scholarly journals Cellular senescence or stemness: hypoxia flips the coin

Author(s):  
Daniel Otero-Albiol ◽  
Amancio Carnero

AbstractCellular senescence is a complex physiological state whose main feature is proliferative arrest. Cellular senescence can be considered the reverse of cell immortalization and continuous tumor growth. However, cellular senescence has many physiological functions beyond being a putative tumor suppressive trait. It remains unknown whether low levels of oxygen or hypoxia, which is a feature of every tissue in the organism, modulate cellular senescence, altering its capacity to suppress the limitation of proliferation. It has been observed that the lifespan of mammalian primary cells is increased under low oxygen conditions. Additionally, hypoxia promotes self-renewal and pluripotency maintenance in adult and embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and cancer stem cells (CSCs). In this study, we discuss the role of hypoxia facilitating senescence bypass during malignant transformation and acquisition of stemness properties, which all contribute to tumor development and cancer disease aggressiveness.

2019 ◽  
Vol 20 (9) ◽  
pp. 2254 ◽  
Author(s):  
Ken Nishimura ◽  
Aya Fukuda ◽  
Koji Hisatake

Pluripotent stem cells (PSCs), including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), hold a huge promise for regenerative medicine, drug development, and disease modeling. PSCs have unique metabolic features that are akin to those of cancer cells, in which glycolysis predominates to produce energy as well as building blocks for cellular components. Recent studies indicate that the unique metabolism in PSCs is not a mere consequence of their preference for a low oxygen environment, but is an active process for maintaining self-renewal and pluripotency, possibly in preparation for rapid response to the metabolic demands of differentiation. Understanding the regulatory mechanisms of this unique metabolism in PSCs is essential for proper derivation, generation, and maintenance of PSCs. In this review, we discuss the metabolic features of PSCs and describe the current understanding of the mechanisms of the metabolic shift during reprogramming from somatic cells to iPSCs, in which the metabolism switches from oxidative phosphorylation (OxPhos) to glycolysis.


2021 ◽  
Vol 129 (Suppl_1) ◽  
Author(s):  
Anichavezhi Devendran ◽  
Rasheed Bailey ◽  
Sumanta Kar ◽  
Francesca Stillitano ◽  
Irene Turnbull ◽  
...  

Background: Heart failure (HF) is a complex clinical condition associated with substantial morbidity and mortality worldwide. The contractile dysfunction and arrhythmogenesis related to HF has been linked to the remodelling of calcium (Ca ++ ) handling. Phospholamban (PLN) has emerged as a key regulator of intracellular Ca ++ concentration. Of the PLN mutations, L39X is intriguing as it has not been fully characterized. This mutation is believed to be functionally equivalent to PLN null (KO) but contrary to PLN KO mice, L39X carriers develop a lethal cardiomyopathy (CMP). Our study aims at using induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) from homozygous L39X carriers to elucidate the role of L39X in human pathophysiology. Our plan also involves the characterization of humanized L39X knock-in mice (KM), which we hypothesize will develop a CMP from mis-localization of PLN and disruption of Ca ++ signalling. Methodology and Results: Mononuclear cells from Hom L39X carriers were obtained to generate 11 integration-free patient-specific iPSC clones. The iPSC-CMs were derived using established protocols. Compared to the WT iPSC-CMs, the Hom L39X derived-CMs PLN had an abnormal cytoplasmic distribution and formed intracellular aggregates, with the loss of perinuclear localization. There was also a 70% and 50% reduction of mRNA and protein expression of PLN respectively in L39X compared to WT iPSC-CMs. These findings indicated that L39X PLN is both under-expressed and mis-localized within the cell. To validate this observation in-vivo, we genetically modified FVB mice to harbour the human L39X. Following electroporation, positively transfected mouse embryonic stem cells were injected into host blastocysts to make humanized KM that were subsequently used to generate either a protamine-Cre (endogenous PLN driven expression) or a cardiac TNT mouse (i.e., CMP specific). Conclusion: Our data confirm an abnormal intracellular distribution of PLN, with the loss of perinuclear accumulation and mis-localization, suggestive of ineffective targeting to or retention of L39X. The mouse model will be critically important to validate the in-vitro observations and provides an ideal platform for future studies centred on the development of novel therapeutic strategies including virally delivered CRISPR/Cas9 for in-vivo gene editing and testing of biochemical signalling pathways.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 728-728 ◽  
Author(s):  
Frances J. Harding ◽  
Charles M. Cameron ◽  
Wei-shou Hu ◽  
Dan S. Kaufman

Abstract Blood cell products, such as red blood cells and platelets suitable for transfusion, are attractive as a first generation human embryonic stem cell (hESC) derived therapy. Development of hESC-derived transfusion therapies will require a better understanding of how to control the differentiation of human ES cells into functional blood cells in sufficient quantities for clinical use. HESCs are known to produce mature hematopoietic cells during differentiation as embryoid bodies (EBs). Previously we have demonstrated development of both hematopoietic progenitor cells and more differentiated cell types of myeloid, lymphoid and erythroid lineages from hESC. A four-fold increase in total cell number was achieved when ESC-derived EBs were differentiated in stirred vessels compared to conventional static cultures. Spinner cultures generated EBs more uniform in size and density. Static- and spinner cultivated EBs produced equivalent percentages of hematopoietic progenitors when assayed by surface antigen expression (CD34+, CD31+ and CD45+) and colony forming potential. Hence, overall a greater yield of hematopoietic cells was generated in spinner cultures. Here we incorporate pH and oxygen control into the stirred vessel system in order to closely regulate environmental conditions at levels conducive to hematopoietic differentiation. Hematopoietic potential is compared under hypoxic and normoxic conditions. Hypoxic conditions were confirmed in the EB tissue mass by 2-nitroimidazole (hypoxyprobe) staining. We observed that the cellular response to hypoxia, monitored by the presence of HIF 1α protein, is transient. Peak levels of HIF 1α were detected within 48 hours of low oxygen culture, falling to baseline levels within 7 days. Under more severe conditions the kinetics of the hypoxic response were accelerated, HIF 1α expression peaking and subsiding earlier in cultures held at 1% dissolved oxygen compared to 5% dissolved oxygen. We show that by manipulating dissolved oxygen concentration we are able to influence the progress of differentiation. This can at least partly be attributed to the upregulation of hypoxia inducible genes, including VEGF-A and EPO, under low oxygen conditions. Expression levels of VEGF-A are dependent on dissolved oxygen concentration, being most highly expressed under 1% dissolved oxygen conditions. The transitory nature of the cellular hypoxic response suggests that short exposure to low oxygen conditions may be sufficient to gain the full beneficial impact of hypoxic signalling on hematopoietic cell generation without decreases in cell proliferation and increase in cell death associated with extended oxygen deprivation. We propose that control of culture parameters such as dissolved oxygen in conjunction with cytokines can specify the cellular microenvironment within EB to yield robust levels of hematopoietic progenitors. This work demonstrates proof-of-principle for hematopoietic cell production from human embryonic stem cells in a scaleable bioreactor system.


Biomolecules ◽  
2020 ◽  
Vol 10 (12) ◽  
pp. 1614
Author(s):  
Paulina Podkalicka ◽  
Jacek Stępniewski ◽  
Olga Mucha ◽  
Neli Kachamakova-Trojanowska ◽  
Józef Dulak ◽  
...  

Inadequate supply of oxygen (O2) is a hallmark of many diseases, in particular those related to the cardiovascular system. On the other hand, tissue hypoxia is an important factor regulating (normal) embryogenesis and differentiation of stem cells at the early stages of embryonic development. In culture, hypoxic conditions may facilitate the derivation of embryonic stem cells (ESCs) and the generation of induced pluripotent stem cells (iPSCs), which may serve as a valuable tool for disease modeling. Endothelial cells (ECs), multifunctional components of vascular structures, may be obtained from iPSCs and subsequently used in various (hypoxia-related) disease models to investigate vascular dysfunctions. Although iPSC-ECs demonstrated functionality in vitro and in vivo, ongoing studies are conducted to increase the efficiency of differentiation and to establish the most productive protocols for the application of patient-derived cells in clinics. In this review, we highlight recent discoveries on the role of hypoxia in the derivation of ESCs and the generation of iPSCs. We also summarize the existing protocols of hypoxia-driven differentiation of iPSCs toward ECs and discuss their possible applications in disease modeling and treatment of hypoxia-related disorders.


2020 ◽  
Vol 295 (30) ◽  
pp. 10245-10254
Author(s):  
Soujanya Vinayagamurthy ◽  
Akansha Ganguly ◽  
Shantanu Chowdhury

Telomeres comprise specialized nucleic acid–protein complexes that help protect chromosome ends from DNA damage. Moreover, telomeres associate with subtelomeric regions through looping. This results in altered expression of subtelomeric genes. Recent observations further reveal telomere length–dependent gene regulation and epigenetic modifications at sites spread across the genome and distant from telomeres. This regulation is mediated through the telomere-binding protein telomeric repeat–binding factor 2 (TRF2). These observations suggest a role of telomeres in extra-telomeric functions. Most notably, telomeres have a broad impact on pluripotency and differentiation. For example, cardiomyocytes differentiate with higher efficacy from induced pluripotent stem cells having long telomeres, and differentiated cells obtained from human embryonic stem cells with relatively long telomeres have a longer lifespan. Here, we first highlight reports on these two seemingly distinct research areas: the extra-telomeric role of telomere-binding factors and the role of telomeres in pluripotency/stemness. On the basis of the observations reported in these studies, we draw attention to potential molecular connections between extra-telomeric biology and pluripotency. Finally, in the context of the nonlocal influence of telomeres on pluripotency and stemness, we discuss major opportunities for progress in molecular understanding of aging-related disorders and neurodegenerative diseases.


Stem Cells ◽  
2013 ◽  
Vol 31 (4) ◽  
pp. 682-692 ◽  
Author(s):  
Yun-Shen Chan ◽  
Jonathan Göke ◽  
Xinyi Lu ◽  
Nandini Venkatesan ◽  
Bo Feng ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (8) ◽  
pp. 2094-2104 ◽  
Author(s):  
Hyung Joon Joo ◽  
Honsoul Kim ◽  
Sang-Wook Park ◽  
Hyun-Jai Cho ◽  
Hyo-Soo Kim ◽  
...  

Abstract Angiopoietin-1 (Ang1) plays a crucial role in vascular and hematopoietic development, mainly through its cognate receptor Tie2. However, little is known about the precise role of Ang1 in embryonic stem cell (ESC) differentiation. In the present study, we used COMP-Ang1 (a soluble and potent variant of Ang1) to explore the effect of Ang1 on endothelial and hematopoietic differentiation of mouse ESCs in an OP9 coculture system and found that Ang1 promoted endothelial cell (EC) differentiation from Flk-1+ mesodermal precursors. This effect mainly occurred through Tie2 signaling and was altered in the presence of soluble Tie2-Fc. We accounted for this Ang1-induced expansion of ECs as enhanced proliferation and survival. Ang1 also had an effect on CD41+ cells, transient precursors that can differentiate into both endothelial and hematopoietic lineages. Intriguingly, Ang1 induced the preferential differentiation of CD41+ cells toward ECs instead of hematopoietic cells. This EC expansion promoted by Ang1 was also recapitulated in induced pluripotent stem cells (iPSCs) and human ESCs. We successfully achieved in vivo neovascularization in mice by transplantation of ECs obtained from Ang1-stimulated ESCs. We conclude that Ang1/Tie2 signaling has a pivotal role in ESC-EC differentiation and that this effect can be exploited to expand EC populations.


2021 ◽  
Vol 2021 ◽  
pp. 1-13
Author(s):  
Weiwei Sun ◽  
Bin Zhang ◽  
Qingli Bie ◽  
Na Ma ◽  
Na Liu ◽  
...  

The biological role of RNA methylation in stem cells has attracted increasing attention. Recent studies have demonstrated that RNA methylation plays a crucial role in self-renewal, differentiation, and tumorigenicity of stem cells. In this review, we focus on the biological role of RNA methylation modifications including N6-methyladenosine, 5-methylcytosine, and uridylation in embryonic stem cells, adult stem cells, induced pluripotent stem cells, and cancer stem cells, so as to provide new insights into the potential innovative treatments of cancer or other complex diseases.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Yuanjie Ding ◽  
Yuanchun Yao ◽  
Xingmu Gong ◽  
Qi Zhuo ◽  
Jinhua Chen ◽  
...  

AbstractThe Jumonji domain-containing protein-3 (JMJD3) is a histone demethylase that regulates the trimethylation of histone H3 on lysine 27 (H3K27me3). H3K27me3 is an important epigenetic event associated with transcriptional silencing. JMJD3 has been studied extensively in immune diseases, cancer, and tumor development. There is a comprehensive epigenetic transformation during the transition of embryonic stem cells (ESCs) into specialized cells or the reprogramming of somatic cells to induced pluripotent stem cells (iPSCs). Recent studies have illustrated that JMJD3 plays a major role in cell fate determination of pluripotent and multipotent stem cells (MSCs). JMJD3 has been found to enhance self-renewal ability and reduce the differentiation capacity of ESCs and MSCs. In this review, we will focus on the recent advances of JMJD3 function in stem cell fate.


Sign in / Sign up

Export Citation Format

Share Document