scholarly journals The Blood Circulating Rare Cell Population. What Is It and What Is It Good for?

Cells ◽  
2020 ◽  
Vol 9 (4) ◽  
pp. 790
Author(s):  
Stefan Schreier ◽  
Wannapong Triampo

Blood contains a diverse cell population of low concentration hematopoietic as well as non-hematopoietic cells. The majority of such rare cells may be bone marrow-derived progenitor and stem cells. This paucity of circulating rare cells, in particular in the peripheral circulation, has led many to believe that bone marrow as well as other organ-related cell egress into the circulation is a response to pathological conditions. Little is known about this, though an increasing body of literature can be found suggesting commonness of certain rare cell types in the peripheral blood under physiological conditions. Thus, the isolation and detection of circulating rare cells appears to be merely a technological problem. Knowledge about rare cell types that may circulate the blood stream will help to advance the field of cell-based liquid biopsy by supporting inter-platform comparability, making use of biological correct cutoffs and “mining” new biomarkers and combinations thereof in clinical diagnosis and therapy. Therefore, this review intends to lay ground for a comprehensive analysis of the peripheral blood rare cell population given the necessity to target a broader range of cell types for improved biomarker performance in cell-based liquid biopsy.

2014 ◽  
Vol 33 (1) ◽  
pp. 48-54 ◽  
Author(s):  
Yoichi Ezura ◽  
Junji Nagata ◽  
Masashi Nagao ◽  
Hiroaki Hemmi ◽  
Tadayoshi Hayata ◽  
...  

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 692-692
Author(s):  
Matthew S. Davids ◽  
Jennifer R. Brown ◽  
Adrian Wiestner ◽  
Anthony G. Letai

Abstract Abstract 692 Treatments for chronic lymphocytic leukemia (CLL) often kill malignant cells in the peripheral blood, but the disease inevitably relapses in the lymph nodes or bone marrow. BH3 profiling was developed in our laboratory to assess the degree to which malignant cells are primed to undergo apoptosis by the mitochondrial pathway, and to identify the anti-apoptotic proteins on which these cells depend for their survival. We hypothesized that BH3 profiling can help elucidate mechanisms underlying stromal-mediated resistance to the BH3-mimetic ABT-737 in CLL. BH3 profiling was performed by exposing malignant CD19+ B cells from 15 CLL patients to a panel of BH3-domain peptides, and the cell death induced was quantified by JC-1 based FACS to assess mitochondrial outer membrane permeabilization, as previously described (Ryan et al., PNAS 2010). To simulate lymph node and bone marrow microenvironments, we co-cultured CLL cells from a subset of these patients for 24 hours in the presence of IL-4 with CD154+ fibroblasts and with HS5 cells, respectively, and then repeated BH3 profiling. The status of the chemokine receptor CXCR4, which can serve as a marker for the residence of CLL cells in stromal microenvironments, was also evaluated by FACS. Additional co-culture experiments were done in the presence or absence of ABT-737 at 100 nM, and CLL cell viability was assessed at 24 hours by Annexin-PI. We also performed BH3 profiling on 7 additional CLL patients with matched peripheral blood, lymph node, and bone marrow samples. Circulating malignant CLL cells were highly primed to undergo apoptosis, and their survival was mainly dependent on Bcl-2, and to a lesser degree Mcl-1. CXCR4 decreased on CLL cells co-cultured for 24 hours with CD154+ fibroblasts (38.6%) compared to cells cultured with parental controls (76.3%) (p = 0.030), but did not decrease on cells cultured with HS5 cells (87.1%) (p > 0.05). When CLL cells were co-cultured with CD154+ fibroblasts in the presence of ABT-737, mean CLL cell viability by Annexin-PI increased to 85.1% compared to 31.8% (p < 0.001) in cells co-cultured with parental controls. BH3 profiling revealed that CD154+ fibroblast exposure led to decreased CLL cell mitochondrial depolarization in response to Bim, Noxa, Hrk, and particularly to ABT-737 (see figure). In contrast, CLL cells exposed to HS5 cells had unchanged CXCR4 status, but still had a decrease in apoptotic priming, which was observed in response to an even broader range of BH3-domain peptides, including Puma and Bmf. When gating on the whole CLL cell population, the pattern and degree of apoptotic priming was similar in matched peripheral blood, lymph node, and bone marrow biopsy samples from 7 additional patients. Interestingly, gating on CXCR4 status revealed heterogeneity in apoptotic priming in the different microenvironments, with a subset of patients showing that CXCR4- bone marrow-derived CLL cells were less primed than their CXCR4+ counterparts. Overall, BH3 profiling demonstrated that circulating primary CLL cells are highly primed to undergo apoptosis, and depend predominantly on the anti-apoptotic protein Bcl-2 for their survival. CLL cells co-cultured with lymph node-like stroma had decreased CXCR4 surface expression and became resistant to ABT-737. BH3 profiling demonstrated that this resistance was accompanied by decreased apoptotic priming in response to several BH3-domain peptides. An even broader decrease in apoptotic priming was observed in response to co-culture with a bone marrow-like microenvironment, apparently unrelated to changes in CXCR4 status. Matched peripheral blood, lymph node, and bone marrow CLL patient samples had similar BH3 profiles overall, but some patients showed decreased apoptotic priming in CXCR4- CLL cells, which likely represent the true bone marrow resident CLL cell population. This heterogeneity in mitochondrial priming may help to explain some of the resistance to therapy observed in bone marrow and lymph nodes as compared to peripheral blood. Disclosures: Letai: Eutropics Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1461-1461
Author(s):  
Serena Marotta ◽  
Giovanna Giagnuolo ◽  
Giulia Scalia ◽  
Maddalena Raia ◽  
Santina Basile ◽  
...  

Abstract Abstract 1461 The differential diagnosis of myelofibrotic disorders encompasses chronic primary myelofibrosis (PMF), myelodysplastic syndromes with fibrosis (MDS-F), acute panmyelosis with myelofibrosis (APMF) and acute megakaryoblastic leukemia (AMKL). Most of these conditions are recognized as distinct entities by the WHO 2008 revised classification of myeloid neoplasms; however, the WHO admits that often a definitive diagnosis is problematic, mostly because of specimens with insufficient cellularity (e.g., “dry tap”). Nevertheless, the correct identification of the most aggressive fibrotic disorders (APMF and AMKL) remains crucial, given their poor prognosis and subsequent need of intensive treatment (including transplantation). Even the most recent molecular studies did not result in any contribution in the differential diagnosis. Here we report our experience on a cohort of about 300 patients who were admitted in our bone marrow failure unit because of cytopenia in the last 7 years. All these patients were evaluated by standard peripheral blood and bone marrow cytology, karyotype analysis and bone marrow thephine biopsy, aiming to a definitive hematological diagnosis. Flow cytometry analysis was performed at initial presentation and then serially during the follow up on both peripheral blood and bone marrow aspirate. All patients were classified according to the WHO 2008 revised classification of myeloid neoplasms, and received the best standard treatment based on the specific disease, age and comorbidities. This report focuses on 8 patients who shared a unique flow cytometry finding of an aberrant megakaryocyte-derived cell population, which seems associated with a distinct disease evolution. Two of these patients received the diagnosis of AMKL according to bone marrow aspirate and trephine biopsy; the karyotype was complex in one case (monosomal karyotype, including a 5q-), whereas no Jak-2 mutation or any other genetic lesions could be demonstrated. Their blast cells were CD34+, CD38+, CD45+, CD117+, CD33+, CD13+; in addition, in the peripheral blood, we detected the presence of an aberrant cell population which was CD45-, CD42b+ (CD34+ in one case and CD34- in the other one). In the blood smear, we observed megakaryocyte fragments which likely correspond to this aberrant cell population, as identified by flow cytometry. Other three patients presented with a severe pancytopenia: all of them had a dry tap, and their trephine biopsies documented a massive fibrosis. They had no previous hematological disorder (one suffered from Behcet syndrome), normal karyotype and absence of any typical genetic lesion (i.e., wild-type Jak-2). All of them did not show splenomegaly, increased LDH or leukoerythroblastosis; their peripheral blood smear showed abnormal giant platelets, often resembling megakaryocyte fragments. Flow cytometry documented in the peripheral blood the presence of a distinct population of CD45-, CD42b+, CD61+ cells, which was also CD34+ in one case. These 3 patients were initially classified as PMF, even if APMF could not be ruled out; however, within 6 months they all progressed to AMKL. At this stage, typical CD34+, CD45+ blast cells were accompanied by a progressive increase of CD45+, CD42b+, CD61+ cells. This aberrant megakaryocyte-derived cell population (which could not be demonstrated in patients with thrombocytopenia) was also identified in 3 additional patients, who have a previous history of hematologic disorders: two had a history of pure red cell aplasia (successfully treated by immunosuppressive therapy), and one a 5q- melodysplastic syndrome (responding to lenalidomide, even with transient cytogenetic remission). In all of them we observed the appearance of CD45-, CD42b+ cells in the peripheral blood, which appeared as giant platelets/megakaryocyte fragments in the blood film; this finding within a few weeks was followed by progression to AMKL (5q- was detected in 2 of 3 cases). In conclusion, we demonstrate that aberrant circulating megakaryocyte-derived cells detected by flow cytometry may be useful in the differential diagnosis of myelofibrotic disorders. These giant platelets or megakaryocyte fragments, regardless the initial diagnosis, were associated with early evolution into AMKL, likely representing a surrogate marker for aggressive neoplasms of the megakaryocytic lineage. Disclosures: Risitano: Alexion: Membership on an entity's Board of Directors or advisory committees, Research Funding.


2013 ◽  
Vol 304 (10) ◽  
pp. L639-L645 ◽  
Author(s):  
Dalize M. Squebola-Cola ◽  
Glaucia C. Mello ◽  
Lorenzo Pissinatti ◽  
André A. Schenka ◽  
Gabriel F. Anhê ◽  
...  

Bone marrow (BM) eosinopoiesis is a common feature during allergen exposure in atopic individuals. Airway exposure to staphylococcal superantigens aggravates allergic airway disease and increases the output of BM eosinophils. However, the exact mechanisms regulating eosinophil mobilization and trafficking to the peripheral circulation and airways remain to be elucidated. Therefore, this study aimed to investigate the mechanisms determining the BM eosinopoiesis in allergic mice under exposure to staphylococcal enterotoxin A (SEA). Ovalbumin (OVA)-sensitized male BALB/C mice were intranasally exposed to SEA (1 μg), and at 4, 12, 24, and 48 h later animals were challenged with OVA (10 μg, twice a day). Measurement of IL-5, eotaxin, and granulocyte-macrophage colony-stimulating factor (GM-CSF) levels, flow cytometry for CCR3+, VLA4+, and CCR3+VLA4+, as well as adhesion assays to VCAM-1 were performed in BM. Prior airway exposure to SEA time dependently increased the BM eosinophil number in OVA-challenged mice. Eosinophils gradually disappear from peripheral blood, being recruited over time to the airways, where they achieve a maximal infiltration at 24 h. SEA exposure increased the levels of IL-5 and eotaxin (but not GM-CSF) in BM of OVA-challenged mice. Marked increases in CCR3+and CCR3+VLA4+expressions in BM eosinophils of OVA-challenged mice were observed, an effect largely reduced by prior exposure to SEA. Adhesion of BM eosinophils to VCAM-1 was increased in OVA-challenged mice, but prior SEA exposure abrogated this enhanced cell adhesion. Accumulation of BM eosinophils by airway SEA exposure takes place through IL-5- and CCR3-dependent mechanisms, along with downregulation of CCR3/VL4 and impaired cell adhesion to VCAM-1.


Blood ◽  
1975 ◽  
Vol 46 (1) ◽  
pp. 91-102
Author(s):  
JK Chamberlain ◽  
L Weiss ◽  
RI Weed

Ultrastructural studies of erythropoietin effects on the bone marrow of control and hypertransfused (65 hct) mice revealed a decrease in adventitial cell cover of the sinus apertures in erythropoietin-treated animals. A more striking finding, however, was the marked inhibition of erythropoietin-induced reticulocytosis by hypertransfusion itself. Hypertransfusion of the erythropoietin-treated animals appeared to decrease the reticulocyte response by inhibiting reticulocyte response by marrow cords in addition to inhibiting erythroid proliferation. This inhibition of reticulocyte response was associated with clustering of reticulocytes around the marrow sinuses which were packed with red cells. Acute lowering of the hematocrit of erythropoietin-treated, hypertransfused animals to normal at the time of maximal reticulocyte response in control animals resulted in more than a twofold increase in reticulocytosis with 2 hr. It is suggested that (1) elevated levels of erythropoietin are associated with a diminution of the normal marrow- peripheral blood barrier, thereby contributing to the premature release of marrow elements and (2) the hematocrit is an important determinant of cell release from the marrow into the peripheral circulation.


2010 ◽  
Vol 2 (2) ◽  
pp. e2010028 ◽  
Author(s):  
Mark E. Johns ◽  
Lynn C. Moscinski ◽  
Lubomir Sokol

We report a case of phenytoin-induced pseudolymphoma in a 28-year-old male with a history of autism and seizure disorder.  The patient presented with bilateral cervical lymphadenopathy that was shown to be moderately to markedly FDG-avid on a whole body PET/CT scan.  Flow cytometry analysis of peripheral blood and bone marrow mononuclear cells detected identical T cell population with aberrant immunophenotype.  Additionally, a TCR beta gene was found to be clonally rearranged in both peripheral blood and bone marrow supporting a clonal origin of atypical T cells. However, no such clonal population of T-cells could be detected in a pathologic specimen obtained from an excisional biopsy of one of the patient’s cervical lymph nodes. After discontinuing the patient’s phenytoin, his lymphadenopathy has nearly completely resolved and circulation clonal T cell population disappeared with 12 months of follow-up.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4140-4140
Author(s):  
Dag Josefsen ◽  
Leiv S. Rusten ◽  
Trond Stokke ◽  
Lise Forfang ◽  
Erlend B. Smeland ◽  
...  

Abstract CD34+ cells isolated from bone marrow include hematopoietic stem cells (HSC) as well as more lineage committed hematopoietic progenitor cells (HPC), demonstrating that CD34+ cells are a relatively heterogeneous cell population. Highly enriched CD34+ cells isolated from peripheral blood (PBPC) after mobilization shows a more immature profile with less expression of lineage restricted markers indicating that CD34+ cells from PBPC are a more homogenous immature cell population than CD34+ cells obtained from bone marrow. By using Hoechst 33342-dye efflux assay, which identifies a population of immature HPC, termed side population (SP) cells we have examined the phenotypical profile of SP+CD34+ cells obtained from bone marrow and SP+CD34+ cells isolated from PBPC. Highly enriched CD34+ cells were isolated from PBPC obtained from patients with Hodgkin lymphoma, and bone marrow was obtained from healthy volunteer donors by iliac crest aspiration after informed consent. To identify the SP+ cells, enriched CD34+ cells were stained with Hoechst 33342 dye. Using flowcytometric techniques (FACStar+, FACSDiva, Becton Dickinson, San Jose, CA) we were able to visualize the dye efflux in SP+ cells. SP+ cells were functionally confirmed using Verapamil staining. The frequenzy of LTC-IC was markedly increased in SP+CD34+ cells compared to SP−CD34+ cells (n=5), in line with previous reports. The percentage of SP+CD34+ cells varied from 0,4 to 18% of the total CD34+ cell population obtained from PBPC (n= 16), whereas the level of SP+CD34+ cells obtained from bone marrow varied between 4–7% of the total CD34+ cell population (n=4). Expression of lineage committed markers, including CD10, CD15 and CD19 was less then 10% of the whole CD34+ cell population obtained from PBPC, whereas we found a higher level of expression of these markers in CD34+ cells isolated from bone marrow. However, when we examined the SP+CD34+ cells from either PBPC or bone marrow, we observed that the phenotypical profile of these cells were similar with almost no expression of lineage markers. Thus, the more lineage-committed cells in the CD34+ cell population obtained from bone marrow seems to be restricted to the SP−CD34+ cell fraction. Examination of CD90 and CD133 expression revealed a higher level in the SP+ CD34+ cell fractions compared to the SP− fractions. Furthermore, we investigated the level of CD38 expression. Previous studies have demonstrated that lack of CD38 expression in CD34+ cells identifies a more immature cell population. Surprisingly, we observed that 30–40% of SP+CD34+ cells obtained from bone marrow were CD38 negative, whereas the level of SP+CD34+CD38− cells from PBPC was 2–5%, which is similar to the level of CD38− cells in the CD34+ cell population isolated from both PBPC and bone marrow. Currently, we are exploring the frequency of LTC-IC in SP+CD34+CD38− cells from bone marrow, and we are also planning cell sorting of these cells for functional analyses. In conclusion, we find that the level of CD38 negative cells in SP+CD34+ subpopulation of CD34+ bone marrow cells are higher than what observed in SP+CD34+ and SP−CD34+ from PBPC as well as in SP−CD34+ from bone marrow. Our ongoing studies will clarify if these results define SP+CD34+CD38− cells from bone marrow as a source of highly enriched primitive HPC.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4170-4170
Author(s):  
Dag Josefsen ◽  
Lise Forfang ◽  
Marianne Dyrhaug ◽  
Gunnar Kvalheim

Abstract Side population (SP) cells are characterised by their ability to exclude Hoechst 33342 dye from the cells. Using this method, it has been demonstrated that cells within the SP+ fraction of mononuclear cells from both murine and human hematopoietic systems are enriched for primitive hematopoietic stem- and progenitor cells. Moreover, most of the SP+ cells did not express CD34, indicating the presence of a CD34 negative hematopoietic stem cell population. To explore this further, we have examined SP+ cells obtained from different cell compartments in human bone marrow and peripheral blood. Human bone marrow (BM) was obtained from healthy volunteer donors by iliac crest aspiration after informed consent. Mononuclear cells (MNC) were obtained by Ficoll grade centrifugation. CD34+ cells were then isolated from MNC. Highly enriched CD34+ cells were isolated from PBPC obtained from patients with Hodgkin lymphoma. To identify the SP+ cells, the cells were stained with Hoechst 33342 dye. Using flowcytometric techniques (FACStar+, FACSDiva, Becton Dickinson, San Jose, CA) we were able to visualize the dye efflux in SP+ cells. SP+ cells were functionally confirmed using Verapamil. Phenotypical characterisation of the different cell populations using flow cytometric methods was performed. The level of SP+ cells in BM-MNC was 1,3% (mean, n=3) In line with previous findings, we observed that SP+ cells obtained from BM-MNC lack expression of several lineage committed markers, including CD15 and CD19. Most of the cells were CD34− (mean=2,2%), which was lower than in the main population (MP; mean=5%). The level of CD133 expression was low and similar in both populations. Furthermore we found a higher fraction of CD3+ T-cells in the SP fraction than in the MP fraction (mean: 69% vs 51%). To further investigate the SP+CD34+ cell fraction, we examined CD34+ cells isolated from both human bone marrow and peripheral blood. The percentage of SP+CD34+ cells varied from 0,4 up to 18% of the total CD34+ cell population obtained from PBPC (n= 16), whereas the level of SP+CD34+ cells obtained from bone marrow was 5% of the total CD34+ cell population (n=3). Expression of lineage committed markers, including CD10, CD15 and CD19 was less then 10% of the whole CD34+ cell population obtained from PBPC, whereas we found a higher level of expression of these markers in CD34+ cells isolated from bone marrow. However, when we examined the SP+CD34+ cells from either PBPC or bone marrow, we observed that the phenotypic profile of these cells were similar with almost no expression of lineage markers. The frequency of LTC-IC was markedly increased in SP+MNC, in line with previous findings. In addition we also observed a marked increase in LTC-IC in SP+CD34+ cells compared to SP-CD34+ cells in both BM and PB (BM: 7-fold increase; PB: 3–4 fold). In conclusion, SP cells are present in different hematopoietic progenitor cell populations, including BM-MNC, BM-CD34+ cells and PB-CD34+ cells. In SP+CD34+ cell fractions from both BM and PB we observed an increased expression of stem cell markers like CD90 and CD133, whereas in SP+MNC we found low levels of CD34, CD90 and CD133 expression. However, the LTC-IC frequency was markedly higher in all SP+fractions compared to MP fractions, suggesting that sorting of SP+ cells from different hematopoietic stem- and progenitor cell compartments identify immature hematopoietic cells.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1395-1395
Author(s):  
Feng Xu ◽  
Qingde Wang ◽  
Hongmei Shen ◽  
Hui Yu ◽  
Yanxin Li ◽  
...  

Abstract Adenosine Deaminases Acting on RNA (ADAR) are RNA-editing enzymes converting adenosine residues into inosine (A-to-I) in many double-stranded RNA substrates including coding and non-coding sequences as well as microRNAs. Disruption of the ADAR1 gene in mice results in fetal liver, but not yolk sac, defective erythropoiesis and death at E11.5 (Wang Q et al, Science 2000). Subsequently, a conditional knockout mouse model confirmed these findings and showed massively increased cell death in the affected organs (Wang Q et al, JBC 2004). However, the actual impact of ADAR1 absence on definitive or adult hematopoiesis has not been examined. To define the role of ADAR1 in adult hematopoiesis, we first examined the expression of ADAR1 in different hematopoietic stem/progenitor cell subsets isolated from bone marrow by real-time RT-PCR. ARAR1 was present in hematopoietic stem cells (HSCs) at relatively low level and increased in hematopoietic progenitor cells (HPCs). A series of functional hematopoietic assays were then undertaken. A conditional deletion of ADAR1 was achieved by transducing Lin− or Lin−cKit+ bone marrow cells from ADAR1-lox/lox mice with a MSCV retroviral vector co-expressing Cre and GFP. PCR analysis confirmed the complete deletion of ADAR1 in the transduced cells within 72 hours after the transduction. This system allowed us to evaluate the acute effect of ADAR1 deletion in a specific hematopoietic cell population. Following 4 days of in vitro culture after transduction, the absolute number of Lin− Sca1+ cells in the Cre transduced group was similar to the input number; however the differentiating Lin+ cells significantly decreased whereas both the Lin−Sca1+ and Lin+ cells in the vector (MSCV carrying GFP alone) transduced group increased during culture. Moreover, the colony forming cell (CFC) assay showed much fewer and smaller colonies that contained dead cells from the gene deleted group as compared to those from the control group (p&lt;0.001). The TUNEL assay showed a dramatic increase of apoptosis in the Lin+ population but not in the Lin− cells. Given the mixed genetic background of the ADAR1-lox/lox mice, repopulation of the transduced hematopoietic cells in vivo was examined in immunodeficient mice. Sublethally irradiated (3.5 Gy) NOD/SCID-γcnull recipient were transplanted with either 1.5 × 105 Cre or vector transduced Lin− ADAR1-lox/lox cells. Multi-lineage engraftment in peripheral blood was monitored monthly. While the vector transduced cells were able to constitute more than 90% in multiple lineages of the peripheral blood at 1 to 3 months, Cre-transduced cells were virtually undetectable at all the time points (n=9 to 13, p&lt;0.001). A similar result was found in the hematopoietic organs, including the bone marrow, spleen and thymus. Interestingly, however, the Lin−Sca1+cKit+ cell population was preserved in the Cre transduced group despite the very low level of total donor-derived cells in the bone marrow (n=6 to 7, p&lt;0.01). Consistently, the single cell culture experiment demonstrated that there was no significant difference between ADAR−/− and wild-type HSCs in terms of survival and division during the first 3 days of culture. Taken together, our current study demonstrates nearly absolute requirement of ADAR1 for hematopoietic repopulation in adult mice and it is also suggested that ADAR1 has a preferential effect on the survival of differentiating progenitor cells over more primitive cells.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5465-5465
Author(s):  
Xiaomiao Li ◽  
Zhongbo Hu ◽  
Marda Jorgenson ◽  
William Slayton

Abstract Background: The bone marrow contains a variety of blood vessels that have different functions in maintaining the bone marrow as the major blood producing organ in adulthood. For instance, arterioles function to control the flow of blood into bone marrow compartments, and the sinusoids serve as a conduit to the blood stream and niches for megakaryocyte development. Most current studies of the bone marrow vasculature, including studies quantifying changes in the marrow vascular by microvascular density, do not differentiate between different types of marrow vessels. Recognizing the changes in different types of blood vessels has important physiologic implications. Here we report a new method to distinguish sinusoids from arterioles in the murine bone marrow. Methods and Results: We used transgenic mice with GFP expressed downstream of the Tie-2 promoter, combined with in vivo acetylated low-density lipoprotein (Ac-LDL) uptake method to differentiate sinusoids from arterioles. We found that Ac-LDL was specifically endocytosed by sinusoids, and Tie-2 expression was more pronounced in the arteries, arterioles, and transitional capillaries. Combining these two functional endothelial markers and using confocal microscopy to obtain three dimensional images, we identified transitional zones where arterioles emptied into the sinusoids. Conclusions: These results demonstrate that the marrow vasculature and specific endothelial cell types are functionally heterogeneous. Methods to study changes in the marrow vasculature and particularly the vascular niche, a function of sinusoids, need to take into account this heterogeneity.


Sign in / Sign up

Export Citation Format

Share Document