scholarly journals Mechanisms of stem cells action: reality and hypotheses

Author(s):  
D. A. Kudlay ◽  
D. A. Ivolgin

Despite the long history of stem cells studying, the mechanisms of their action are still not fully understood. A classic example is the described non-hematological effect of hematopoietic stem cells (HSC), presumably due to the ability to transdifferentiation and cell fusion. The focus of clinical application in the field of regenerative medicine is mesenchymal stromal cells (MSC), and the understanding of the ways of implementfnion of their regenerative potential has evolved significantly. MSC were discovered and described as a structural unit of the HSC niche responsible for the repair of connective tissue through differentiation. However, it later turned out that they are a regulator of various processes in the body as a whole. These processes include anti-inflammatory, antifibrotic, and immunomodulatory effects, which are realized in various ways. Among these pathways, the paracrine mechanism has already been identified – the release of various growth factors, exosomes and microvesicles, the mechanism of direct transfer of mitochondria and other cellular organelles from cell to cell using tunneling nanotubes, the mechanism of efferocytosis. One of the latest discoveries in this field was the immunomodulatory effect of apoptotic MSC.In general, the study of the stem cells mechanisms of action is a living, developing field of science in which the last word will not be said soon.

2021 ◽  
Vol 22 (17) ◽  
pp. 9231
Author(s):  
Alexander Belyavsky ◽  
Nataliya Petinati ◽  
Nina Drize

In the bone marrow of vertebrates, two types of stem cells coexist—hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). Hematopoiesis only occurs when these two stem cell types and their descendants interact. The descendants of HSCs supply the body with all the mature blood cells, while MSCs give rise to stromal cells that form a niche for HSCs and regulate the process of hematopoiesis. The studies of hematopoiesis were initially based on morphological observations, later extended by the use of physiological methods, and were subsequently augmented by massive application of sophisticated molecular techniques. The combination of these methods produced a wealth of new data on the organization and functional features of hematopoiesis in the ontogenesis of mammals and humans. This review summarizes the current views on hematopoiesis in mice and humans, discusses the development of blood elements and hematopoiesis in the embryo, and describes how the hematopoietic system works in the adult organism and how it changes during aging.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 249-249
Author(s):  
Junke Zheng ◽  
HoangDinh Huynh ◽  
Chengcheng Zhang

Abstract Abstract 249 The physiological role of Angiopoietin-like proteins (Angptls) in hematopoietic system is unknown. Here we showed that Angptl3 is expressed by both hematopoietic stem cells (HSCs) and bone marrow stromal cells. In particular, the expression of Angptl3 in the bone marrow stromal cells is significantly increased upon transplantation, suggesting that this protein may play an important role in the bone marrow under stress. We asked whether Angptl3 expression had a functional role in HSCs by utilizing mice ablated for Angptl3. Using Hoechst/pyronin Y staining and Brdu incorporation analysis, we found that HSCs in Angptl3-null mice exhibited significantly decreased quiescence compared to those in wild-type mice. To test the role of Angptl3 in the stress response of hematopoietic cells, we treated mice with 5-fluorouracil (5-FU), which is toxic to cycling cells and accelerates the entry of HSCs into the cell cycle. The survival of Angptl3-null mice was significantly lower than that of wild-type mice. To further identify the role of Angptl3 in stress response of HSCs, we examined whether Angptl3 affected DNA damage in HSCs upon transplantation. To this end, we transplanted WT bone marrow cells into lethally irradiated Angptl3 null recipients or WT mice. We found that HSCs in the Angptl3 null recipient mice had significantly increased gamma-H2AX foci compared to WT recipients, suggesting that Angptl3 protects HSCs from DNA damage. We further used the competitive reconstitution analysis to determine the roles of Angptl3 on HSC activity. Importantly, both Angptl3-null HSCs transplanted to wild-type recipients and wild-type HSCs transplanted to Angptl3-null recipients showed impaired repopulation. These results conclude that Angptl3 has both cell-autonomous and environmental effects that support the in vivo activity of HSCs. To identify the intracellular target of Angptl3 in HSCs, we performed DNA microarray and real-time RT-PCR analyses to compare the gene expression in HSCs isolated from WT and Angptl3 null mice. We found that Angptl3-null HSCs had increased expression of transcription factor Ikaros. Consistently, extrinsic treatment of HSCs by Angptl3 also suppressed the expression of Ikaros. Ikaros is a zinc finger transcription factor important for differentiation of lymphoid, myeloid, and erythroid cells, and its expression is low in multi-potent HSCs, but high in progenitors with lymphoid-myeloid potential. Since Angptl3 downregulates the expression of Ikaros in HSCs, we examined the effect of forced expression of Ikaros on HSC activities. Indeed, overexpression of Ikaros enhanced HSC cycling and DNA damage, and diminished their repopulation activity, indicating the downregulation of Ikaros by extrinsic Angptl3 maintains the stemness of HSCs. We studied the spatial relationship of Angptl3-expressing cells and the bone-marrow HSCs using immunohistochemical tools. We showed that 58.6% of Angptl3-producing cells were in contact with sinusoidal endothelial cells in bone marrow and that 60.8% of HSCs are adjacent to Angptl3-producing cells in the bone marrow. To directly test whether Angptl3-producing bone marrow stromal cells support HSC expansion, we co-cultured HSCs and CD45-SSEA4+ cells and used competitive reconstitution analysis to measure HSC activity. HSCs co-cultured with WT CD45-SSEA4+ cells had significantly increased repopulation relative to those co-cultured with Angptl3 null CD45-SSEA4+ cells (36% vs. 17%). This result demonstrated that bone marrow CD45-SSEA4+ cells support expansion of HSCs, and provided the functional evidence that Angptl3-producing stromal cells are a part of HSC niche in the bone marrow. Thus, Angptl3-producing cells are an important component of the HSC niche. Our experiments demonstrate a unique example of the extrinsic control of stemness by cell-autonomous effects from stem cells per se and by environmental effects from the niche cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5004-5004
Author(s):  
Fumio Nakahara ◽  
Sandra Pinho ◽  
Daniel K. Borger ◽  
Qiaozhi Wei ◽  
Maria Maryanovich ◽  
...  

Hematopoietic stem cells (HSCs) are maintained by bone marrow (BM) niches in vivo, but the ability of niche cells to maintain HSCs ex vivo is markedly diminished. Expression of niche factors (Scf, Cxcl12, Vcam1 and Angpt1) by Nestin-GFP+ mesenchymal-derived stem cells (MSCs) is downregulated upon culture and lose its effect of maintaining HSC in vitro, suggesting that transcriptional rewiring may contribute to this reduced potential in cultured MSCs. To gain further insight, we searched RNA sequencing data for transcriptional regulators that were highly expressed in Nestin-GFP+ stroma, revealing 40 potential candidates. We compared the expression of these genes by real-time quantitative PCR (qPCR) in freshly isolated Nestin-GFP+ or Nestin-GFP- BM CD45-Ter119-CD31- cells, with that of cultured Nestin-GFP+ stroma. These analyses yielded 28 candidate genes after the elimination of 12 genes due to non-specific expression or lack of downregulation after culture. We cultured stromal cells isolated from Scf-GFP knock-in mice in which GFP expression reflects endogenous Scf mRNA synthesis. Upon culture, GFP expression was rapidly downregulated in these cells, demonstrating the potential of using GFP to screen for factors capable of revitalizing niche activity in cultured MSCs. We generated lentiviral vectors expressing 28 selected genes and transduced the viral mixture into cultured stromal cells derived from Scf-GFP mice. Five days after transduction, we observed re-emergence of GFP+ cells and these GFP+ cells were sorted and plated in limiting dilutions to isolate single cell-derived clones. Using this approach, we generated 16 independent GFP+ single cell-derived clones. To determine the specific combination of genes that enables cultured stromal cells to regain their capacity to maintain and expand HSCs in vitro, lineage-negative (Lin-) BM cells were co-cultured with each single cell-derived clone or control stroma. Thus, we identified 5 transcription factors (Klf7, Ostf1, Xbp1, Irf3, and Irf7; KOXII) that restored HSC niche function in cultured BM-derived MSCs. These revitalized MSCs (rMSCs) exhibited enhanced synthesis of HSC niche factors while retaining their mesenchymal differentiation capacity. In contrast to HSCs co-cultured with control MSCs, HSCs expanded with rMSCs in vitro showed higher repopulation capacity and enabled lethally irradiated recipient mice to survive better. Competitive reconstitution assays revealed 7-fold expansion of functional HSCs by rMSCs. Moreover, rMSCs prevented the accumulation of DNA damage in cultured HSCs, a hallmark of ageing and replication stress. To investigate the revitalization mechanism, we performed ATAC-seq in freshly sorted Scf-GFP- CD45-Ter119-CD31- cells, Scf-GFP+ CD45-Ter119-CD31- cells, rMSCs and control vector-transduced stroma. We found that revitalization of MSCs led to 9,623 peaks of open chromatin in rMSCs when compared to control MSCs. Of these, 626 open peaks were also detected in freshly isolated Scf-GFP+ cells when compared to Scf-GFP- cells. Motif analyses of the sequence at these 626 peaks revealed that myocyte enhancer factor 2c (Mef2c) was among the most significantly enriched transcription regulators. Mef2c was also expressed at high levels in both rMSCs and freshly isolated Scf-GFP+ cells compared to control cultured MSCs and freshly isolated Scf-GFP- cells by RNA-seq and real-time qPCR. To evaluate the role of Mef2c in rMSCs, we knocked down Mef2c in rMSCs by short hairpin RNA lentiviral transduction (shMef2c). We found that the expression of niche factors (Scf, Cxcl12 and Vcam1) was reduced in shMef2c-transduced compared to parental rMSCs. In addition, shMef2c transduced-rMSCs exhibited reduced (by 43%) capacity to expand HSCs in co-culture compared to shCntrl transduced-rMSCs. These results suggest a role for Mef2c as a downstream effector mediating MSC revitalization. We are now exploring the method to make these rMSCs to form new niches in vivo. Our results suggest that combination of KOXII genes are able to fully restore the niche activity in MSCs ex vivo and establish a new platform that provides critical insight in the regulatory network of the HSC niche leading to the basis toward the engineering of supportive niches for curative cell therapies. Disclosures Wei: Albert Einstein College of Medicine, Inc: Patents & Royalties. Frenette:Albert Einstein College of Medicine, Inc: Patents & Royalties; Ironwood Pharmaceuticals: Research Funding; Cygnal Therapeutics: Equity Ownership; Pfizer: Consultancy.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 505-505
Author(s):  
Sandra Pinho ◽  
Julie Lacombe ◽  
Maher Hanoun ◽  
Ingmar Bruns ◽  
Yuya Kunisaki ◽  
...  

Abstract Abstract 505 Few markers have thus far been identified on native mesenchymal stem cells (MSCs), both in the mouse and human systems. Most markers cited in the literature are indeed based on expression analyses on heterogeneous cultured cell populations, which may not have self-renewal properties if rigorously tested by transplantation assays. Previous studies using Nestin (Nes)-Gfp transgenic mice showed that Nes-GFP+ cells are self-renewing MSCs, a major constituent of the hematopoietic stem cell (HSC) niche in the bone marrow (BM) (Nature 2010; 466:829). However, the cytoplasmic location of Nestin precludes prospective live cell isolation outside of the transgenic mice. Hence, finding a combination of surface markers labeling Nestin+ cells in situ would be valuable to isolate bona fide MSCs and characterize niche cells. Screening analyses toward this end revealed that PDGFRα and CD51 expression among CD45− Ter119− CD31− BM stromal cells comprised a large fraction (∼60%) of Nes-GFP+ cells. Upon gating first on PDGFRα+ and CD51+, double-positive cells were also highly enriched in Nes-GFP+ cells (∼75%), and represented a rare fraction (∼2%) of the stromal population. Endogenous Nestin expression was also enriched in PDGFRα+ CD51+ cells, compared to single-positive or double-negative stromal cells (control subsets). Cell sorting of BM PDGFRα+ CD51+ and control subsets revealed that PDGFRα+ CD51+ significantly enriched (> 10-fold, p<0.05) for colony forming unit-fibroblastic (CFU-F) and multipotent clonal mesenspheres (> 7-fold, p<0.01) that differentiate robustly along the osteoblastic, chondrocytic and adipocytic lineages. To test in vivo self-renewal capacity, clonal spheres or polyclonal freshly sorted PDGFRα+ CD51+ cells and control subsets were transplanted into recipient mice by different approaches (renal capsule implants, collagen and/or HA/TCP carrier grafts). After 2 months, secondary sphere formation assays and histological analyses revealed the in vivo self-renewal and heterotopic BM niche regeneration capacity of PDGFRα+ CD51+ cells, but not the control subsets. In addition, the PDGFRα+ CD51+ fraction of Nestin+ cells was markedly enriched in major HSC regulatory genes (Cxcl12, Vcam1, Angpt1, Opn and Scf), supporting the notion that niche activity co-segregates with MSC activity in the BM. Next, we investigated whether PDGFRα+ CD51+ cells also labeled putative Nestin+ MSCs in the human BM. To this end, we analyzed the fetal human BM (13–19 gw), a period during which hematopoietic activity is nascent. At this stage, we found that PDGFRα+ CD51+ cells comprised ∼3% of stromal cells, contained most of the CFU-F activity (6.3 ± 0.8 CFU-Fs/102 cells) in the BM, and also expressed Nestin and HSC regulatory factors. PDGFRα+ CD51+ cells could also form mesenspheres that can self-renew in vivo after heterotopic transplantation. Furthermore, we found that human BM PDGFRα+ CD51+ cells represented a subset of CD146+ cells previously suggested to mark human MSCs (Cell 2007; 131:324), as ∼30% of the CD146high cells also expressed PDGFRα and CD51, and ∼65% of PDGFRα+ CD51+ cells were CD146high. To evaluate functionally the HSC niche properties of human PDGFRα+ CD51+ cells, we set up a co-culture system of human BM CD34+ cells with PDGFRα+ CD51+ mesenspheres. We found that mesenspheres were capable of expanding the number of human CD45+ Lin− CD38− CD34+ CD90+ CD49f+ cells (hHSCs) by 11-fold (p<0.05) compared to input (day 0). In addition, hHSC expansion was 2-fold greater (p<0.05) using mesenspheres compared to serum-free media alone with hematopoietic growth factors (SCF, TPO, Flt3L). Recent studies have suggested that SCF production in the BM niche is derived from perivascular and endothelial cells distinct from Nestin+ cells (Nature 2012; 481:457), although Nestin+ MSCs express high levels of SCF (Nature 2010; 466:829). Immunofluorescence analyses of human PDGFRα+ CD51+ mesenspheres showed that all cells forming the sphere uniformly expressed both Nestin and SCF. Moreover, in the absence of SCF from the media, PDGFRα+ CD51+ mesenspheres rescued hHSCs expansion, yielding a 46- and 5-fold (p<0.001) expansion, as compared to control media alone and input, respectively. These results thus indicate that the HSC niche is conserved between the murine and human species and suggest that highly purified non-adherent cultures of niche cells may represent a useful novel technology to expand hHSCs in vitro. Disclosures: No relevant conflicts of interest to declare.


Hematology ◽  
2013 ◽  
Vol 2013 (1) ◽  
pp. 389-393 ◽  
Author(s):  
John A. Zaia ◽  
Stephen J. Forman

Abstract With the advent of effective antiretroviral therapy, the treatment of patients with HIV-related malignancies, especially lymphoma, has greatly improved, yielding results comparable to those seen in patients with lymphoma unrelated to HIV. The platform of transplantation of hematopoietic stem cells has facilitated studies of genetically modified stem cells engineered to express antiretroviral genes to resist infection by the HIV virus, testing the concept that engraftment of these cells will lead to HIV resistance and elimination of the reservoir of virus in the body. Results in patients with HIV and lymphoma have now led to studies that will test these principles in HIV patients without concomitant malignancy. In addition, in a patient with HIV and acute myeloid leukemia, the success of an allogeneic transplantation from an unrelated donor carrying a mutation in the CCR5 genes has demonstrated that, in principle, such an approach could also lead to cure of patients with HIV. Case studies in HIV patients with leukemia undergoing allogeneic transplantation also suggest that there may be a therapeutic effect on the HIV reservoir that could alter the natural history of HIV in the allogeneic setting.


2020 ◽  
Vol 15 (3) ◽  
pp. 250-262
Author(s):  
Maryam Islami ◽  
Fatemeh Soleimanifar

Transplantation of hematopoietic stem cells (HSCs) derived from umbilical cord blood (UCB) has been taken into account as a therapeutic approach in patients with hematologic malignancies. Unfortunately, there are limitations concerning HSC transplantation (HSCT), including (a) low contents of UCB-HSCs in a single unit of UCB and (b) defects in UCB-HSC homing to their niche. Therefore, delays are observed in hematopoietic and immunologic recovery and homing. Among numerous strategies proposed, ex vivo expansion of UCB-HSCs to enhance UCB-HSC dose without any differentiation into mature cells is known as an efficient procedure that is able to alter clinical treatments through adjusting transplantation-related results and making them available. Accordingly, culture type, cytokine combinations, O2 level, co-culture with mesenchymal stromal cells (MSCs), as well as gene manipulation of UCB-HSCs can have effects on their expansion and growth. Besides, defects in homing can be resolved by exposing UCB-HSCs to compounds aimed at improving homing. Fucosylation of HSCs before expansion, CXCR4-SDF-1 axis partnership and homing gene involvement are among strategies that all depend on efficiency, reasonable costs, and confirmation of clinical trials. In general, the present study reviewed factors improving the expansion and homing of UCB-HSCs aimed at advancing hematopoietic recovery and expansion in clinical applications and future directions.


Author(s):  
Valentina Orticelli ◽  
Andrea Papait ◽  
Elsa Vertua ◽  
Patrizia Bonassi Signoroni ◽  
Pietro Romele ◽  
...  

Biomolecules ◽  
2021 ◽  
Vol 11 (5) ◽  
pp. 667
Author(s):  
Meera Krishnan ◽  
Sahil Kumar ◽  
Luis Johnson Kangale ◽  
Eric Ghigo ◽  
Prasad Abnave

Adult stem cells (ASCs) are the undifferentiated cells that possess self-renewal and differentiation abilities. They are present in all major organ systems of the body and are uniquely reserved there during development for tissue maintenance during homeostasis, injury, and infection. They do so by promptly modulating the dynamics of proliferation, differentiation, survival, and migration. Any imbalance in these processes may result in regeneration failure or developing cancer. Hence, the dynamics of these various behaviors of ASCs need to always be precisely controlled. Several genetic and epigenetic factors have been demonstrated to be involved in tightly regulating the proliferation, differentiation, and self-renewal of ASCs. Understanding these mechanisms is of great importance, given the role of stem cells in regenerative medicine. Investigations on various animal models have played a significant part in enriching our knowledge and giving In Vivo in-sight into such ASCs regulatory mechanisms. In this review, we have discussed the recent In Vivo studies demonstrating the role of various genetic factors in regulating dynamics of different ASCs viz. intestinal stem cells (ISCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), and epidermal stem cells (Ep-SCs).


2015 ◽  
Vol 39 (10) ◽  
pp. 1099-1110 ◽  
Author(s):  
Iordanis Pelagiadis ◽  
Eftichia Stiakaki ◽  
Christianna Choulaki ◽  
Maria Kalmanti ◽  
Helen Dimitriou

Blood ◽  
1993 ◽  
Vol 81 (2) ◽  
pp. 365-372 ◽  
Author(s):  
JP Wineman ◽  
S Nishikawa ◽  
CE Muller-Sieburg

We show here that mouse pluripotent hematopoietic stem cells can be maintained in vitro on stroma for at least 3 weeks at levels close to those found in bone marrow. The extent of stem cell maintenance is affected by the nature of the stromal cells. The stromal cell line S17 supported stem cells significantly better than heterogeneous, primary stromal layers or the stromal cell line Strofl-1. Stem cells cultured on S17 repopulated all hematopoietic lineages in marrow-ablated hosts for at least 10 months, indicating that this culture system maintained primitive stem cells with extensive proliferative capacity. Furthermore, we demonstrate that, while pluripotent stem cells express c-kit, this receptor appears to play only a minor role in stem cell maintenance in vitro. The addition of an antibody that blocks the interaction of c-kit with its ligand essentially abrogated myelopoiesis in cultures. However, the level of stem cells in antibody-treated cultures was similar to that found in untreated cultures. Thus, it seems likely that the maintenance of primitive stem cells in vitro depends on yet unidentified stromal cell-derived factor(s).


Sign in / Sign up

Export Citation Format

Share Document