Reactive Oxygen Species Control Hematopoietic Stem Cell-Niche Interaction through the Regulation of N-Cadherin.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 86-86
Author(s):  
Kentaro Hosokawa ◽  
Fumio Arai ◽  
Hiroki Yoshihara ◽  
Keiyo Takubo ◽  
Keisuke Ito ◽  
...  

Abstract Interaction of tissue stem cells with their particular microenvironments, known as stem cell niches, is critical for maintaining the stem cell properties, including self-renewal capacity and the ability of differentiation into single or multiple lineages. We previously reported that the regulation of reactive oxygen species (ROS) is critical for the self-renewal activity of HSCs (Ito et al., Nature 2004, Ito et al., Nat Med 2006). Accumulation of ROS in HSCs induced defects in repopulating ability and in maintenance of quiescence through the activation of p38MAPK and p16Ink4a, indicating that the oxidative stress contributes to exhaustion of the stem cell population. From these findings, we hypothesized that loss of quiescence was associated with the impaired HSC-niche interaction. Oxidative stress may affect not only intrinsic function of HSC but the interaction between HSCs and their niche. In this study, we investigated the effects of ROS in the interaction of HSC with osteoblastic niche. We have found that the side-population (SP) in c-Kit+Sca-1+ Lin− (KSL) fraction was the quiescent HSCs in the osteoblastic niche and KSL-SP cells expressed N-cadherin (NCAD) (Arai et al., Cell 2004), and NCAD mediated cell adhesion induced quiescence of HSCs. Then we analyzed the role of ROS in the maintenance of NCAD-mediated cell-cell adhesion and detachment of HSCs from the niche under the myelosuppressive condition. Administration of 5-FU to mice decreases the dividing cells (non-SP fraction), but not quiescent cells (SP fraction) in BM on day 2. On day 6, HSC population was shifted from SP to non-SP fraction. This event might be associated with the cell cycle activation and detachment of HSCs from the niche. We found that 5-FU treatment transiently increased a level of intracellular ROS in HSCs and downregulated the expression of NCAD in HSCs. Administration of anti-oxidant, N-Acetyl Cysteine (NAC), in 5-FU treated mice maintained NCAD expression in HSCs, suggesting that increased ROS suppressed the expression of NCAD in HSCs. In addition, NAC treatment inhibited the transition of HSCs from SP to non-SP fraction on day 6. Moreover, 5-FU induced upregulation of G-CSFR and Flt3 expression in HSCs on day 2, while NAC treatment inhibited the expressions of growth factor receptors. These data indicated that intracellular ROS was a trigger for the detachment of HSCs from the niche, and inhibition of oxidative stress in HSCs by ant-oxidant preserved NCAD-mediated cell adhesion and blocked cell cycle activation after myelosuppression. Furthermore, these data led us the possibility that normal quiescent HSCs maintains low oxidative stress by existing in the low oxygen environment. To confirm this hypothesis, we evaluated the redox status of fractionated hematopoietic cells (Lin+, Lin−, KSL-non SP, and KSL-SP) by hypoxic marker pimonidazole. And we confirmed that >80 % of KSL-SP cells were pimonidazole positive, suggesting that quiescent HSCs resided in the hypoxic niche. Altogether, our data suggest that regulation of oxidative stress is critical for the interaction between HSCs and BM niche. And osteoblastic niche maintains HSCs in low oxidative stress.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1223-1223
Author(s):  
Cassandra J Reiling ◽  
Dianna Howard ◽  
Christian M Paumi

Abstract Abstract 1223 Hematopoietic stem cells (HSCs) are currently used therapeutically to treat diseases such as leukemia; however, a greater understanding in both the molecular and environmental requirements for HSCs self-renewal will hopefully increase the success rate of these therapeutic uses. It is increasingly evident that reactive oxygen species (ROS) and cellular oxidative stress play an important role in HSC self-renewal and differentiation. Therefore a better understanding of intracellular oxidative stress regulation is critical to increasing transplant success and increasing successful treatment of Leukemia. Our studies take aim at determining the role of multidrug resistance-associated protein 1 (MRP1) in regulating intracellular ROS in HSC's. MRP1 is expressed at slightly higher levels in HSCs than in mature blood cells. The expression pattern of MRP1 in HSCs suggests a possible role in hematopoietic stem cell integrity and differentiation. A major function of MRP1 is to help maintain the oxidative balance of the cell by transporting reduced glutathione (GSH), oxidized glutathione (GSSG), and glutathione-4-hydroxy-nonenal (HNE-SG) conjugates out of the cell. We have hypothesized that MRP1-dependent efflux of GSH and GSSG in HSC increases intracellular reactive oxygen species (ROS) resulting in a loss of self-renewing and increased differentiation of HSCs. In our current studies we have used C57BL/6 FVB and Mrp1-disrupted FVB [Mrp1 (−/−)] mice to investigate the role of MRP1 in HSC differentiation. Our experiments have revealed an increase in LT-HSC and ST-HSC and a corresponding decrease in MPP's in the MRP1 −/− mice as compared to WT matched controls. To determine if MRP1 plays a role in regulating HSC intracellular oxidative stress levels via GSH and GSSG efflux, we measured cellular oxidative stress as a function of DCF-DA and relative GSH levels as a function of glutathione-monochlorobimane (GS-MCB) conjugate fluorescence by flow cytometry. Our studies revealed higher intracellular ROS in WT mice as compared to MRP1 −/− mice and decreased GS-MCB in our WT mice as compared to the MRP1 −/− mice. Taken together the DCF-DA and MCB assays support our hypothesis that MRP1-dependent efflux of GSH/GSSG decreases cellular GSH resulting in higher level of ROS. Our hypothesis is further supported by analysis of lineage marked cells (Lin+), which showed a distinct differentiation pattern between the cells derived from WT and MRP1−/− bone marrow (BM). These studies are supported by results from colony forming cell (CFC) assays. Interestingly, analysis of whole blood did not result in a robust phenotype with regards to leukocytes; however, we found an increase in the number of platelets in MRP1−/− mice when compared to the WT. The increase in platelets is an intriguing result under further investigation. In light of our recent results we have initiated long-term transplant assays to determine if MRP1 does indeed play a role in HSC differentiation and self-renewal. If our hypothesis is true as suggested by our current studies then we expect that expression of MRP1 will negatively effect the ability of HSC's to successfully transplant in the long-term. Overall our data supports our hypothesis that MRP1-dependent efflux of GSH and GSSG in HSC increases intracellular ROS thereby decreasing HSC self-renewing potential and increasing HSC differentiation. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Parthive H. Patel ◽  
Clothilde Pénalva ◽  
Michael Kardorff ◽  
Marianne Roca ◽  
Bojana Pavlović ◽  
...  

Abstract Epithelia are exposed to diverse types of stress and damage from pathogens and the environment, and respond by regenerating. Yet, the proximal mechanisms that sense epithelial damage remain poorly understood. Here we report that p38 signaling is activated in adult Drosophila midgut enterocytes in response to diverse stresses including pathogenic bacterial infection and chemical and mechanical insult. Two upstream kinases, Ask1 and Licorne (MKK3), are required for p38 activation following infection, oxidative stress, detergent exposure and wounding. Ask1-p38 signaling in enterocytes is required upon infection to promote full intestinal stem cell (ISC) activation and regeneration, partly through Upd3/Jak-Stat signaling. Furthermore, reactive oxygen species (ROS) produced by the NADPH oxidase Nox in enterocytes, are required for p38 activation in enterocytes following infection or wounding, and for ISC activation upon infection or detergent exposure. We propose that Nox-ROS-Ask1-MKK3-p38 signaling in enterocytes integrates multiple different stresses to induce regeneration.


2015 ◽  
Vol 2015 ◽  
pp. 1-8 ◽  
Author(s):  
O. G. Lyublinskaya ◽  
Ya. G. Borisov ◽  
N. A. Pugovkina ◽  
I. S. Smirnova ◽  
Ju. V. Obidina ◽  
...  

The present study focuses on the involvement of reactive oxygen species (ROS) in the process of mesenchymal stem cells “waking up” and entering the cell cycle after the quiescence. Using human endometrial mesenchymal stem cells (eMSCs), we showed that intracellular basal ROS level is positively correlated with the proliferative status of the cell cultures. Our experiments with the eMSCs synchronized in the G0phase of the cell cycle revealed a transient increase in the ROS level upon the quiescence exit after stimulation of the cell proliferation. This increase was registered before the eMSC entry to the S-phase of the cell cycle, and elimination of this increase by antioxidants (N-acetyl-L-cysteine, Tempol, and Resveratrol) blocked G1–S-phase transition. Similarly, a cell cycle arrest which resulted from the antioxidant treatment was observed in the experiments with synchronized human mesenchymal stem cells derived from the adipose tissue. Thus, we showed that physiologically relevant level of ROS is required for the initiation of human mesenchymal stem cell proliferation and that low levels of ROS due to the antioxidant treatment can block the stem cell self-renewal.


2021 ◽  
Vol 11 ◽  
Author(s):  
Zhen Cheng ◽  
Shuang Yu ◽  
Weiman He ◽  
Jie Li ◽  
Tianyi Xu ◽  
...  

Thyroid cancer is the most common endocrine malignancy, and its incidence has increased in the past decades. Selenium has been shown to have therapeutic effects against several tumors. However, its role in thyroid cancer and its underlying molecular mechanism remains to be explored. In the present study, we demonstrated that sodium selenite significantly decreased cell viability and induced G0/G1 cell cycle arrest and apoptosis in thyroid cancer cells in a dose-dependent manner. Transcriptomics revealed that sodium selenite induced intracellular reactive oxygen species (ROS) by promoting oxidative phosphorylation. Increased intracellular ROS levels inhibited the AKT/mTOR signaling pathway and upregulated EIF4EBP3. Intracellular ROS inhibition by N-acetylcysteine (NAC) ameliorated the cellular effects of sodium selenite. The in vitro findings were reproduced in xenograft thyroid tumor models. Our data demonstrated that sodium selenite exhibits strong anticancer effects against thyroid cancer cells, which involved ROS-mediated inhibition of the AKT/mTOR pathway. This suggests that sodium selenite may serve as a therapeutic option for advanced thyroid cancer.


2021 ◽  
Author(s):  
◽  
Natelle C H Quek

<p>Natural products offer vast structural and chemical diversity highly sought after in drug discovery research. Saccharomyces cerevisiae makes an ideal model eukaryotic organism for drug mode-of-action studies owing to ease of growth, sophistication of genetic tools and overall homology to higher eukaryotes. Equisetin and a closely related novel natural product, TA-289, are cytotoxic to fermenting yeast, but seemingly less so when yeast actively respire. Cell cycle analyses by flow cytometry revealed a cell cycle block at S-G2/M phase caused by TA-289; previously described oxidative stress-inducing compounds causing cell cycle delay led to further investigation in the involvement of equisetin and TA-289 in mitochondrial-mediated generation of reactive oxygen species. Chemical genomic profiling involving genome-wide scans of yeast deletion mutant strains for TA-289 sensitivity revealed sensitization of genes involved in the mitochondria, DNA damage repair and oxidative stress responses, consistent with a possible mechanism-of-action at the mitochondrion. Flow cytometric detection of reactive oxygen species (ROS) generation caused by TA-289 suggests that the compound may induce cell death via ROS production. The generation of a mutant strain resistant to TA-289 also displayed resistance to a known oxidant, H2O2, at concentrations that were cytotoxic to wild-type cells. The resistant mutant displayed a higher basal level of ROS production compared to the wild-type parent, indicating that the resistance mutation led to an up-regulation of antioxidant capacity which provides cell survival in the presence of TA-289. Yeast mitochondrial morphology was visualized by confocal light microscopy, where it was observed that cells treated with TA-289 displayed abnormal mitochondria phenotypes, further indicating that the compound is acting primarily at the mitochondrion. Similar effects observed with equisetin treatment suggest that both compounds share the same mechanism, eliciting cell death via ROS production in the mitochondrial respiratory chain.</p>


2008 ◽  
Vol 3 (10) ◽  
pp. 823-826 ◽  
Author(s):  
Attila Feher ◽  
Krisztina Ötvös ◽  
Taras P. Pasternak ◽  
Aladár Pettkó-Szandtner

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-19-SCI-19
Author(s):  
Toshio Suda

Abstract Hematopoietic stem cells (HSCs) play a key role in the lifelong maintenance of hematopoiesis through self-renewal and multi-lineage differentiation. Adult HSCs reside within a specialized microenvironment of the bone marrow (BM), called "niche", in which they are maintained in a quiescent state in cell cycle. Most of HSCs within BM show quiescence under the hypoxic niche. Since the loss of HSC quiescence leads to the exhaustion or aging of stem cells through excess cell division, the regulation of quiescence in HSCs is essential for hematopoietic homeostasis. On the other hand, cellular metabolism has been suggested to play a critical role in many biological processes including the regulation of stem cell properties and functions. However, the metabolic condition and adaptation of stem cells remain largely unaddressed. First, we have analyzed HSC metabolism using metabolomics approaches. With step-wise differentiation of stem cells, the cell metabolism associated with each differentiation stage may be different. A feature of quiescent HSCs is their low baseline energy production; quiescent HSCs rely on glycolysis and exhibit low mitochondrial membrane potential (ΔΨm). Likewise, HSCs with a low ΔΨm show higher reconstitution activity in BM hematopoiesis, compared to cells with high ΔΨm. By contrast, upon stress hematopoiesis, HSCs actively divide and proliferate. However, the underlying mechanism for the initiation of HSC division still remains unclear. In order to elucidate the mechanism underlying the transition of cell cycle state in HSCs, we analyzed the change of mitochondria activity in HSCs after BM suppression induced by 5-fluoruracil (5-FU). Upon 5-FU treatment, cycling progenitors are depleted and then quiescent HSCs start to divide. We found that HSCs initiate cell division after exhibiting enhanced ΔΨm, as a result of increased intracellular Ca2+ level. We hypothesize that extracellular adenosine, derived from hematopoietic progenitors, inhibits the calcium influx and mitochondrial metabolism. While further activation of Ca2+-mitochondria pathway led to loss of the stem cell function after cell division, the appropriate suppression of intracellular Ca2+ level by nifedipine, a blocker of L-type voltage-gated Ca2+ channels, prolonged cell division interval in HSCs, and simultaneously achieved both cell division and HSC maintenance (self-renewal division). Thus, our results indicate that the adenosine-Ca2+-mitochondria pathway induces HSC division critically to determine HSC cell fate. Next, to examine the mitochondria oxidative metabolism and purinergic pathways, we introduced the study on a tumor suppressor, Folliculin (FLCN). Conditional deletion of FLCN in HSC compartment using the Mx1-Cre or Vav-iCre system disrupted HSC quiescence and BM homeostasis dependently on the lysosomal stress response induced by TFE3. Together all, we propose that the change in cellular metabolism involving mitochondria is crucial for HSC homeostasis in the stress settings. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 470-470 ◽  
Author(s):  
Kentaro Hosokawa ◽  
Fumio Arai ◽  
Toshio Suda

Abstract Hematopoietic stem cells (HSCs) are responsible for blood cell production throughout the lifetime of individuals. Interaction of HSCs with their particular microenvironments, known as stem cell niches, is critical for maintaining the stem cell properties, including self-renewal capacity and the ability of differentiation into single or multiple lineages. The niche cells produce signaling molecules, extracellular matrix, and cell adhesion molecules, and regulate stem cell fates. Recently, it was clarified that long-term bone marrow (BM) repopulating (LTR) HSCs exist frequently in BM trabecular bone surface, and that N-cadherin + spindle-shaped osteoblasts (OBs) are identified as a major niche component. We found that side-population (SP) in c-Kit +Sca-1 +Lin −(KSL) fraction, which is the quiescent HSC in the OB niche, expressed N-cadherin. Expression of N-cadherin in both of the quiescent HSCs and OBs thought to be essential for an adherens junction between HSCs and OBs in the niche. However, the role of N-cadherin in hematopoiesis is still unclear. In this study, we focused on the function of N-cadherin in the maintenance of the stem cell specific property, such as cell adhesion, quiescence, and LTR-activity. To clarify the function of N-cadherin in hematopoiesis, we prepared the retroviruses expressing wild-type N-cadherin, transfected retroviruses into OP9 stromal cell line and KSL cells, and performed the coculture. After coculture of KSL cells with OP9 cells, long-term culture-initiating cells (LTC-ICs) were maintained on OP9 cells overexpressing WT-N-cadherin (OP9/WT-NCAD). In addition, overexpression of WT-N-cadherin in both of the KSL cells and stromal cells enhanced cobblestone formation. N-cadherin overexpressing KSL cell showed slow-cell division from the single cell, when they cultured on OP9/WT-N-cedherin or N-cadherin-Fc protein coated plates, suggesting that N-cadherin-mediated cell-cell adhesion between HSCs and stromal cells enhances the quiescence of HSCs and keeps HSCs in immature state in in vitro. To clarify the role of N-cadherin in the BM reconstitution ability of HSC, we transfected control-IRES-GFP, WT-N-cadherin-IRES-GFP and N-cedherin/390Δ-IRES-GFP retrovirus into the Ly5.1 BM mononuclear cells and transplanted into lethally irradiated Ly5.2 mice. N-cedherin/390Δ, which is a mutant N-cadherin with a deletion at the extracellular domain, exhibits a dominant negative effect on the activity of endogenous cadherins. Control and WT-N-cadherin expressing cell reconstitute the recipient mice BM, while N-cadherin/390Δ expressing cells did not. It suggests that the adhesion between HSCs and BM niche cell is indispensable for the LTR-activity. In addition, we found that WT-N-cadherin overexpressing HSCs were enriched in the SP fraction after 4 months of BM transplantation, indicating that N-cadherin-mediated cell adhesion induced HSCs in the quiescent and kept quiescent HSCs in the niche. Altogether, these observations suggest that N-cadherin is a critical niche factor for the maintenance of the quiescence and self-renewal activity of HSCs. N-cadherin promotes tight adhesion of HSCs to the niche and keeps HSCs in the quiescent state


2019 ◽  
Vol 20 (6) ◽  
pp. 1508 ◽  
Author(s):  
Chang Lee ◽  
Jongsung Lee ◽  
Myeong Nam ◽  
Youn Choi ◽  
See-Hyoung Park

Tomentosin is a natural sesquiterpene lactone extracted from various plants and is widely used as a medicine because it exhibits essential therapeutic properties. In this study, we investigated the anti-carcinogenic effects of tomentosin in human osteosarcoma MG-63 cells by performing cell migration/viability/proliferation, apoptosis, and reactive oxygen species (ROS) analysis assays. MG-63 cells were treated with various doses of tomentosin. After treatment with tomentosin, MG-63 cells were analyzed using the MTT assay, colony formation assay, cell counting assay, wound healing assay, Boyden chamber assay, zymography assay, cell cycle analysis, FITC Annexin V apoptosis assay, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, western blot analysis, and ROS detection analysis. Our results indicated that tomentosin decreased cell viability and migration ability in MG-63 cells. Moreover, tomentosin induced apoptosis, cell cycle arrest, DNA damage, and ROS production in MG-63 cells. Furthermore, tomentosin-induced intracellular ROS decreased cell viability and induced apoptosis, cell cycle arrest, and DNA damage in MG-63 cells. Taken together, our results suggested that tomentosin exerted anti-carcinogenic effects in MG-63 cells by induction of intracellular ROS.


Sign in / Sign up

Export Citation Format

Share Document