scholarly journals Calvariae-Resident Osteomacs That Are Phenotypically and Functionally Distinct from Marrow-Derived Macrophages Interact with Megakaryocytes to Regulate Hematopoietic Stem Cell Function

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 28-28 ◽  
Author(s):  
Safa F Mohamad ◽  
Linlin Xu ◽  
Himes R Evan ◽  
Hao Wu ◽  
Marta Alvarez ◽  
...  

Abstract Maintenance of stem cell function is an orchestrated event requiring the participation of multiple cell types within the hematopoietic niche. Precise networking between hematopoietic stem cells (HSC) and these cell types is critical for the maintenance of the stem cell pool. Evidence is accumulating that multiple cell types cooperate to collectively maintain HSC function in the hematopoietic niche. We report here a detailed characterization of calvariae-resident osteomacs (OM) and outline how these cells require cooperation from megakaryocytes (MK) to sustain HSC function. We also describe in detail discriminating phenotypic and functional properties that clearly distinguish OM from marrow-derived macrophages (Mφ). Osteomacs, identified as CD45+F4/80+ cells, were easily detectable in calvarial cell (CC) preparations (3-5% of total CC) collected by the enzymatic digestion of calvariae from 2d-old pups. To assess the effect of MK, a known regulator of osteoblast (OB) proliferation and differentiation, on OM, we performed co-cultures using CC and MK prepared from fetal liver. In the absence of MK, OM did not increase in numbers over a period of 5 days in culture and remained approximately 5% of total cultured cells. However, in the presence of MK, OM significantly increased to become between 25% and 30% of total cells demonstrating that MK regulate OM proliferation. Clonogenic assays established that OM support hematopoiesis enhancing activity of OB and that this activity can be upregulated by MK. Interestingly, marrow-derived Mφ were unable to mediate the same hematopoiesis enhancing activity regardless of whether MK were present in the co-culture or not. These results were validated via primary and secondary transplantations in lethally irradiated hosts whereby the highest repopulating potential was observed among marrow-derived LSK cells co-cultured for 5 days with a mixture of OB, OM, and MK. Using eight surface markers and flow cytometric analysis, we established that although marrow-derived Mφ and OM share many phenotypic similarities (CD45, F4/80, CD68, CD11b, Mac2, and GR-1), only OM expressed MCSFR and CD166, thus providing a distinct and unique profile for these cells. To assess changes in pathway activation between resting and MK-activated OM, we performed single cell genomic analysis. This approach detected the upregulation of several canonical pathways important in HSC maintenance such as Ephrin receptor signaling, PDGF signaling, and leukocyte extravasation signaling in MK-stimulated OM. Single cell genomic analysis between CC-derived OM and marrow-derived Mφ (isolated from each tissue as CD45+F4/80+ cells) revealed 39 genes to be significantly different between the two cell types. Strikingly, many genes such as IGF1, KITL and NOTCH2 that have previously been implicated in HSC regulation were upregulated in OM. MCSFR1 a known regulator of proliferation, differentiation and survival of Mφ was also upregulated in OM corroborating the data previously collected from flow cytometric analyses. However, OM did not respond to exogenous MCSF stimulation suggesting that MCSF alone is not sufficient to induce OM proliferation or that direct contact with MK is required for induction of proliferation. To investigate changes at the protein translational level, we examined both cell types using CyTOF and a panel of 24 surface and intracellular antibodies. The surface marker CD169 which was previously associated with HSC retention when present on cellular components of the hematopoietic niche was expressed on OM but not on Mφ. Intriguingly, OM expressed both CD86 and CD206 which are known M1 and M2 Mφ markers, respectively. TNF-α, TIMP2, FGF2 and MCP1 which are known HSC regulators were also upregulated in OM. Finally, the majority of OM expressed embigin and IL-18, both of which have been implicated in the maintenance of HSC function. These data demonstrate that although bone-associated OM share many properties with marrow-derived Mφ, they are phenotypically and functionally distinct and are critical for the maintenance of HSC function. Furthermore, the function of OM, and consequently that of the two components of CC, namely OB and OM, is significantly augmented by interactions with MK demonstrating that the crosstalk between OM, OB and MK form a novel network in supporting HSC function. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2020 ◽  
Vol 135 (23) ◽  
pp. 2049-2058 ◽  
Author(s):  
Christine R. Keenan ◽  
Nadia Iannarella ◽  
Gaetano Naselli ◽  
Naiara G. Bediaga ◽  
Timothy M. Johanson ◽  
...  

Abstract Loss of heterochromatin has been proposed as a universal mechanism of aging across different species and cell types. However, a comprehensive analysis of hematopoietic changes caused by heterochromatin loss is lacking. Moreover, there is conflict in the literature around the role of the major heterochromatic histone methyltransferase Suv39h1 in the aging process. Here, we use individual and dual deletion of Suv39h1 and Suv39h2 enzymes to examine the causal role of heterochromatin loss in hematopoietic cell development. Loss of neither Suv39h1 nor Suv39h2 individually had any effect on hematopoietic stem cell function or the development of mature lymphoid or myeloid lineages. However, deletion of both enzymes resulted in characteristic changes associated with aging such as reduced hematopoietic stem cell function, thymic involution and decreased lymphoid output with a skewing toward myeloid development, and increased memory T cells at the expense of naive T cells. These cellular changes were accompanied by molecular changes consistent with aging, including alterations in nuclear shape and increased nucleolar size. Together, our results indicate that the hematopoietic system has a remarkable tolerance for major disruptions in chromatin structure and reveal a role for Suv39h2 in depositing sufficient H3K9me3 to protect the entire hematopoietic system from changes associated with premature aging.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. SCI-15-SCI-15
Author(s):  
Lluis Espinosa ◽  
Anna Bigas

Abstract Abstract SCI-15 The Notch pathway controls the generation of different cell types in most tissues including blood, and dysregulation of this pathway is strongly associated with oncogenic processes. In many systems, Notch is also required for the maintenance of the stem cell populations. However, in the adult hematopoietic system this link between Notch and stemness has not been established. Instead, work of several groups, including ours, has clearly demonstrated that Notch has a prominent role in the generation of hematopoietic stem cells (HSC) during embryonic development. Although the first wave of blood cells appears in the mouse embryo around day 7.5 of development and is independent of Notch function, embryonic HSC are formed around day 10 of development from endothelial-like progenitors that reside in the embryonic aorta surrounded by the gonad and mesonephros, also called AGM region. By analyzing different Notch pathway mutant mouse embryos, we have demonstrated the involvement of the Jagged1-Notch1-GATA2 axis in this event. However, the formal demonstration that Notch regulates the GATA2 gene during HSC generation is still lacking. We have now found that GATA2 is a direct Notch target in vivo during embryonic HSC generation. However, whereas Notch positively activates GATA2 transcription in the HSC precursors, it simultaneously activates hes1 transcription, which acts a repressor of the same GATA2 gene. This finding directly implicates hes1 in the regulation of HSC development although further studies using loss-of-function mutant embryos are still needed. Altogether, our results indicate that both Notch and hes1 are required to finely regulate the levels, distribution, and likely the timing of GATA2 expression through an incoherent feed-forward loop. In parallel, we have identified other downstream targets of Notch in the AGM region by ChIP-on-chip and expression microarray analysis that we are currently characterizing. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol Publish Ahead of Print ◽  
Author(s):  
Joydeep Ghosh ◽  
Roy El Koussa ◽  
Safa F. Mohamad ◽  
Jianyun Liu ◽  
Melissa A. Kacena ◽  
...  

2011 ◽  
Vol 208 (3) ◽  
pp. 421-428 ◽  
Author(s):  
Armin Ehninger ◽  
Andreas Trumpp

Stem cell niches are defined as the cellular and molecular microenvironments that regulate stem cell function together with stem cell autonomous mechanisms. This includes control of the balance between quiescence, self-renewal, and differentiation, as well as the engagement of specific programs in response to stress. In mammals, the best understood niche is that harboring bone marrow hematopoietic stem cells (HSCs). Recent studies have expanded the number of cell types contributing to the HSC niche. Perivascular mesenchymal stem cells and macrophages now join the previously identified sinusoidal endothelial cells, sympathetic nerve fibers, and cells of the osteoblastic lineage to form similar, but distinct, niches that harbor dormant and self-renewing HSCs during homeostasis and mediate stem cell mobilization in response to granulocyte colony-stimulating factor.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2576-2576
Author(s):  
Safa F. Mohamad ◽  
Joydeep Ghosh ◽  
Andrea M. Gunawan ◽  
Rachel Blosser ◽  
Malgorzata Kamocka ◽  
...  

Abstract Networking between hematopoietic stem cells (HSC) and cells of the hematopoietic niche is critical for the maintenance of stem cell renewal and function. HSC maintenance in the hematopoietic niche is considered to be the product of intimate interactions between cellular and soluble elements of the niche and stem cells. Among the cellular components of the niche participating in this function are a group of specialized bone-resident macrophages known as osteomacs (OM). Previously, we established the importance of osteoblasts (OB) in hematopoiesis and quite recently, we described the importance of OM and their interactions with OB and megakaryocytes (MK) in sustaining HSC function. We have also illustrated that CD166 is a critical functional marker of stem cell function and competence of the hematopoietic niche. Interestingly, immature OB which are CD166+ mediate the highest level of hematopoietic enhancing activity. We report here the importance of CD166 on calvarie-resident OM (identified as CD45+F4/80+ cells) and outline how these cells require cooperation from MK to increase CD166 expression and sustain HSC function. Bone resident-osteomacs, which are phenotypically similar but functionally different from bone marrow-derived macrophages, were collected by the enzymatic digestion of neonatal calvarial cells (NCC) or long bones of adult mice. Transplantation assays indicated that OM are relatively radioresistant and survive several weeks post lethal radiation. However, they eventually deplete and are replenished by progeny of donor HSC. To understand the importance of OM-OB-MK interactions in maintaining HSC function in the niche, we performed 3D cytometry on fixed and stained bone marrow sections that revealed intimate spatial interactions between OM, OB, MK and HSC. To assess changes in gene expression observed due to these interactions, we cultured NCC for 16hr in the absence or presence of MK prepared from fetal liver followed by sorting out OM from each group. These cells were then captured as single cells and sequenced to identify potential targets through which OM enhanced hematopoietic activity. Strikingly, several genes involved in the hematopoietic stem cell differentiation pathway including lmo2, fli1 and ikzf1 were upregulated in OM cultured in the presence of MK. Other genes that were upregulated were embigin and PF-4, both of which have been implicated in the maintenance of HSC function. Interestingly, OM express embigin, angiogenin and IL-18 mRNA similar to proximal osteolineage cells which we previously described as HSC regulators. To investigate changes at the translational level, we performed single cell proteomics using CyTOF. NCC were cultured for 2 days in the absence and presence of MK followed by staining for a panel of 29 surface and intracellular markers. Expression of markers such as CD166, embigin, mac-2 and STAT3 amongst others was elevated on OM cultured with MK compared to OM cultured without. These data informed our decision to focus our future investigations on CD166 and embigin. Next CD166+OM and CD166-OM were isolated by cell sorting and used in co-culture assays with OB to support the production of clonogenic cells in vitro. Only the CD166+ fraction of OM maintained hematopoietic activity similar to unsorted OM, implicating CD166 as one of the mediators of OM function. These results were validated using recombinant CD166 protein to substitute for OM function. Under these conditions, recombinant CD166 supported the hematopoietic enhancing activity of OB in the absence of OM. Recombinant Angiogenin and IL-18 were unable to augment the CD166-mediated support of hematopoiesis. Interestingly, CD166 knockout OM were unable to mediate the same hematopoietic enhancing activity observed with WT OM regardless of the presence or absence of MK in culture. In vivo transplantation studies to corroborate these findings have been initiated and are very early to yield meaningful conclusions. These data demonstrate that CD166 is one of the important mediators through which OM maintain HSC function. However, CD166-OM mediated HSC function is only maintained in conjunction with OB-MK interactions. Our data indicate the importance of crosstalk between OM, OB and MK which leads to the expression of novel mediators such as CD166 to support HSC function. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1487-1487
Author(s):  
C. Matthias Wilk ◽  
Akos G. Czibere ◽  
Ron-Patrick Cadeddu ◽  
Sebastian Buest ◽  
Frank A. Schildberg ◽  
...  

Abstract Abstract 1487 Poster Board I-510 TIMP–1 protein (Tissue Inhibitor of Metalloproteinases) is a recently identified tetraspanin interacting cell surface protein in the immortalized human breast epithelial cell line MCF10A. Tetraspanins like CD63 are proteins that consist of four transmembrane domains and are known to interact with integrins. Integrins play a crucial role in hematopoietic stem cell homing and mobilization. We first screened gene array data sets of CD34+ human hematopoietic stem and progenitor cells (HSPCs) and found TIMP–1 mRNA expression. In this study we show that TIMP–1 co-localizes with the tetraspanin CD63 and Beta-1-Integrin. Furthermore, we found a functional interaction of TIMP-1 with its receptor complex on G-CSF mobilized HSPCs. All experiments were carried out using highly enriched CD34+ cells. Co-immunoprecipitation shows that TIMP–1 binds to CD63. Using high resolution Stimulated Emission Depletion (STED) microscopy we could confirm co-localization of TIMP-1 and CD63 as well as Beta-1 Integrin and CD63. To further characterize the role of TIMP-1 in the Beta-1-Integrin signaling, we used an antibody specific to the active form of Beta-1- Integrin. Flow cytometric analysis revealed a significantly higher number of active Beta-1-Integrin in TIMP-1 stimulated cells suggesting TIMP-1 to activate the receptor complex on CD34+ cells. For functional analysis of the receptor complex formation, transwell migration assays were performed showing significantly increased migratory capacities of TIMP–1 treated cells. Additionally, TIMP–1 stimulation leads to a significantly increased adhesion rate of CD34+ cells to the fibronectin-coated dish. To assess a potential role of TIMP–1 in apoptosis, CD34+ HSPCs were co-incubated with thapsigargin and TIMP–1 or DMSO as a control. Subsequent flow cytometric analysis of cleaved Caspase-3 revealed a decrease of apoptotic cells in the TIMP-1 treated samples. In summary, we can show that TIMP-1, CD63 and Beta-1-Integrin form a complex on CD34+ HSPCs. TIMP-1 activates the Beta-1-Integrin signaling in HSPCs and alters the adhesive as well as the migratory behavior of CD34+ HSPCs. Furthermore, TIMP-1 induces an antiapoptotic effect in CD34+ cells. The functional effects of TIMP-1 in HSPCs might be of relevance in clinical hematopoietic stem cell transplantation so that we are currently about to verify these effects in an in vivo model. Disclosures: No relevant conflicts of interest to declare.


2018 ◽  
Vol 2 (21) ◽  
pp. 3063-3069 ◽  
Author(s):  
Samuel J. Wattrus ◽  
Leonard I. Zon

Abstract Each stem cell resides in a highly specialized anatomic location known as the niche that protects and regulates stem cell function. The importance of the niche in hematopoiesis has long been appreciated in transplantation, but without methods to observe activity in vivo, the components and mechanisms of the hematopoietic niche have remained incompletely understood. Zebrafish have emerged over the past few decades as an answer to this. Use of zebrafish to study the hematopoietic niche has enabled discovery of novel cell–cell interactions, as well as chemical and genetic regulators of hematopoietic stem cells. Mastery of niche components may improve therapeutic efforts to direct differentiation of hematopoietic stem cells from pluripotent cells, sustain stem cells in culture, or improve stem cell transplant.


2022 ◽  
Author(s):  
Ling Liu ◽  
Matthew T Buckley ◽  
Jaime M Reyes ◽  
Soochi Kim ◽  
Lei Tian ◽  
...  

Exercise has the ability to rejuvenate stem cells and improve tissue homeostasis and regeneration in aging animals. However, the cellular and molecular changes elicited by exercise have not been systematically studied across a broad range of cell types in stem cell compartments. To gain better insight into the mechanisms by which exercise affects niche and stem cell function, we subjected young and old mice to aerobic exercise and generated a single cell transcriptomic atlas of muscle, neural and hematopoietic stem cells with their niche cells and progeny. Complementarily, we also performed whole transcriptome analysis of single myofibers from these animals. We identified common and unique pathways that are compromised across these tissues and cell types in aged animals. We found that exercise has a rejuvenating effect on subsets of stem cells, and a profound impact in the composition and transcriptomic landscape of both circulating and tissue resident immune cells. Exercise ameliorated the upregulation of a number of inflammatory pathways as well as restored aspects of cell-cell communication within these stem cell compartments. Our study provides a comprehensive view of the coordinated responses of multiple aged stem cells and niche cells to exercise at the transcriptomic level.


2021 ◽  
Vol 118 (14) ◽  
pp. e2015748118
Author(s):  
Jun Xia ◽  
Zhixin Kang ◽  
Yuanyuan Xue ◽  
Yanyan Ding ◽  
Suwei Gao ◽  
...  

During vertebrate embryogenesis, fetal hematopoietic stem and progenitor cells (HSPCs) exhibit expansion and differentiation properties in a supportive hematopoietic niche. To profile the developmental landscape of fetal HSPCs and their local niche, here, using single-cell RNA-sequencing, we deciphered a dynamic atlas covering 28,777 cells and 9 major cell types (23 clusters) of zebrafish caudal hematopoietic tissue (CHT). We characterized four heterogeneous HSPCs with distinct lineage priming and metabolic gene signatures. Furthermore, we investigated the regulatory mechanism of CHT niche components for HSPC development, with a focus on the transcription factors and ligand–receptor networks involved in HSPC expansion. Importantly, we identified an endothelial cell-specific G protein–coupled receptor 182, followed by in vivo and in vitro functional validation of its evolutionally conserved role in supporting HSPC expansion in zebrafish and mice. Finally, comparison between zebrafish CHT and human fetal liver highlighted the conservation and divergence across evolution. These findings enhance our understanding of the regulatory mechanism underlying hematopoietic niche for HSPC expansion in vivo and provide insights into improving protocols for HSPC expansion in vitro.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 876-876
Author(s):  
Simon Haas ◽  
Chiara Baccin ◽  
Jude Al-Sabah ◽  
Lars Velten ◽  
Steinmetz Lars ◽  
...  

Abstract Coordinated interaction of many cell types is required to facilitate hematopoietic and mesenchymal stem cell maintenance and differentiation in the bone marrow. However, the molecular factors and cell types involved in this complex interplay remain poorly understood. Here we developed a combined single cell and spatial transcriptomics approach to address this problem. Large-scale single-cell transcriptional profiling in conjunction with a multi-layered sorting approach allowed us to generate a complete and evenly sampled transcriptional map of all major bone and bone marrow populations. Our dataset covers all cell types or differentiation trajectories involved in mesenchymal and hematopoietic stem cell differentiation, osteogenesis, adipogenesis, myelopoiesis, erythropoiesis, lymphopoiesis, memory T cell formation as well as bone marrow neural innervation and vascularization at the single cell level. Using this data, we derive fundamental properties of the described cell types, clarify the cellular source of signals affecting stem cell differentiation processes and provide a systems view on putative intercellular interactions. Systematic spatial transcriptomics, using laser-capture microdissection of selected bone marrow niches followed by transcriptional profiling and bioinformatic cellular deconvolution, allowed us to confirm predicted interactions and map the cellular composition of distinct bone marrow niches. Our analyses highlight the importance of pre-adipogenic CXCL12 abundant reticular cells as key niche cells for stem cell maintenance, provides a holistic systems view of the hematopoietic stem cell niche and offers a novel approach to systematically deconvolute the molecular, cellular and spatial composition of complex tissues. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document