scholarly journals Quantification of bone marrow interstitial pH and calcium concentration by intravital ratiometric imaging

Author(s):  
Shu-Chi Yeh ◽  
Jue Hou ◽  
Juwell Wu ◽  
Fernando Camargo ◽  
Charles Lin

Abstract The fate of hematopoietic stem cells (HSCs) in the bone marrow can be directed by microenvironmental factors including extracellular calcium ion concentration (T h can vary significantly with bone remodeling, but the local e around individual HSCs in vivo remains unknown. Here we developed an intravital ratiometric imaging approach to quantify the absolute pH and in the mouse calvarial bone marrow, taking into account the pH sensitivity of the calcium probe and the wavelength-dependent optical loss through bone to ensure unbiased ratiometric analyses. We uncovered substantial heterogeneity in f (1.0 - 3.6 mM) surrounding steady-state HSCs. While the lowest a (0.3 mM) was found in regions dominated by bone formation, HSCs were not found in those locations. This work thus established a tool to further investigate t and pH in the HSC niche under malignant or stressed conditions and can be broadly applied to other tissue types.

2015 ◽  
Vol 112 (5) ◽  
pp. 1350-1355 ◽  
Author(s):  
Hee-Sun Han ◽  
Elisabeth Niemeyer ◽  
Yuhui Huang ◽  
Walid S. Kamoun ◽  
John D. Martin ◽  
...  

Multiplexed, phenotypic, intravital cytometric imaging requires novel fluorophore conjugates that have an appropriate size for long circulation and diffusion and show virtually no nonspecific binding to cells/serum while binding to cells of interest with high specificity. In addition, these conjugates must be stable and maintain a high quantum yield in the in vivo environments. Here, we show that this can be achieved using compact (∼15 nm in hydrodynamic diameter) and biocompatible quantum dot (QD) -Ab conjugates. We developed these conjugates by coupling whole mAbs to QDs coated with norbornene-displaying polyimidazole ligands using tetrazine–norbornene cycloaddition. Our QD immunoconstructs were used for in vivo single-cell labeling in bone marrow. The intravital imaging studies using a chronic calvarial bone window showed that our QD-Ab conjugates diffuse into the entire bone marrow and efficiently label single cells belonging to rare populations of hematopoietic stem and progenitor cells (Sca1+c-Kit+ cells). This in vivo cytometric technique may be useful in a wide range of structural and functional imaging to study the interactions between cells and between a cell and its environment in intact and diseased tissues.


Blood ◽  
1996 ◽  
Vol 87 (10) ◽  
pp. 4136-4142 ◽  
Author(s):  
I Kawashima ◽  
ED Zanjani ◽  
G Almaida-Porada ◽  
AW Flake ◽  
H Zeng ◽  
...  

Using in utero transplantation into fetal sheep, we examined the capability of human bone marrow CD34+ cells fractionated based on Kit protein expression to provide long-term in vivo engraftment. Twelve hundred to 5,000 CD34+ Kit-, CD34+ Kit(low), and CD34+ Kit(high) cells were injected into a total of 14 preimmune fetal sheep recipients using the amniotic bubble technique. Six fetuses were killed in utero 1.5 months after bone marrow cell transplantation. Two fetuses receiving CD34+ Kit(low) cells showed signs of engraftment according to analysis of CD45+ cells in their bone marrow cells and karyotype studies of the colonies grown in methylcellulose culture. In contrast, two fetuses receiving CD34+ Kit(high) cells and two fetuses receiving CD34+ Kit- cells failed to show evidence of significant engraftment. Two fetuses were absorbed. A total of six fetuses receiving different cell populations were allowed to proceed to term, and the newborn sheep were serially examined for the presence of chimerism. Again, only the two sheep receiving CD34+ Kit(low) cells exhibited signs of engraftment upon serial examination. Earlier in studies of murine hematopoiesis, we have shown stage-specific changes in Kit expression by the progenitors. The studies of human cells reported here are in agreement with observations in mice, and indicate that human hematopoietic stem cells are enriched in the Kit(low) population.


Blood ◽  
1997 ◽  
Vol 89 (11) ◽  
pp. 3919-3924 ◽  
Author(s):  
Jean C.Y. Wang ◽  
Monica Doedens ◽  
John E. Dick

Abstract We have previously reported the development of in vivo functional assays for primitive human hematopoietic cells based on their ability to repopulate the bone marrow (BM) of severe combined immunodeficient (SCID) and nonobese diabetic/SCID (NOD/SCID) mice following intravenous transplantation. Accumulated data from gene marking and cell purification experiments indicate that the engrafting cells (defined as SCID-repopulating cells or SRC) are biologically distinct from and more primitive than most cells that can be assayed in vitro. Here we demonstrate through limiting dilution analysis that the NOD/SCID xenotransplant model provides a quantitative assay for SRC. Using this assay, the frequency of SRC in cord blood (CB) was found to be 1 in 9.3 × 105 cells. This was significantly higher than the frequency of 1 SRC in 3.0 × 106 adult BM cells or 1 in 6.0 × 106 mobilized peripheral blood (PB) cells from normal donors. Mice transplanted with limiting numbers of SRC were engrafted with both lymphoid and multilineage myeloid human cells. This functional assay is currently the only available method for quantitative analysis of human hematopoietic cells with repopulating capacity. Both CB and mobilized PB are increasingly being used as alternative sources of hematopoietic stem cells in allogeneic transplantation. Thus, the findings reported here will have important clinical as well as biologic implications.


Blood ◽  
2011 ◽  
Vol 117 (2) ◽  
pp. 451-458 ◽  
Author(s):  
Mamiko Noda ◽  
Yoshiki Omatsu ◽  
Tatsuki Sugiyama ◽  
Shinya Oishi ◽  
Nobutaka Fujii ◽  
...  

Abstract Natural killer (NK) cells are granular lymphocytes that are generated from hematopoietic stem cells and play vital roles in the innate immune response against tumors and viral infection. Generation of NK cells is known to require several cytokines, including interleukin-15 (IL-15) and Fms-like tyrosine kinase 3 ligand, but not IL-2 or IL-7. Here we investigated the in vivo role of CXC chemokine ligand-12 (CXCL12) and its primary receptor CXCR4 in NK-cell development. The numbers of NK cells appeared normal in embryos lacking CXCL12 or CXCR4; however, the numbers of functional NK cells were severely reduced in the bone marrow, spleen, and peripheral blood from adult CXCR4 conditionally deficient mice compared with control animals, probably resulting from cell-intrinsic CXCR4 deficiency. In culture, CXCL12 enhanced the generation of NK cells from lymphoid-primed multipotent progenitors and immature NK cells. In the bone marrow, expression of IL-15 mRNA was considerably higher in CXCL12-abundant reticular (CAR) cells than in other marrow cells, and most NK cells were in contact with the processes of CAR cells. Thus, CXCL12-CXCR4 chemokine signaling is essential for NK-cell development in adults, and CAR cells might function as a niche for NK cells in bone marrow.


1987 ◽  
Vol 5 (3) ◽  
pp. 231-241 ◽  
Author(s):  
Vincent S. Gallicchio ◽  
Thomas D. Watts ◽  
George P. Casale ◽  
Philip M. Bartholomew

Blood ◽  
2007 ◽  
Vol 110 (7) ◽  
pp. 2276-2285 ◽  
Author(s):  
Maria De La Luz Sierra ◽  
Paola Gasperini ◽  
Peter J. McCormick ◽  
Jinfang Zhu ◽  
Giovanna Tosato

The mechanisms underlying granulocyte-colony stimulating factor (G-CSF)–induced mobilization of granulocytic lineage cells from the bone marrow to the peripheral blood remain elusive. We provide evidence that the transcriptional repressor growth factor independence-1 (Gfi-1) is involved in G-CSF–induced mobilization of granulocytic lineage cells from the bone marrow to the peripheral blood. We show that in vitro and in vivo G-CSF promotes expression of Gfi-1 and down-regulates expression of CXCR4, a chemokine receptor essential for the retention of hematopoietic stem cells and granulocytic cells in the bone marrow. Gfi-1 binds to DNA sequences upstream of the CXCR4 gene and represses CXCR4 expression in myeloid lineage cells. As a consequence, myeloid cell responses to the CXCR4 unique ligand SDF-1 are reduced. Thus, Gfi-1 not only regulates hematopoietic stem cell function and myeloid cell development but also probably promotes the release of granulocytic lineage cells from the bone marrow to the peripheral blood by reducing CXCR4 expression and function.


Blood ◽  
1985 ◽  
Vol 66 (6) ◽  
pp. 1460-1462 ◽  
Author(s):  
ME Pietrzyk ◽  
GV Priestley ◽  
NS Wolf

It was found in a long-term bromodeoxyuridine (BrdU) infusion study that two or more different subpopulations of bone marrow stem cells exist in mice. One of these subpopulations appears to be noncycling and forms approximately 10% of eight-day CFU-S. Another one, a subpopulation of slowly cycling bone marrow cells, is represented as 14- day CFU-S. The 14-day CFU-S have a regular increment in the percentage of the subpopulation entering the cycle over time, with a cell generation half-time of 21 days. The cycling status in these experiments was ascertained by in vivo continuous long-term BrdU infusion. An improved method is presented for long-term BrdU infusion with UV killing of cycled cells.


2021 ◽  
Author(s):  
Zixian Liu ◽  
Jinhong Wang ◽  
Miner Xie ◽  
Peng Wu ◽  
Yao Ma ◽  
...  

Hematopoietic stem cells (HSCs) have been considered to progressively lose their self-renewal and differentiation potentials prior to the commitment to each blood lineage. However, recent studies have suggested that megakaryocyte progenitors are generated at the level of HSCs. In this study, we newly identified early megakaryocyte lineage-committed progenitors (MgPs) in CD201-CD48- cells and CD48+ cells separated from the CD150+CD34-Kit+Sca-1+Lin- HSC population of the bone marrow in C57BL/6 mice. Single-cell transplantation and single-cell colony assay showed that MgPs, unlike platelet-biased HSCs, had little repopulating potential in vivo, but formed larger megakaryocyte colonies in vitro (on average eight megakaryocytes per colony) than did previously reported megakaryocyte progenitors (MkPs). Single-cell RNA-sequencing supported that these MgPs lie between HSCs and MkPs along the megakaryocyte differentiation pathway. Single-cell colony assay and single-cell RT-PCR analysis suggested the coexpression of CD41 and Pf4 is associated with megakaryocyte colony-forming activity. Single-cell colony assay of a small number of cells generated from single HSCs in culture suggested that MgPs are not direct progeny of HSCs. In this study, we propose a differentiation model in which HSCs give rise to MkPs through MgPs.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 276-276
Author(s):  
Xin GAO ◽  
Philip Boulais ◽  
Masato Tanaka ◽  
Christopher Richard Marlein ◽  
Paul S. Frenette

Hematopoietic stem cells (HSCs), residing at the apex of the hematopoietic hierarchy, are a rare and heterogeneous cell population. Although HSC subsets with various repopulation capacities and lineage bias have been identified, there is no available information about whether each HSC has an equal chance of being mobilized or whether there are specific pools of mobilizable or non-mobilizable HSCs in bone marrow (BM). Here, we identify a BM resident HSC subset based on the expression of the macrophage marker F4/80. Flow cytometry analysis revealed that F4/80 was expressed on 75% of HSCs (Lin- Sca1+cKit+CD150+CD34-; hereafter referred to as HSCM) in the BM. In competitive transplantation assays, HSCM exhibited significantly lower engraftment potential, but no lineage bias, compared to F4/80- HSCs (HSCM: 19.7±4.8%; F4/80- HSC: 37.1±4.5%, P<0.05). We next analyzed the ability of HSCM and F4/80- HSCs to be mobilized after G-CSF treatment and found that F4/80- HSCs, but not HSCM, were exclusively mobilized (F4/80- HSC: 5042±912.5; HSCM: 6.6±6.6 per mL blood, P<0.001). Similar selective mobilization was observed after administration of AMD3100 or macrophage depletion using clodronate liposomes. To confirm this observation in a genetic model, we crossed transgenic mice expressing Cre recombinase knocked into the Cd169 locus, a marker of BM macrophages (MΦ), with ROSA26-loxP-stop-loxP-tdTomato (CD169/Tomato). We found CD169/Tomato labelled a large fraction of HSCM (31.7±8.4%), in contrast to F4/80- HSCs which were largely unlabelled (2.1±1.2%). Remarkably, CD169/Tomato+ HSCs were not mobilized into the circulation after G-CSF treatment (CD169/Tomato- HSC: 4573±1416; CD169/Tomato+ HSC: 90.6±90.6 per mL blood, P<0.01). Although HSCs are reported to express Csf1r, the expression of macrophage markers on HSCs (F4/80 and CD169) was unexpected. To ascertain the expression origin, we transplanted CD169/Tomato+ and CD169/Tomato- sorted HSCs into lethally irradiated CD45.1 congenic mice. If HSCs expressed CD169, we would expect that donor HSC tomato expression would be retained. However, to our surprise, we found that the proportion of Tomato+ HSCs was higher in the CD169/Tomato- group, suggesting that F4/80 and CD169 expression may be acquired, rather than expressed within HSCs. Indeed, co-culture of TdTomato+ BM MΦ with GFP+ HSCs consistently revealed the acquisition of tdTomato (GFP+ TdTomato+: 9.9±1.9%). Transwell experiments revealed that cell contact was important as GFP+ cells did not show any TdTomato expression in the absence of direct cell contact (GFP+TdTomato+: 0%). To investigate the mechanism of HSC retention, we analyzed CXCR4 expression on HSCM and F4/80- HSC subsets. These analyses revealed that CXCR4 was expressed at higher expression levels on HSCM compared to F4/80- HSCs (HSCM: 79.4±9.1% CXCR4+;F4/80- HSC: 45.6±8.5% CXCR4+, P<0.001). In accordance with their increased CXCR4 expression, HSCM exhibited a 4.7-fold increase in pERK1/2 after CXCL12 stimulation, while CXCL12 only triggered a modest increase (1.5-fold) in pERK1/2 in F4/80- HSCs, suggesting that HSCM have a higher signaling response to CXCL12, and thus exhibit enhanced BM retention. To further confirm that the cell transfer occurred in vivo, we used xenografted mice in which human HSCs were transplanted into NOG mice. We found a significant presence of murine F4/80 on human CD34+ HSCs in xenografted huNOG mice, confirming the F4/80 transfer from host MΦ into human HSCs in vivo. To investigate further the mechanism of cell transfer, we carried out co-culture experiments of GFP+ lineage-depleted BM cells with TdTomato+ MΦ in the presence of inhibitors known to prevent cell transfer through membrane and cytoskeleton remodeling. Of all conditions tested, only the presence of the GAP junction inhibitor carbenoxolone (CBX) could inhibit the transfer of TdTomato from MΦ to GFP+ cells (control: 21.4±3.0%; CBX: 12.6±2.1%, P<0.001). Furthermore, treatment of wild-type mice with CBX led to a marked suppression of F4/80 transfer in vivo, similar to that observed in vitro, suggesting that GAP junctions are responsible for the direct transfer of cellular content from BM MΦ to HSCs. Our results thus identify a novel mechanism by which macrophages can assign HSC residence in BM. Manipulation of macrophage-mediated transfer may enhance the mobilizable HSC pool and provide a new method to improve HSC yields after mobilization. Disclosures Frenette: Cygnal Therapeutics: Equity Ownership; Ironwood Pharmaceuticals: Research Funding; Albert Einstein College of Medicine, Inc: Patents & Royalties; Pfizer: Consultancy.


Sign in / Sign up

Export Citation Format

Share Document