Prolactin, Growth Factors, and Cell Growth*

Author(s):  
Charles S. Nicoll ◽  
Thomas R. Anderson ◽  
Nora J. Hebert ◽  
Sharon M. Russell
Keyword(s):  
Cell ◽  
1978 ◽  
Vol 15 (3) ◽  
pp. 813-822 ◽  
Author(s):  
Martin C. Raff ◽  
Erika Abney ◽  
Jeremy P. Brockes ◽  
Ann Hornby-Smith

PLoS ONE ◽  
2018 ◽  
Vol 13 (11) ◽  
pp. e0207404
Author(s):  
Gokul Raj Kathamuthu ◽  
Kadar Moideen ◽  
Vaithilingam V. Banurekha ◽  
Dina Nair ◽  
R. Sridhar ◽  
...  

2020 ◽  
Vol 76 (12) ◽  
pp. 684-689
Author(s):  
Madej J.A.

The author describes DNA oncoviruses and RNA oncoviruses, their ways of infiltrating the host’s cells, and the possibilities of neoplastic transformation of cells by these microorganisms. The role of protooncogenesis and oncogenesis in both humans and animals is discussed. The transformation of cells by viruses is normally insufficient for oncogenesis; the cells also need to gain “immortality,” which usually requires 4-5 genetic changes (the so-called clonal development of cells), (Fig. 1). Oncoviruses remove suppressor growth factors while enhancing the effects that stimulate cell growth through e.g. hormones, cytokines, or transcription activators. In addition, the author discusses the role of the optimization principle in neogenesis.


2020 ◽  
Author(s):  
Lucía Benítez ◽  
Lucas Barberis ◽  
Luciano Vellón ◽  
Carlos Alberto Condat

Abstract Background: Cancer stem cells are important for the development of many solid tumors. These cells receive promoting and inhibitory signals that depend on the nature of their environment (their niche) and determine cell dynamics. Mechanical stresses are crucial to the initiation and interpretation of these signals. Methods: A two-population mathematical model of tumorsphere growth is used to interpret the results of a series of experiments recently carried out in Tianjin, China, and extract information about the intraspecific and interspecific interactions between cancer stem cell and differentiated cancer cell populations. Results: The model allows us to reconstruct the time evolution of the cancer stem cell fraction, which was not directly measured. We find that, in the presence of stem cell growth factors, the interspecific cooperation between cancer stem cells and differentiated cancer cells induces a positive feedback loop that determines growth, independently of substrate hardness. In a frustrated attempt to reconstitute the stem cell niche, the number of cancer stem cells increases continuously with a reproduction rate that is enhanced by a hard substrate. For growth on soft agar, intraspecific interactions are always inhibitory, but on hard agar the interactions between stem cells are collaborative while those between differentiated cells are strongly inhibitory. Evidence also suggests that a hard substrate brings about a large fraction of asymmetric stem cell divisions. In the absence of stem cell growth factors, the barrier to differentiation is broken and overall growth is faster, even if the stem cell number is conserved. Conclusions: Our interpretation of the experimental results validates the centrality of the concept of stem cell niche when tumor growth is fueled by cancer stem cells. Niche memory is found to be responsible for the characteristic population dynamics observed in tumorspheres. A specific condition for the growth of the cancer stem cell number is also obtained.


Sign in / Sign up

Export Citation Format

Share Document