scholarly journals Heme oxygenase‐1 deficiency triggers exhaustion of hematopoietic stem cells

EMBO Reports ◽  
2019 ◽  
Vol 21 (2) ◽  
Author(s):  
Krzysztof Szade ◽  
Monika Zukowska ◽  
Agata Szade ◽  
Witold Nowak ◽  
Izabella Skulimowska ◽  
...  
Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1353-1353
Author(s):  
Yu-An Cao ◽  
Amy J. Wagers ◽  
Holger Karsunky ◽  
Hui Zhao ◽  
Robert Reeves ◽  
...  

Abstract Hematopoietic stem cells (HSCs) must be able to balance their self-renewal and differentiation activities in order to preserve their compartment in response to hematopoietic insults for efficient and life-long hematopoiesis while ensuring sufficient blood production to meet the increased hematopoietic demand. Mechanism(s) that regulate this balance during stress hematopoiesis remain to be fully understood. Heme oxygenase 1 (HO-1) is an important stress-inducible protein and a key enzyme of heme degradation that produces iron, bilirubin, and carbon monoxide (CO). CO is a gaseous regulator of cellular responses to a variety of insults. HO-1 deficiency results in reduced stress defense due to inadequate levels of its products. We report here that mice lacking one allele of HO-1 (HO-1−/+ mice) recovered more rapidly from myelotoxic injury and that sufficient HO-1−/+ bone marrow (BM) transplants engrafted lethally irradiated hosts with accelerated kinetics. These effects of HO-1 deficiency on regenerative hematopoiesis were associated with accelerated cell division of lineage-depleted (lin-) Sca-1+ cells. However, in other experimental settings, the converse can be seen in HO-1 deficiency. Repeated treatment of HO-1−/+ mice with 5-FU significantly reduced the number of HSCs in the marrow, relative to wild-type animals, and following adoptive transfer, the HO-1−/+ HSC compartment was smaller compared to that of HO-1+/+ in recipient animals. As a consequence, limited numbers of HO-1−/+ BM cells failed to provide adequate radio-protection of lethally irradiated recipients, and HO-1−/+ HSCs showed significantly compromised capacity to serially repopulate myeloablated hosts. We also noted that expression levels of the cyclin-dependent kinase inhibitor p21Cip/WAF1 were lower in lin- HO-1−/+ BM cells in contrast to that in HO-1+/+ cells under steady-state conditions, and that p38 mitogen-activated protein kinase (p38MAPK) was insufficiently activated in HO-1−/+ multipotent progenitors (MPPs) after heme challenge, compared to that in wild-type MPPs. We propose a model wherein HO-1 plays an intracellular regulatory and cyto-protective role in maintaining HSC compartment, especially during stress hematopoiesis. It follows that reduced levels of p38MAPK activation, presumably due to insufficient production of CO when HO-1 is deficient, result in accelerated proliferation of MPPs. This would potentially skew the balance between self-renewal and differentiation of HSC and deplete the primitive cell compartment, leading to ultimate hematopoietic exhaustion.


2018 ◽  
Author(s):  
Krzysztof Szade ◽  
Monika Zukowska ◽  
Agata Szade ◽  
Witold N Nowak ◽  
Maciej Ciesla ◽  
...  

While intrinsic changes in aging hematopoietic stem cells (HSCs) are well-characterized, it remains unclear how hematopoietic niche affects HSC aging. Here, we demonstrate that cells in the niche - endothelial cells (ECs) and CXCL12-abundant reticular cells (CARs) - highly express the heme-degrading enzyme, heme oxygenase 1 (HO-1), but then decrease its expression with age. RNA-sequencing shows that ECs and CARs from HO-1-deficient animals (HO-1-/-) produce less hematopoietic factors. Consequently, HSCs from young HO-1-/- animals lose quiescence and regenerative potential. Young HO-1-/- HSCs exhibit features of premature aging on the transcriptional and functional level. HO-1+/+ HSCs transplanted into HO-1-/- recipients exhaust their regenerative potential early and do not reconstitute secondary recipients. In turn, transplantation of HO-1-/- HSCs to the HO-1+/+ recipients recovers the regenerative potential of HO-1-/- HSCs and reverses their transcriptional alterations. Thus, HSC-extrinsic activity of HO-1 prevents HSCs from premature aging and may restore the function of aged HSCs.


Blood ◽  
2008 ◽  
Vol 112 (12) ◽  
pp. 4494-4502 ◽  
Author(s):  
Yu-An Cao ◽  
Amy J. Wagers ◽  
Holger Karsunky ◽  
Hui Zhao ◽  
Robert Reeves ◽  
...  

Abstract An effective response to extreme hematopoietic stress requires an extreme elevation in hematopoiesis and preservation of hematopoietic stem cells (HSCs). These diametrically opposed processes are likely to be regulated by genes that mediate cellular adaptation to physiologic stress. Herein, we show that heme oxygenase-1 (HO-1), the inducible isozyme of heme degradation, is a key regulator of these processes. Mice lacking one allele of HO-1 (HO-1+/−) showed accelerated hematopoietic recovery from myelotoxic injury, and HO-1+/− HSCs repopulated lethally irradiated recipients with more rapid kinetics. However, HO-1+/− HSCs were ineffective in radioprotection and serial repopulation of myeloablated recipients. Perturbations in key stem cell regulators were observed in HO-1+/− HSCs and hematopoietic progenitors (HPCs), which may explain the disrupted response of HO-1+/− HPCs and HPCs to acute stress. Control of stem cell stress response by HO-1 presents opportunities for metabolic manipulation of stem cell–based therapies.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 8-8
Author(s):  
Magdalena Kucia ◽  
Kamila Bujko ◽  
Andrzej Ciechanowicz ◽  
Monika Cymer ◽  
Katarzyna Sielatycka ◽  
...  

Background . The SARS-CoV-2 pandemic, with its high mortality, has become an urgent clinical problem. It is well established that SARS-CoV-2 enters human cells after binding to the angiotensin-converting enzyme 2 (ACE2) receptor and utilizes a spike protein (S) for attachment and entry into the cells, leading to their lysis or damage. This infection damages several organs, including lungs, heart, blood vessels, kidneys, and intestines and may lead to a fatal complication known as a "cytokine storm", which is the result of uncontrolled hyperactivation of the innate immunity-initiated response and the release of several pro-inflammatory cytokines. An important potential aspect of SARS-CoV-2 infection is damage to the stem cell compartment, which may lead to severe complications from the infection. The ACE2 receptor has been described as being expressed on the surfaces of certain types of stem cells, including specified hematopoietic stem cells (HSCs) and endothelial progenitor cells (EPCs). These cells could be potentially damaged and lysed after virus entry or could undergo pyroptosis due to hyperactivation of Nlrp3 inflammasomes. Supporting this possibility, we recently reported that all Nlrp3 inflammasome components are expressed in HSCs (Adamiak, M et al. Nlrp3 Inflammasome Signaling Regulates the Homing and Engraftment of Hematopoietic Stem Cells (HSPCs) by Enhancing Incorporation of CXCR4 Receptor into Membrane Lipid Rafts. Stem Cell Rev and Rep (2020).https://doi.org/10.1007/s12015-020-10005-w). Hypothesis. We hypothesized that one of the triggers of a cytokine storm could be interaction of the ACE2 receptor with the SARS-CoV-2 spike protein, leading to hyperactivation of Nlrp3 inflammsomes in target cells including population of stem cells.Materials and Methods. Experiments were performed on human stem cells at different levels of specification, including very small CD34+CD133+lin-CD45- cells, which may become specified into HSCs and EPCs, as well as human CD34+Lin-CD45+ HSCs and CD34+ CD133+ KDR+ CD31+ EPCs. These cells were phenotyped for expression of ACE2 and the SARS-CoV-2 entry-facilitating transmembrane protease TMPRSS2 at the mRNA level and by FACS at the protein level. Next, we exposed these cells to the NCP-CoV (2019-nCoV) spike protein (S1+S2 ECD, expressed with a His-tag; Sino Biological) at a concentration of 10 nM. After 16 h of incubation, the cells were lysed, and total RNA was isolated for qRT-PCR analysis of Nlrp3 and essential Nlrp3 inflammasome components, including ASC, caspase 1, IL1b, and IL18. In some experiments, UCB-derived HSCs were plated into 96-well plates and stimulated with NCP-CoV (2019-nCoV) spike protein, as described above, alone or together with angiotensin 1-7 or the anti-inflammatory heme oxygenase 1 (HO-1) activator CoPP. Results. First, we observed that the ACE2 receptor and SARS-CoV-2 entry-facilitating transmembrane protease TMPRSS2 are expressed by all types of stem cells evaluated in our studies. Moreover, we detected activation of Nlrp3 inflammasomes in response to viral spike protein. This activation was inhibited by exposure of the stimulated cells to angiotensin 1-7 or CoPP. Conclusions. We envision that, in addition to directly infecting target cells, virus can hyperactivate the Nlrp3 inflammasome in stem cells, which may trigger their pyroptosis. Therefore, since we still do not have an effective SARS-CoV-2 vaccine in hand, the results presented in our current work suggest that inhibition of the Nlrp3 inflammasome by the small-molecule inhibitor MCC950 or application of Nlrp3 inflammasome inhibitors, such as Ang (1-7) or heme oxygenase 1 activators, could find potential clinical application to prevent the onset of a cytokine storm and cell pyroptosis. Disclosures No relevant conflicts of interest to declare.


Antioxidants ◽  
2021 ◽  
Vol 10 (1) ◽  
pp. 94
Author(s):  
Wojciech Krzeptowski ◽  
Patryk Chudy ◽  
Grzegorz Sokołowski ◽  
Monika Żukowska ◽  
Anna Kusienicka ◽  
...  

G-quadruplexes (G4) are stacked nucleic acid structures that are stabilized by heme. In cells, they affect DNA replication and gene transcription. They are unwound by several helicases but the composition of the repair complex and its heme sensitivity are unclear. We found that the accumulation of G-quadruplexes is affected by heme oxygenase-1 (Hmox1) expression, but in a cell-type-specific manner: hematopoietic stem cells (HSCs) from Hmox1−/− mice have upregulated expressions of G4-unwinding helicases (e.g., Brip1, Pif1) and show weaker staining for G-quadruplexes, whereas Hmox1-deficient murine induced pluripotent stem cells (iPSCs), despite the upregulation of helicases, have more G-quadruplexes, especially after exposure to exogenous heme. Using iPSCs expressing only nuclear or only cytoplasmic forms of Hmox1, we found that nuclear localization promotes G4 removal. We demonstrated that the proximity ligation assay (PLA) can detect cellular co-localization of G-quadruplexes with helicases, as well as with HMOX1, suggesting the potential role of HMOX1 in G4 modifications. However, this colocalization does not mean a direct interaction was detectable using the immunoprecipitation assay. Therefore, we concluded that HMOX1 influences G4 accumulation, but rather as one of the proteins regulating the heme availability, not as a rate-limiting factor. It is noteworthy that cellular G4–protein colocalizations can be quantitatively analyzed using PLA, even in rare cells.


2006 ◽  
Author(s):  
Hideyo Hirai ◽  
Pu Zhang ◽  
Tajhal Dayaram ◽  
Christopher Hetherington ◽  
Shin-ichi Mizuno ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document