scholarly journals Mitigating oxygen stress enhances aged mouse hematopoietic stem cell numbers and function

2021 ◽  
Vol 131 (1) ◽  
Author(s):  
Maegan L. Capitano ◽  
Safa F. Mohamad ◽  
Scott Cooper ◽  
Bin Guo ◽  
Xinxin Huang ◽  
...  
2018 ◽  
Vol 64 ◽  
pp. S41-S42
Author(s):  
Emanuele Azzoni ◽  
Vincent Frontera ◽  
Joe Harman ◽  
Sten Eirik Jacobsen ◽  
Marella De Bruijn

2016 ◽  
Vol 37 (1) ◽  
pp. 18-21
Author(s):  
Deniz Cagdas ◽  
Selin Aytac ◽  
Barış Kuskonmaz ◽  
Tadashi Ariga ◽  
Mirjam van der Burg ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (16) ◽  
pp. 2812-2822 ◽  
Author(s):  
Tao Wang ◽  
Vijayalakshmi Nandakumar ◽  
Xiao-Xia Jiang ◽  
Lindsey Jones ◽  
An-Gang Yang ◽  
...  

Key Points Mysm1 is required to maintain the quiescence and pool size of HSC, and its deletion severely impairs the survival and function of HSC. Mysm1 controls HSC homeostasis by regulating Gfi1 expression via modulating histone modifications and transcriptional factors recruitment.


2015 ◽  
Vol 209 (1) ◽  
pp. 2091OIA63 ◽  
Author(s):  
Melania Tesio ◽  
Yilang Tang ◽  
Katja Müdder ◽  
Massimo Saini ◽  
Lisa von Paleske ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1274-1274
Author(s):  
Elizabeth Csaszar ◽  
Daniel Kirouac ◽  
Mei Yu ◽  
Caryn Ito ◽  
Peter W. Zandstra

Abstract Abstract 1274 Clinical outcomes of hematopoietic stem cell (HSC) transplantation are correlated with infused progenitor cell dose. Limited cell numbers in a typical umbilical cord blood (UCB) unit restricts the therapeutic potential of UCB and motivates ex vivo expansion of these cells. Strategies to grow HSCs have relied on the supplement of molecules acting directly on the stem cell population; however, in all cases, sustained HSC growth is limited by the concurrent growth of more mature cells and their endogenously produced inhibitory signaling factors. Despite increasing evidence for the important role of intercellular (between cell) communication networks, the identity and impact of non-stem cell autonomous feedback signaling remains poorly understood. Simultaneous kinetic tracking of more than 30 secreted factors produced during UCB culture, including TGF-b1, MIP-1b, and MCP-1, in combination with computational simulations of cell population dynamics, enabled us to develop a global control strategy predicted to reduce inhibitory paracrine signaling and, consequently, increase HSC self-renewal. By maintaining endogenously produced ligands at specified levels using a tuneable fed-batch (automated media dilution) strategy, we achieved significant improvements in expansions of total cell numbers (∼180-fold), CD34+ cells (∼80-fold), and NOD/SCID/IL-2Rgc-null (NSG) repopulating cells (∼11-fold, detected at limiting dilution). The fed-batch strategy has been integrated into an automated bioreactor, allowing for the generation of a clinically-relevant cell product after 12 days of culture, with minimal user manipulation. As this strategy targets the HSC environment and not the stem cells directly, it has the ability to act in combination with other expansion strategies to produce synergistic results. Unexpectedly, supplementation of the soluble protein, TAT-HOXB4, to the system, yielded the expected boost in progenitor expansion only in “sub-optimal” control conditions but not in the fed-batch system. Hypothesizing that the efficacy of HOXB4 may be dependent on the skewing of supportive vs. non-supportive cell populations, and the consequent impact of paracrine ligand production, we performed kinetic tracking of 20 hematopoietic cell types during several supportive (fed-batch, HOXB4 supplemented, Notch ligand Delta1 supplemented) vs. non-supportive (control) cultures. Meta analysis of these data revealed a non-autonomous link between HOXB4, increased megakaryocyte production, and stem cell proliferation, as well as between Notch delta-1 ligand, decreased myeloid cell production, and a decrease in the growth inhibition of stem cells. These predictions have been experimentally validated using co-cultures of sorted purified HSCs and CD41+ megakaryocykes and CD14+ monocytes. Our results identify complex connections between mature cell lineages and stem cell fate decisions and we expect to report a direct link between cell-cell interactions emerging from culture manipulations and the resulting impact on HSC self-renewal. Collectively, these studies support a dominant role for non-stem cell autonomous feedback signaling in the regulation of HSC self-renewal. Overcoming cell non-autonomous inhibition of HSC self-renewal has allowed for novel strategies to enhance HSC numbers ex vivo, thereby facilitating the production of clinically relevant quantities of stem and progenitor cells and enabling more effective strategies to treat hematologic disease. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1970-1970 ◽  
Author(s):  
Sima T Bhatt ◽  
Darlene Monlish ◽  
Eric J. Duncavage ◽  
John Luke Keller ◽  
Bilski Rachel ◽  
...  

Abstract Myelodysplastic syndromes (MDS) are hematopoietic stem cell (HSC) disorders characterized by ineffective hematopoiesis and cellular dysplasia with high rates of transformation to leukemia. The only cure is hematopoietic stem cell transplant, which is associated with significant morbidity and mortality, thus there is a need to better delineate the pathogenesis of MDS and identify novel targets for treatment. Recent studies suggest that deregulated innate immune signaling, and, in particular, enhanced toll like receptor (TLR) signaling, may contribute to the pathogenesis of MDS. The TLRs are a family of pattern recognition receptors that play a central role in innate immunity. Specifically, TLR2 and its binding partners, TLR1 and TLR6, are markedly elevated in the CD34+ cells of patients with MDS compared to healthy controls (Wei et al, Leukemia 2013). Thus, we hypothesize that enhanced TLR2 signaling may contribute to the pathogenesis of MDS. To elucidate the contribution of TLR2 signaling to MDS, we used a well-characterized mouse model of MDS (mice expressing the NUP98-HOXD13 fusion from the hematopoietic Vav-1 promoter) to determine the effects of loss or gain of TLR2 signaling on disease progression. These "NHD13" mice have cytopenias, dysplasia, marrow hypercellularity, and increased apoptosis by 4-7 months of age; transgenic mice die by 14 months of age of leukemia or cytopenias (Lin et al, Blood 2005).Of note, similar to human HSCs, we find increased expression of TLR2 by flow cytometry on the HSCs of NHD13 mice compared to controls. Given the association of enhanced TLR2 signaling with MDS, we predicted that loss of TLR2 would improve outcomes in NHD13 mice. To this end, these mice were crossed to Tlr2-/- mice to generate 4 groups: NHD13+;Tlr2-/-; NHD13+;Tlr2+/+, NHD13-;Tlr2-/- and NHD13-;Tlr2+/+. Surprisingly, loss of TLR2 is associated with significantly worse survival with a median survival of 313 days for NHD13+;Tlr2-/-, and 367 days for NHD13+;Tlr2+/+(p=0.01; with the majority of mice dying from leukemia); and loss of TLR2 did not improve cytopenias. Ongoing experiments are aimed at determining the effects of TLR2 loss on HSC cycling, apoptosis, and function in the NHD13 mice and their wild-type (WT) sibling controls. Conversely, we are asking how stimulation of TLR2 affects disease by treating NHD13 mice chronically with a TLR1/2 agonist (PAM3CSK4) or a TLR2/6 agonist (PAM2CSK4) or water control. NHD13 mice receiving PAM2CSK4 had elevated white blood cell and hemoglobin counts after five months as compared to mice receiving PAM3CSK4 and control mice, while mice receiving PAM3CSK4 demonstrated a higher incidence of thrombocytopenia, suggesting that TLR2 signaling may contribute to cytopenias. Strikingly, however, we found that treatment with the TLR2/6 agonist (PAM2CSK4) significantly (p < 0.0001) accelerates the time to leukemia and death (but not the TLR1/2 agonist, PAM3CSK4), with deaths occurring as early as 70 days of life, mostly from leukemia. After four months of treatment, 100% of control and 93% of PAM3CSK4 treated NHD13 mice were alive, while only 37.5% of PAM2CSK4 treated NHD13 mice were alive (p= 0.0002). This raises the intriguing possibility that there are heterodimer-specific effects of TLR2 signaling on premalignant HSCs. Historically, the association of TLR2 with TLR1 versus TLR6 was thought to expand the ligand spectrum without altering downstream signaling (Farhat et al, J Leuk Biol 2008), however recent data (and our own findings) suggest that there may be heterodimer-specific differences (Rolf et al, Euro J of Immunology 2015). Furthermore, while expression of TLR2 itself correlates with low-risk disease and longer survival, high expression of TLR6 is associated with higher-risk disease and increased marrow blasts (Wei et al, Leukemia 2013). We are currently exploring the mechanisms behind the heterodimer-specific effects of TLR2 signaling on leukemogenesis and survival in the NHD13 mice by elucidating the effects of PAM3CSK4 versus PAM2CSK4 on HSC cycling, apoptosis, and function, as well as determining the downstream signaling pathways (utilizing mass cytometry) and target genes that are uniquely activated by the different agonists. Ultimately, an understanding of TLR1/2 versus TLR2/6 stimulation effects on WT and premalignant HSCs is critical to the development of targeted therapies toward this pathway. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 125 (14) ◽  
pp. 2206-2216 ◽  
Author(s):  
Wenhuo Hu ◽  
James Dooley ◽  
Stephen S. Chung ◽  
Dhruva Chandramohan ◽  
Luisa Cimmino ◽  
...  

Key Points miR-29a maintains HSC function by targeting Dnmt3a.


2016 ◽  
Vol 44 (9) ◽  
pp. S49
Author(s):  
Eric Pietras ◽  
Cristina Mirantes-Barbeito ◽  
Sarah Fong ◽  
Dirk Loeffler ◽  
Larisa Kovtonyuk ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document