quiescence, dormancy and reactivation of CSCs, metastasis and immune evasion of hepatocellular carcinoma.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 779-779
Author(s):  
Vaia Stavropoulou ◽  
Susanne Kaspar ◽  
Laurent Brault ◽  
Sabine Juge ◽  
Alexander Tzankov ◽  
...  

Abstract Acute myeloid leukemia (AML) is a genetically and clinically heterogeneous disease. Chromosomal translocations causing fusions of the Mixed Lineage Leukemia (MLL) gene are associated with pediatric and adult de novo and therapy-related acute leukemia that are characterized by variable disease outcome. To date, only a limited number of genetic lesions have been implicated in AML disease variability. To address the impact of cellular origin on disease heterogeneity of AMLs, we studied AMLs originating from long-term hematopoietic stem cells (LT-HSCs) and more committed progenitors using a newly established inducible “iMLL-AF9” transgenic mouse model for the t(9:11)(p22;q23) translocation associated MLL-AF9 oncogene. Ex vivo immortalized cells displayed several origin-related growth and drug resistance characteristics and gene expression signatures. Only iMLL-AF9 expressing LT-HSCs formed novel, particularly dispersed colonies, expanded in lineage restrictive stem cell medium and were resistant to genotoxic stress. iMLL-AF9 induction in vivo resulted in fully reversible myelo-monoblastic AML in all animals. Intriguingly, induction of iMLL-AF9 in LT-HSCs caused a particularly aggressive AML phenotype in 15% of recipient mice while the remainder LT-HSCs, as well as short-term HSCs, common myeloid and granulocyte macrophage progenitors induced a more moderate AML. The aggressive LT-HSC-derived AMLs were all characterized by a drastically shorter latency (37 versus 72 days median latency), higher white blood counts, increased invasion capacity and chemo-resistance of leukemic blast, and were associated with expression of genes previously implicated in cell migration, invasion, inflammation and the epithelial-mesenchymal transition (EMT) of solid cancers. shRNA based knock-down experiments demonstrated functional importance of selected candidate genes in cell migration and invasion. Importantly, comparative gene expression analyses between mouse and human revealed that among the genes associated with aggressive AMLs in mice, elevated expression of 66, 11 and 40 human orthologous genes was significantly associated with poor overall survival of t(9;11) (n=21), 11q23-lesion positive, (n=54) and all AMLs (n=662) (p<0.05). Collectively, our data indicates that expression of MLL-AF9 in HSCs results in a particularly aggressive disease driven by expression of common MLL targets and origin-dependent targets previously associated with migration, invasion and EMT of aggressive solid cancers. Remarkably, origin-related genetic signatures associated with the aggressive murine disease revealed a large number of novel MLL-AF9 fusion targets and many highly significant genetic prognostic markers for the overall survival in human AML irrespective of the underlying genetic alterations. Our data experimentally support the previously disputed theory that human AML may also arise from stem and/or oligo-potent progenitors contributing thus to the great heterogeneity of AML including drug resistance and post therapy relapse. Validation of the novel identified target genes in a broader spectrum of human leukemia will facilitate the design of accurate personalized therapeutic interventions. Disclosures No relevant conflicts of interest to declare.


Author(s):  
Martina Mang Leng Lei ◽  
Terence Kin Wah Lee

Cancer stem cells (CSCs) are subpopulations of undifferentiated cancer cells within the tumor bulk that are responsible for tumor initiation, recurrence and therapeutic resistance. The enhanced ability of CSCs to give rise to new tumors suggests potential roles of these cells in the evasion of immune surveillance. A growing body of evidence has described the interplay between CSCs and immune cells within the tumor microenvironment (TME). Recent data have shown the pivotal role of some major immune cells in driving the expansion of CSCs, which concurrently elicit evasion of the detection and destruction of various immune cells through a number of distinct mechanisms. Here, we will discuss the role of immune cells in driving the stemness of cancer cells and provide evidence of how CSCs evade immune surveillance by exerting their effects on tumor-associated macrophages (TAMs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), T-regulatory (Treg) cells, natural killer (NK) cells, and tumor-infiltrating lymphocytes (TILs). The knowledge gained from the interaction between CSCs and various immune cells will provide insight into the mechanisms by which tumors evade immune surveillance. In conclusion, CSC-targeted immunotherapy emerges as a novel immunotherapy strategy against cancer by disrupting the interaction between immune cells and CSCs in the TME.


Sign in / Sign up

Export Citation Format

Share Document