In Situ Fate Mapping of Native and Stress Myelopoiesis Reveals a Unique Niche for Mono- and Dendritic Cell -Poiesis

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 38-39
Author(s):  
Jizhou Zhang ◽  
Qingqing Wu ◽  
Courtney Johnson ◽  
Giang Pham ◽  
Jeremy M. Kinder ◽  
...  

In contrast to virtually all other tissues in the body the anatomy of differentiation in the bone marrow remains unknown. This is due to the lack of strategies to examine blood cell production in situ, which are required to better understand differentiation, lineage commitment decisions, and to define how spatial organizing cues inform tissue function. We developed approaches to image and fate map -using confetti mice- myelopoiesis in situ and generated 3D atlases of granulocyte and monocyte/dendritic cell differentiation during homeostasis and after emergency myelopoiesis induced by infection with Listeria monocytogenes. Figure 1 shows stepwise differentiation during myelopoiesis. We have found that -in imaging studies- CD11b-Ly6C-CD117+CD115+ cells are MDP; Lin-CD117+CD16/32+CD115- cells are GMP; CD11b-Ly6C+CD117+CD115+ are MOP; CD11b-CD117+CD115-Ly6C+ are GP; CD11b+CD115+Ly6Chi and CD11b+CD115+Ly6Clo cells are Ly6Chi and Ly6Clo monocytes; and MHCIIhi reticular cells are dendritic cells (DC). We used these markers to map every myeloid cell in the sternum and assessed the relationships between myeloid progenitors, their offspring and candidate niches in situ with single cell resolution. To test whether the interactions observed were specific we obtained the X, Y and Z coordinates for every hematopoietic cell in the sternum (detected using αCD45 and αTer119). We then used these coordinates to randomly place each type of myeloid cell, at the same frequencies found in vivo, through the BM to generate random distributions for each myeloid cell type. We found that myeloid progenitors do not localize with HSC indicating that they leave the HSC niche during differentiation. In the steady-state GP, MOP, and MDP are found as single cells that do not associate with each other indicating that granulo-, mono-, and dendritic cell-poiesis take place in different location. Myeloid progenitors are specifically recruited to sinusoids but are depleted near endosteum and arterioles (e.g. mean MDP distance to sinusoids, arterioles, and endosteum observed 5, 134, and 105μm vs 9, 86, and 69µm in the random simulation). GP form clusters with preneutrophils and immature neutrophils, in situ fate mapping demonstrated that these clusters are oligoclonal and that additional GP are serially recruited to the cluster as the old ones differentiate. Ly6Clo monocytes and dendritic cells are selectively enriched near MDP (2.0 DC and 4.4 Ly6Clo monocytes observed within 50µm of an MDP vs 0.9 DC and 1.8 Ly6Clo monocytes in the random simulation p=0.02 and p<0.0001). Fate mapping experiments demonstrated that the monocytes around MOP and monocytes and dendritic cells around the MDP are oligoclonal but are not the derived from the MOP/MDP they associate with. These indicate that Ly6Clo monocytes and DC are produced elsewhere but are then selectively recruited to regions enriched in MDP. The results above suggest that different sinusoids might be responsible for supporting different myeloid lineages. We found that dendritic cells localize to a unique subset (8% of all vessels) of colony stimulating factor 1 (CSF1, the major regulator of monopoiesis) -expressing sinusoids. Csf1 deletion in the vasculature disrupted MDP interaction with sinusoids, leading to reduced MDP numbers and differentiation ability, with subsequent loss of Ly6Clo monocytes and dendritic cells. L. monocytogenes infection dramatically changed the architecture of myelopoiesis and caused massive expansion of myeloid progenitors leading to the formation of monoclonal GP clusters and oligoclonal MOP clusters whereas MDP are still found as single cells associated with dendritic cells. Even after this massive insult granulopoiesis and mono/DC poiesis remained spatially segregated to different sinusoids. Csf1 deletion in the vasculature prevented generation of MDP and dendritic cells in response to infection. In summary we have developed strategies to image and fate map myelopoiesis in situ; revealed spatial segregation of -and distinct clonal architectures for- granulopoiesis and mono/DCpoiesis; and identified rare CSF1+ sinusoids that maintain mono/DCpoiesis in the steady-state and after infection. These data indicate that there is a specific spatial organization of definitive hematopoiesis and that local cues produced by distinct blood vessels are responsible for this organization. Figure Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 528-528
Author(s):  
Jizhou Zhang ◽  
Qingqing Wu ◽  
Courtney Johnson ◽  
André Olsson ◽  
Anastasiya Slaughter ◽  
...  

Monocytes, macrophages and dendritic cells are indispensable for innate immunity. Myelopoiesis takes place in the bone marrow (BM) through differentiation of a common myeloid progenitor into monocyte dendritic cell progenitors (MDP) or granulocyte monocyte progenitors (GMP). MDP differentiate into myeloid progenitors (MoP) or common dendritic progenitors (cDP) that give rise to monocytes or dendritic cells (DC) repectively. GMP can also generate neutrophil-like monocytes via an intermediate monocyte progenitor [MP; Immunity. 2017 47(5):890] and neutrophils via a granulocyte progenitor (GP). CSF1 (M-CSF) is the key cytokine that regulates monopoiesis, CSF1 loss causes profound defects in monocyte, dendritic cell, macrophage and osteoclast generation. Classical studies using Csf1-/- chimeric mice demonstrated that CSF1 is produced by BM stromal cells. However the identity of these CSF1-producing cells is unknown. A major limitation is the lack of immunofluorescence protocols to map progenitor interaction with candidate niches. We have found that CD11b-Ly6C-CD117+CD115+ cells are MDP; Lin-Ly6C-CD117+CD16/32+CD115- cells are GMP; CD11b-Ly6C+CD117+CD115+ are MP/MoP; CD11b-Ly6C+CD117+CD115- are GP; CD11b+CD115+Ly6Chi and CD11b+CD115+Ly6Clo cells are classical and non-classical monocytes; and MHCIIhi reticular cells are DC. We used the markers above to map the 3D position of every myeloid cell in the sternum and assessed the relationships between myeloid progenitors, their offspring and candidate niches in situ with single cell resolution. To test whether the interactions observed were specific we obtained the X, Y and Z coordinates for every hematopoietic cell in the sternum (detected using αCD45 and αTer119). We then used these coordinates to randomly place each type of myeloid cell, at the same frequencies found in vivo, through the BM to generate a random distribution for each myeloid cell type. HSC localize to sinusoidal, arteriolar and endosteal niches. However, myeloid progenitors are exclusively perisinusoidal (mean MDP distance to sinusoids, arterioles, and endosteum observed 5, 134, and 105μm vs 9, 86, and 69µm in the random simulation). Myeloid progenitors rarely localize with HSC indicating that progenitors abandon the HSC niche upon differentiation. Strikingly, we found that granulopoiesis, monopoiesis and DCpoiesis occur in distinct sinusoidal locations and that MDP are tightly associated with sinusoids, dendritic cells and Ly6Clo monocytes (2.0 DC and 4.4 Ly6Clo monocytes observed within 50µm of an MDP vs 0.9 DC and 1.8 Ly6Clo monocytes in the random simulation p=0.02 and p<0.0001) but not with MoP/MP or Ly6Chi classical monocytes. The results above suggest that the stromal cells that provide the signals that regulate MDP will localize to the sinusoids. Analyses of Csf1 expression in two recently published scRNAseq studies of BM stroma showed that perivascular stromal cells and osteoblastic cells are the major CSF1 sources with sinusoidal endothelial cells expressing much lower levels. Csf1 deletion in perivascular cells using LepR-cre mice and in osteoblastic cells using Ocn-cre mice did not impact Ly6Chi classical or Ly6Clo non-classical monocytes in peripheral blood. We also did not find any defects in BM MDP, GMP, MoP numbers or colony forming activity or in monocyte or dendritic cell numbers. In sharp contrast we found that conditional Csf1 deletion in endothelial cells using Cdh5-cre mice led to a 3.9-fold defect in Ly6Clo non-classical monocytes in the blood (1.89 vs 0.47 x105/ml in the +/- controls vs Cdh5-cre:Csf1-/Δ mice; p=0.03). In the BM these mice showed a 1.4 reduction in MDP numbers (0.72 vs 0.5x104/femur; p=0.04) further compounded by a 2.7-fold loss in MDP-derived CFU-M (22 vs 8 colonies/100 cells; p=0.009) indicating a dramatic reduction in MDP function. This in turn led to a 2.3-fold reduction in Ly6Clo non classical monocytes (9.5 vs 4.1x104/femur; p=0.01) and a 1.2-fold reduction in cDC (2.7 vs 2.1 x104cDC/femur p=0.005). In summary we have imaged for the first time myeloid progenitors; mapped their differentiation into mature myeloid cells; quantified their interaction with candidate niche cells; showed that sinusoids are the exclusive site of monocyte and dendritic cell production; and demonstrated that endothelial cells are a niche that regulates MDP numbers and function via CSF1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2001 ◽  
Vol 97 (11) ◽  
pp. 3333-3341 ◽  
Author(s):  
Markus G. Manz ◽  
David Traver ◽  
Toshihiro Miyamoto ◽  
Irving L. Weissman ◽  
Koichi Akashi

It has been proposed that there are at least 2 classes of dendritic cells (DCs), CD8α+ DCs derived from the lymphoid lineage and CD8α− DCs derived from the myeloid lineage. Here, the abilities of lymphoid- and myeloid-restricted progenitors to generate DCs are compared, and their overall contributions to the DC compartment are evaluated. It has previously been shown that primitive myeloid-committed progenitors (common myeloid progenitors [CMPs]) are efficient precursors of both CD8α+ and CD8α− DCs in vivo. Here it is shown that the earliest lymphoid-committed progenitors (common lymphoid progenitors [CLPs]) and CMPs and their progeny granulocyte-macrophage progenitors (GMPs) can give rise to functional DCs in vitro and in vivo. CLPs are more efficient in generating DCs than their T-lineage descendants, the early thymocyte progenitors and pro-T cells, and CMPs are more efficient DC precursors than the descendant GMPs, whereas pro-B cells and megakaryocyte-erythrocyte progenitors are incapable of generating DCs. Thus, DC developmental potential is preserved during T- but not B-lymphoid differentiation from CLP and during granulocyte-macrophage but not megakaryocyte-erythrocyte development from CMP. In vivo reconstitution experiments show that CLPs and CMPs can reconstitute CD8α+ and CD8α− DCs with similar efficiency on a per cell basis. However, CMPs are 10-fold more numerous than CLPs, suggesting that at steady state, CLPs provide only a minority of splenic DCs and approximately half the DCs in thymus, whereas most DCs, including CD8α+ and CD8α− subtypes, are of myeloid origin.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2273-2273
Author(s):  
Fumihiko Ishikawa ◽  
Tadafumi Iino ◽  
Hiroaki Niiro ◽  
Shuro Yoshida ◽  
Toshihiro Miyamoto ◽  
...  

Abstract Dendritic cells play a key role in host defense by presenting exogenous antigens to T cells. Two dendritic cell subsets, conventional dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs), express distinct repertoire of Toll-like-receptors and recognize different antigens. We previously reported that murine cDCs and pDCs differentiate via either the myeloid or the lymphoid pathway (Shigematsu et al. Immunity ). It is, however, still unclear whether human cDCs and pDCs develop from myeloid, lymphoid or both lineages. In order to analyze the in vivo differentiation of human dendritic cells, we employed the newly-developed xenotrasplant assay system which utilizes newborn NOD-scid/IL2rgnull mice (Ishikawa et al., Blood, in press). Transplantation of 104 Lin-CD34+CD38- hematopoietic stem cells into sublethally irradiated newborn NOD-scid/IL2rgnull mice resulted in generation of all hematopoietic and lymphoid components for a long-term via physiological intermediates such as common myeloid progenitors (CMP) and common lymphoid progenitors (CLP). We found that in this system, dendritic cell subcomponents such as hCD11c+hIL3Ralow cDCs and hCD11c-hIL3Rahigh pDCs, efficiently developed in recipients’ bone marrow, spleen and peripheral blood. To elucidate the origin of human mDCs and pDCs, we purified CMP or CLP from the cord blood, and transplanted these cells into sublethally irradiated newborn NOD-scid/IL2rgnull mice via facial vein. At 4-6 weeks post-transplantation, CMP gave rise only to myeloid cells such as erythroid cells, platelets and granulocytes, while CLP exclusively generated T, B and NK cells. Interestingly, in either mouse group injected with CMP or CLP, cDCs and pDCs were easily detected in the spleen and in the bone marrow. Phenotypic and RT-PCR analyses of purified CMP- or CLP-derived DCs revealed that DCs possessed similar phenotypic characteristics, and transcription profiles in TLR families, BDCA antigens and costimulation molecules, irrespective of their lineage origin. Thus, human cDCs and pDCs develop through both myeloid and lymphoid pathways as in case of mouse hematopoiesis. Further characterization of DCs of different lineage origin is currently performed by microarray analyses in order to find genes specifically expressed in each DC subset.


2017 ◽  
Vol 2017 ◽  
pp. 1-7 ◽  
Author(s):  
Grzegorz Dyduch ◽  
Katarzyna Ewa Tyrak ◽  
Anna Glajcar ◽  
Joanna Szpor ◽  
Magdalena Ulatowska-Białas ◽  
...  

Background. Dendritic cells could be involved in immune surveillance of highly immunogenic tumors such as melanoma. Their role in the progression melanocytic nevi to melanoma is however a matter of controversy. Methods. The number of dendritic cells within epidermis, in peritumoral zone, and within the lesion was counted on slides immunohistochemically stained for CD1a, CD1c, DC-LAMP, and DC-SIGN in 21 of dysplastic nevi, 27 in situ melanomas, and 21 invasive melanomas. Results. We found a significant difference in the density of intraepidermal CD1c+ cells between the examined lesions; the mean CD1c cell count was 7.00/mm2 for invasive melanomas, 2.94 for in situ melanomas, and 13.35 for dysplastic nevi. The differences between dysplastic nevi and melanoma in situ as well as between dysplastic nevi and invasive melanoma were significant. There was no correlation in number of positively stained cells between epidermis and dermis. We did not observe any intraepidermal DC-LAMP+ cells neither in melanoma in situ nor in invasive melanoma as well as any intraepidermal DC-SIGN+ cells in dysplastic nevi. Conclusion. It was shown that the number of dendritic cells differs between dysplastic nevi, in situ melanomas, and invasive melanomas. This could eventually suggest their participation in the development of melanoma.


2002 ◽  
Vol 196 (8) ◽  
pp. 1091-1097 ◽  
Author(s):  
Kang Liu ◽  
Tomonori Iyoda ◽  
Marzena Saternus ◽  
Yukino Kimura ◽  
Kayo Inaba ◽  
...  

Peripheral immune tolerance is believed to be induced by the processing and presentation of self-tissues that die during physiologic tissue turnover. To examine the mechanism that mediates tolerance, we injected mice with dying syngeneic TAP−/− splenocytes loaded with small amounts of the protein antigen, ovalbumin (OVA). After ingestion and presentation of cell-associated OVA by the CD8+ subset of dendritic cells in situ, large numbers of antigen-reactive, CD8+ T cell receptor (TCR) transgenic T lymphocytes were driven into cell cycle, but then the T cells were deleted. The animals were also tolerant to challenge with OVA in complete Freund's adjuvant. An agonistic anti-CD40 monoclonal antibody was then administered together with the OVA-loaded splenocytes, so that the dendritic cells in the recipient mice would mature. In contrast to observations made in the steady state, the antigen-reactive T cells expanded in numbers for 1–2 wk and produced large amounts of interleukin 2 and interferon γ, while the animals retained responsiveness to antigen rechallenge. The specific tolerance that develops when dendritic cells process self tissues in the steady state should prevent or reduce the development of autoimmunity when dying cells are subsequently processed during infection.


2015 ◽  
Vol 2015 ◽  
pp. 1-14 ◽  
Author(s):  
Arunima Chatterjee ◽  
Péter Gogolak ◽  
Hervé M. Blottière ◽  
Éva Rajnavölgyi

Vitamin A plays an essential role in the maintenance of gut homeostasis but its interplay with chemokines has not been explored so far. Using anin vitromodel system we studied the effects of human colonic epithelial cells (Caco2, HT-29, and HCT116) derived inflammatory stimuli on monocyte-derived dendritic cells and macrophages. Unstimulated Caco2 and HT-29 cells secreted CCL19, CCL21, and CCL22 chemokines, which could attract dendritic cells and macrophages and induced CCR7 receptor up-regulation by retinoic-acid resulting in dendritic cell migration. The chemokines Mk, CXCL16, and CXCL7 were secreted by all the 3 cell lines tested, and upon stimulation by IL-1βor TNF-αthis effect was inhibited by ATRA but had no impact on CXCL1, CXCL8, and CCL20 secretion in response to IL-1β. In the presence of ATRA the supernatants of these cells induced CD103 expression on monocyte-derived dendritic cells and when conditioned by ATRA and cocultured with CD4+T-lymphocytes they reduced the proportion of Th17 T-cells. However, in the macrophage-T-cell cocultures the number of these effector T-cells was increased. Thus cytokine-activated colonic epithelial cells trigger the secretion of distinct combinations of chemokines depending on the proinflammatory stimulus and are controlled by retinoic acid, which also governs dendritic cell and macrophage responses.


2013 ◽  
Vol 31 (1) ◽  
pp. 563-604 ◽  
Author(s):  
Miriam Merad ◽  
Priyanka Sathe ◽  
Julie Helft ◽  
Jennifer Miller ◽  
Arthur Mortha

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 297-297
Author(s):  
Qingqing Wu ◽  
Jizhou Zhang ◽  
Courtney Johnson ◽  
Benjamin Weinhaus ◽  
Anastasiya Slaughter ◽  
...  

Abstract Knowledge of the anatomy of each tissue and the relationships between progenitors and daughter cells is necessary to understand physiology and pathology. The anatomy of hematopoiesis in the marrow remains largely unknown. Here we identify strategies to image all steps of blood cell production in the mouse sternum using confocal microscopy. We show that long-distance migration of multipotent progenitors, lineage-committed progenitor recruitment to vessels, and generation of lineage-specific oligoclonal structures that are the main production sites for immature cells are key features of the anatomy of blood production. This structural organization is extremely durable and resilient to insults as it was maintained after hemorrhage, Listeria monocytogenes infection, and aging (80-weeks). Importantly, this anatomy is also enabled with local plasticity as production sites for each blood lineage selectively expand/contract in respond to insults followed by a return to homeostasis. We used immunophenotyping to identify fifty-six surface markers that can be combined to image any populations of interest. For example, we found that ESAM is selectively expressed in 100% of LT-HSC, 90% of ST-HSC, 70% of MPP2 and MPP3, 30% of MPP4, 10% of CMP, and 90% of MkP and megakaryocytes but absent in more mature cells. Transplantation experiments revealed that all functional LT- and ST-HSC, MPP2, MPP3 and CMP were contained -exclusively- in the ESAM positive fraction (p<0.05 when compared with ESAM- cells n= 7 mice per group). ESAM + MPP4 displayed 5-fold more engraftment than ESAM - MPP4 (p<0.05). Combining ESAM with classical HSPC markers allowed imaging of all LT-HSC, ST-HSC, MkP, Pre-MegE, MPP2 and a mixed population of MPP3, CMP and MPP4. We developed similar strategies to map erythropoiesis (Pre-MegE → Pre-CFU-E → CFU-E → early erythroblast → late erythroblast → reticulocyte → erythrocyte) and lymphopoiesis (CLP→ PreProB → ProB →PreB→ Immature B). All strategies allowed clonal fate-mapping using Ubc-cre ERT2:confetti mice. In this model tamoxifen treatment leads to irreversible expression of one out of four fluorescent proteins. We found that multipotent and oligopotent progenitors are found as single cells, evenly distributed through the marrow (e.g. mean LT-HSC distance to closest ST-HSC, MPP2, MPP3, MkP, Pre-MegE >100 μm, no different from random simulations, n=41 LT-HSC from 5 sternum sections of 4 wild-type mice) and are clonally unrelated between them. Multipotent and oligopotent progenitors reside near sinusoids (mean distance =9.7 μm) but this association is not different from that observed for random cells. In contrast, as progenitors become lineage-restricted, they localize to arterioles (for lymphoid progenitors) or sinusoids (all other progenitors) where they enter oligoclonal structures that are the main production sites for immature cells in each lineage. Each production site has distinct architectures: lymphoid sites are characterized by tight clusters of PreProB cells surrounding CLP; erythroid sites are characterized by strings of 4-21 CFU-E decorating the surface of sinusoids with early erythroblasts differentiating orthogonally to the vessel surface; in megakaryocyte sites one or two megakaryocyte progenitors produce megakaryocytes that decorate blood vessels over large (>200μm 3) marrow regions. We previously showed that production sites for neutrophils contain 1 or 2 granulocyte progenitors tightly clustered with preneutrophils and that sites for monocytes/dendritic cells contain loose clusters of dendritic cells surrounding monocyte dendritic cell progenitors (Zhang Nature 2021). This spatial architecture is durable and resilient and is maintained after acute challenge via phlebotomy, L. monocytogenes infection, or physiological aging (80-week-old mice). However, we also observed plasticity of production sites. Two days after phlebotomy we found increases in erythroid site numbers (368 vs 945 per mm 3, p<0.05). These expansions were reversed by day 8 after phlebotomy. Similarly, infection led to increases in the size of neutrophil and dendritic cell production sites (~2-fold by day 6 post-infection) but these changes are reverted by day 20 post-infection. In summary, we have developed strategies that allow imaging of differentiation in situ and defined a complex - but durable and plastic- anatomy for the hematopoietic tissue. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Jizhou Zhang ◽  
Qingqing Wu ◽  
Courtney B. Johnson ◽  
Andre Olsson ◽  
Anastasiya Slaughter ◽  
...  

SummaryIn contrast to virtually all other tissues in the body the anatomy of differentiation in the bone marrow remains unknown. This is due to the lack of strategies to examine blood cell production in situ, which are required to better understand differentiation, lineage commitment decisions, and to define how spatial organizing cues inform tissue function. Here we developed imaging approaches to map all myeloid cells in whole bones and generated 3D atlases of granulocyte and monocyte/dendritic cell differentiation during homeostasis. We found that myeloid progenitors leave the hematopoietic stem cell niche during differentiation. Granulocyte and monocyte dendritic cell progenitors (MDP) do not interact, instead they localize to different sinusoids where they give rise to clusters of immature cells. MDP cluster with Ly6Clo monocytes and conventional dendritic cells; these localize to a unique subset of colony stimulating factor 1 (CSF1, the major regulator of monopoiesis1) -expressing sinusoids. Csf1 deletion in the vasculature disrupted the MDP clusters and their interaction with sinusoids, leading to reduced MDP numbers and differentiation ability, with subsequent loss of peripheral Ly6Clo monocytes and dendritic cells. These data indicate that there is a specific spatial organization of definitive hematopoiesis and that local cues produced by distinct blood vessels are responsible for this organization. These maps provide a blueprint for in situ analyses of hematopoiesis in blood disorders.


Sign in / Sign up

Export Citation Format

Share Document