scholarly journals Single-Cell Analysis of Human B Lymphoid and Neutrophil/Monocyte Lineage Restriction

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 4291-4291
Author(s):  
Fangwu Wang ◽  
Colin Hammond ◽  
Connie J. Eaves

Abstract Lifelong production of most types of mature blood cells is sustained by a small population of cells with extensive regenerative potential. However, the detailed steps that restrict multipotent or even bipotent human hematopoietic cells to any single lineage remain poorly understood. Those that segregate the human B-lymphoid and neutrophil/monocyte (NM) lineages are of particular interest as these appear to identify a stage that might control the different properties of human leukemias that display perturbed NM and/or B-cell programs. To undertake a refined analysis of this normal lineage restriction process in human cells, we first devised a culture system that permits it to be tracked clonally and efficiently (50%). Initial experiments showed this could be achieved using a combination of multiple stromal cell types and human growth factor-supplemented medium. To elucidate the intervening transitional steps we then used multiplexed flow cytometry to compare the progeny generated in this culture system over a 2-week time course from previously defined lymphoid progenitor-enriched (P-L), NM progenitor-enriched (P-NM) and less restricted P-mix cord blood (CB) subsets. The results suggested that gain of CD45RA (RA) expression and loss of CLEC12A (C) expression appeared to accompany the sequential restriction of early CD34+ progenitors first to cells with dual NM+B-lineage potential and then just to B-lineage potential. Subsequent tracking of the lineage outputs of CD34+ RA-C- cells initially produced in larger numbers from unfractionated CD34+ CB cells either in vitro or in xenografted immunodeficient mice, confirmed the CD34+ RA-C- subset to be highly enriched in cells with dual NM+B potential. In contrast, co-generated CD45RA+ (RA+C-) and RA+CLEC12A+ (RA+C+) phenotypes displayed separate B- and NM lineage-restricted activity, respectively, as indicated by their largely exclusive clonal outputs of CD19+ pre-B and CD14+/CD15+ NM precursors. In agreement with these phenotypically established separate NM and B-lineage outputs, RA+C- cells were found to contain higher levels of B-lineage-associated gene transcripts (e.g., DNTT, CD79A, and EBF1), whereas RA+C+ cells contained higher levels of the NM transcription factor mRNAs encoded by SPI1 and CEBPA. In a further optimized stroma-free liquid culture system, the RA-C- cells could be shown to produce continuously RA+C- and RA+C+ progeny after another 3-4 days, and also RA-C- progeny which are not produced from more restricted RA+C- and RA+C+ cells, suggesting that the acquired expression of RA precedes the separation of NM and B-lineage potential that is then marked by the differential activation of C expression in RA+ cells. To examine more precisely how the process of B+NM restriction to a single lineage might be related to successive cell cycles, we labeled RA-C- cells with carboxyfluorescein diacetate succinimidyl ester (CFSE) to enable the phenotypes and growth potential of the the progeny obtained after 6 days to be directly related to their prior division histories. This revealed extensive heterogeneity in the overall distribution of the initial progeny cell cycle times but with a clear segregation in the outputs of the faster and slower dividing cells. Notably, the faster dividers produced ultimately small M+B or uni-lineage clones whereas the initially slow dividers subsequently produced larger clones, 25% of which still contained CD34+ cells or N+M+B progeny. Taken together, these findings identify new hallmark phenotypic changes that identify a critical step in the lineage restriction of primitive human hematopoietic cells with dual NM+B-lineage potential and a previously unknown association of this process with a shortening of their cell cycle transit time. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2579-2579
Author(s):  
Yan Sun ◽  
Zack Zhengyu Wang ◽  
Wen-Shu Wu

Abstract Abstract 2579 Slug belongs to the highly conserved Slug/Snail family of zinc-finger transcription factors found in diverse species ranging from C. elegans to humans. Our previous study has already demonstrated that Slug serves intrinsic role in the regulation of HSC fates (Blood, 2010, 115(9):1709-1717). There is bone defect in Slug-/- mice, while Slug is expressed in osteoblast cells which are niche cells for HSCs, suggesting that Slug might be also extrinsically required for HSC regulation. To address this issue, the following experiments and assays were performed. Firstly, the cell proliferation of wild-type (WT) hematopoietic stem/progenitor cells (HS/PCs) (loaded with carboxyfluorescein succinimidyl ester (CFSE)) placed on bone marrow (BM) stromal layers (prepared from either Slug-/- or WT mice) was analyzed. Co-culture proliferation assay illustrated a clear increasing in the ability of Slug-/-BM stroma to support normal proliferation of WT HS/PCs, suggesting a significant and qualitative change in Slug-/- BM stromal cells so that they were able to support normal proliferation by Slug-competent HS/PCs. Secondly, BM microenvironment altered by Slug deficiency was defined by RT-PCR. Slug-/- BM had altered cell cycle profiles associated with increased stromal Notch1, N-cadherin, Vcam and Angiopoietin-1 expression. These findings indicate that Slug may participate in signaling in BM niche cells and thus is capable of influencing their function. Thirdly, the transplantation analysis was performed to address whether Slug impact was stem cell stroma dependent. WT hematopoietic cells in Slug knockout environment (8 weeks after transplantation) displayed a dramatic increase in LSK and LSK-Flk-2 ratio and in total number in BM, but not in spleen, indicating a stroma-determined effect by Slug on HSCs. It also showed a mobilization phenotype similar to that originally observed in Slug+/+ environment. Moreover, the annexin V apoptosis assay and cell-cycle analysis by the Edu incorporation assay suggested that Slug deficiency microenvironment promoted HSC expansion largely by increasing their proliferation, but not cell survival. Fourthly, each cell lineage in BM and spleen after WT BM transplanted into Slug+/+ and Slug-/- recipients was fully analyzed. The total number and ratio of T cells (CD3e, CD4+CD8-, CD4-CD8+ and CD4+CD8+) was markedly decreased in BM, but not in spleen, while the other lineages (granulocytes CD11b+Gr-1+, macrophages CD11b+, immature B lymphoid IgM-B220+, mature B lymphoid IgM+B220+, mature Erythroid CD71-Ter119+, and immature erythroid CD71+Ter119+) had an equivalent number and ratio in BM and spleen. Overall, Slug-/- BM microenvironment results in T cell loss. Finally, the recent study shows BM adipocytes as negative regulators of haematopoietic microenvironment, while the in vivo and in vitro evidences indicate that Slug is a key regulator of the adipocyte differentiation. Thus, in this study, we try to address whether Slug as a key factor adjusts adipocytes in BM niche. The fat of the whole body and the femurs and tibias were measured by DXA (Dual-energy X-ray Absorptiometry). Our data showed Slug-/- mice at 8 wk of age had lowest total fat, as well as ROI (region of interest) fat compared to Slug+/+ mice. In vitro adipocyte differentiation assay indicated there was less adipocyte formation from Slug-/- MSCs from BM. Furthermore, a dramatic loss of adipocytes in the femur in Slug-/- recipient was observed in 8 weeks after transplanting WT hematopoietic cells into Slug+/+ and Slug-/- recipients. Combining the above-mentioned data from WT hemtopoisis in Slug-/- microenvironment showed a dramatic increase in LSK and LSK-Flk-2 ratio and total numbers, lack of BM adipocytes after irradiation in fatless mice (Slug-/- recipient), suggesting a role of Slug-/- BM microenvironment in enhancing haematopoietic progenitor expansion via inhibition of BM adipocytes and post-transplant recovery. Taken together, our previously and present findings demonstrate that Slug serves intrinsic and extrinsic roles in the regulation of HSC fates. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2504-2504
Author(s):  
Russell Garrett ◽  
Gerd Bungartz ◽  
Alevtina Domashenko ◽  
Stephen G. Emerson

Abstract Abstract 2504 Poster Board II-481 Polyinosinic:polycytidlyic acid (poly I:C) is a synthetic double-stranded RNA used to mimic viral infections in order to study immune responses and to activate gene deletion in lox-p systems employing a Cre gene responsive to an Mx-1 promoter. Recent observations made by us and others have suggested hematopoietic stem cells, responding to either poly I:C administration or interferon directly, enter cell cycle. Twenty-two hours following a single 100mg intraperitoneal injection of poly I:C into 10-12 week old male C57Bl/6 mice, the mice were injected with a single pulse of BrdU. Two hours later, bone marrow was harvested from legs and stained for Lineage, Sca-1, ckit, CD48, IL7R, and BrdU. In two independent experiments, each with n = 4, 41 and 33% of Lin- Sca-1+ cKit+ (LSK) IL-7R- CD48- cells from poly I:C-treated mice had incorporated BrdU, compared to 7 and 10% in cells from PBS-treated mice. These data support recently published reports. Total bone marrow cellularity was reduced to 45 and 57% in the two experiments, indicating either a rapid death and/or mobilization of marrow cells. Despite this dramatic loss of hematopoietic cells from the bone marrow of poly I:C treated mice, the number of IL-7R- CD48- LSK cells increased 145 and 308% in the two independent experiments. Importantly, the level of Sca-1 expression increased dramatically in the bone marrow of poly I:C-treated mice. Both the percent of Sca-1+ cells and the expression level of Sca-1 on a per cell basis increased after twenty-four hours of poly I:C, with some cells acquiring levels of Sca-1 that are missing from control bone marrow. These data were duplicated in vitro. When total marrow cells were cultured overnight in media containing either PBS or 25mg/mL poly I:C, percent of Sca-1+ cells increased from 23.6 to 43.7%. Within the Sca-1+ fraction of poly I:C-treated cultures, 16.7% had acquired very high levels of Sca-1, compared to only 1.75% in control cultures. Quantitative RT-PCR was employed to measure a greater than 2-fold increase in the amount of Sca-1 mRNA in poly I:C-treated cultures. Whereas the numbers of LSK cells increased in vivo, CD150+/− CD48- IL-7R- Lin- Sca-1- cKit+ myeloid progenitors almost completely disappeared following poly I:C treatment, dropping to 18.59% of control marrow, a reduction that is disproportionately large compared to the overall loss of hematopoietic cells in the marrow. These cells are normally proliferative, with 77.1 and 70.53% accumulating BrdU during the 2-hour pulse in PBS and poly I:C-treated mice, respectively. Interestingly, when Sca-1 is excluded from the analysis, the percent of Lin- IL7R- CD48- cKit+ cells incorporating BrdU decreases following poly I:C treatment, in keeping with interferon's published role as a cell cycle repressor. One possible interpretation of these data is that the increased proliferation of LSK cells noted by us and others is actually the result of Sca-1 acquisition by normally proliferating Sca-1- myeloid progenitors. This new hypothesis is currently being investigated. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2397-2397
Author(s):  
Hirohito Minami ◽  
Kohshi Ohishi ◽  
Masahiro Masuya ◽  
Naoyuki Katayama

Abstract The regulatory mechanism of human early T-lymphoid differentiation remains less defined. We previously reported that human telomerized bone marrow stromal cells support the generation of CD7+ CD56- early T- as well as CD10+ CD19+ early B-lymphoid precursors from human hematopoietic precursors. Here we examined whether and how early T lymphopoiesis is regulated by interaction with stromal cells. Low or no levels of LFA-1 were expressed on CD34+ CD38- CD45RA- immature hematopoietic precursors. However, high levels of LFA-1 were detected on CD34+ CD38- CD45RA+ CD10+ CD7+/- CD19- immature lymphoid precursors, while those of LFA-1 were diminished on CD34+ CD38+ CD45RA+ CD10+ CD19+ more mature pro B cells. On the other hand, ICAM-1 and ICAM-2 were expressed in a portion of the telomerized stromal cells. ICAM-3 was not detected. Various levels of ICAM-1, ICAM-2, or ICAM-3 were also expressed on hematopoietic and lymphoid precursors. To examine the role of LFA-1-mediated adhesion to stromal cells or adjacent hematopoietic precccursors in early lymphoid differentiation, we examined the effect of anti-LFA-1 blocking antibody (Ab) on the differentiation of CD34+ CD45RA- CD7- CD10- CD38lo/- hematopoietic precursors in the cultures on stromal cells or with conditioned medium (CM) obtained from cultures of stromal cells. In the cultures on stromal cells, anti-LFA-1 Ab strongly inhibited the generation of CD7+ CD10- CD45RA+ and CD7- CD10+ CD45RA+ lymphoid precursors from hematopoietic precursors after 21 days of culture. Significant number of CD14+ monocytic cells was generated with or without anti-LFA-1 Ab. In the cultures with CM, anti-LFA-1 Ab showed marginable effect on lymphoid differentiation. To elucidate the effect of anti-LFA-1 Ab on more mature lymphoid precursors, CD7+ CD10- CD45RA+ or CD7- CD10+ CD45RA+ cells were isolated after culture of hematopoietic cells for 14 days and cultured on stromal cells in the presence or absence of anti-LFA-1 Ab. Anti-LFA-1 Ab remarkably inhibited the generation of CD7+ and CD10+ CD19+ lymphoid cells from the CD7+ CD10- CD45RA+ early T-lymphoid precursors. Notably, few or no CD45RA+ CD14- lymphoid cells were detected. Anti-LFA-1 Ab did not affect B-lineage differentiation from CD10+ CD19- CD45RA+ early B-lymphoid precursors to CD10+ CD19+ CD45RA+ proB cells. We next examined which ICAM ligand is responsible for the observed effects by anti-LFA-1 Ab. Anti-ICAM-2 Ab inhibited the generation of CD7+ CD45RA+ and CD10+ CD45RA+ lymphoid precursors from CD34+ CD45RA- CD7- CD10- CD38lo/- hematopoietic precursors on stromal cells, as observed with anti-LFA-1 Ab. No effect was observed with anti-ICAM-1 Ab. Anti-ICAM-2 Ab further suppressed the generation of CD7+ and CD10+ lymphoid cells from the cultured CD7+ CD10- CD45RA+ early T-lymphoid precursors, but did not depress B-lymphoid differentiation from the cultured CD7- CD10+ CD45RA+ early B-lymphoid precursors. Taken together, these data indicate that LFA-1-mediated adhesion to ICAM-2 is essential for stromal cell-dependent early lymphoid differentiation of hematopoietic and CD7+ early T-lymphoid precursors. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 380-380
Author(s):  
Sonia Rodriguez ◽  
Christen Mumaw ◽  
Cristina LoCelso ◽  
Leonor Sarmento ◽  
Keiichi Nakayama ◽  
...  

Abstract Abstract 380 S-phase kinase-associated protein 2 (SKP2) is the F-box subunit of the ubiquitin-ligase complex SCFSKP2 that targets the cyclin-dependent kinases (CDKs) p27Kip1, p21Cip1 and p130 for ubiquitination and degradation. SKP2 protein levels are modulated during cell cycle and in response to mitogenic stimuli: its expression is essential for cell cycle entry whereas its downregulation is critical for cell cycle arrest. The fact that SKP2 controls the destruction of three CKIs underscores its central role as a cell cycle regulator. We have previously shown that Notch1 activation induces the transcriptional activation of SKP2, promoting downregulation of the CKIs and inducing accelerated cell cycle progression in hematopoietic cells. Thus, the Notch/SKP2/CKIs pathway links directly environmental cues to the cell cycle machinery and may play an important role in controlling stem cell pool size and expansion of hematopoietic progenitor cells, in steady-state and during stress conditions. However, it is unclear how SKP2 regulates these CKIs in distinct hematopoietic subpopulations and its function in normal hematopoiesis has not been investigated. Analysis of SKP2 expression on murine bone marrow (BM) hematopoietic cells showed high expression levels in the myeloid progenitors, erythroid cells, B-and T-cells; lower levels were found in Lin- cells, whereas LSK cells showed a little expression of SKP2, in agreement with their more quiescent status. To better define the role of SKP2 during hematopoietic differentiation we analyzed the effects of SKP2 deficiency in the BM compartment of SKP2 null mice at steady-state conditions. As anticipated, in SKP2 knockout mice loss of SKP2 expression correlated with p27Kip1 accumulation and overall reduced cell cycle, both in vivo and in vitro. Loss and decreased levels of SKP2 in SKP2 -/- and SKP2+/- mice correlated with a significant increase in stem cells (LSK, averages: 3.2% in WT vs 6.2% KO; p=0.007), suggesting that low rates of proliferation favor quiescence and decrease the egress of cells from the HSC pool to the more mature compartments. Further immunophenotypical analysis of the BM subsets revealed that SKP2 loss resulted in the specific decrease of granulocyte-monocyte (GMP) and megacaryocytic-erythroid (MEP; averages 16.5% vs 7.6%; p=0.045) progenitor pools. Interestingly, the decrease in GMPs was associated to increased (rather than decreased) percentages of maturing myeloid progenitors Gr1+Mac1+ (averages: 26% vs 33%; p=0.049). An increase in rate of myeloid differentiation in the absence of SKP2 was further confirmed by in vitro differentiation of SKP2-/- progenitors and by overexpression of a SKP2 dominant negative into the PLB myeloid cell line. In conclusion, this data suggests that SKP2 levels may play a distinct role in regulating quiescence in the pool of HSC and rate of proliferation and differentiation in myeloid progenitors. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 8-8
Author(s):  
Alessandra Suelen Jardim Silva ◽  
Gustavo Henrique de Medeiros Oliveira ◽  
Lenilton Silva DA Silva Júnior ◽  
Hugo Henrique de Freitas Ferreira ◽  
Maria das Graças Pereira Araujo ◽  
...  

The acute lymphoblastic leukemia (ALL) is a malignant disease of the immune system and hematologic characterized by accumulation of neoplastic B lymphoid precursors or T (lymphoblasts) in the bone marrow and / or peripheral blood. The diagnosis of these leukemias occurs by morphological classification of French-American-British (L1, L2 or L3) associated with features of immunological profile T or B cell malignancies, based on the expression profile of monoclonal antibodies (MoAb) directed against the antigens of cell differentiation by flow cytometry (FC). Several studies have shown that blast cell immunophenotypes of cases of acute lymphoblastic leukemia does not always exhibit characteristics of lymphoid differentiation normal but exhibit aberrant immunophenotypes. Thus, blasts some cases of acute lymphoblastic leukemia of B lineage may show myeloid or T antigens. Also blasts of cases of acute lymphoblastic leukemia T cell determinants may possess B or myeloid cells.Objective:To determine the immunophenotypic profile by FC in 88 patients with ALL (B or T lineage) diagnosed in the Laboratory of Flow Cytometry Blood Center of Dalton Cunha - HEMONORTE, from State of Rio Grande do Norte, Brazil.Methods:All samples from peripheral blood and / or bone marrow were subjected to FC immunophenotyping using a panel of MoAb specific for diagnosis of acute leukemia (AL) directly conjugated to fluorochromes as fluorescein isothiocyanate (FITC), phycoerythrin (PE), peridinin-chlorophyll protein (PerCP) and allophycocyanin (APC).Results:The patients' age range was between 1 month and 84 years old, with an average of 20.3 years, with 62.5% of the patients being male. The most frequently observed strain was B and the most evident subtype was the Common Pre-B ALL. Among the cell markers evaluated, the most expressed in lineage B were CD19, CD10, HLADR and cCD79a and the antigens most frequently expressed in lineage T were cytoplasmic CD3 (cCD3) and membrane (mCD3), CD7, CD5 and CD2. A small percentage (6.8%) were doubly positive T cells.Conclusion:It is concluded that individuals with ALL in this study have demographic, clinical and immunophenotypic characteristics similar to those observed in other studies, demonstrating that CF immunophenotyping is an essential methodology in the diagnosis of follow-up of these leukemias. Disclosures No relevant conflicts of interest to declare.


2011 ◽  
Vol 236 (11) ◽  
pp. 1342-1350 ◽  
Author(s):  
Yukio Hirabayashi ◽  
Yoshihiro Hatta ◽  
Jin Takeuchi ◽  
Isao Tsuboi ◽  
Tomonori Harada ◽  
...  

Hematopoiesis occurs in the bone marrow, where primitive hematopoietic cells proliferate and differentiate in close association with a three-dimensional (3D) hematopoietic microenvironment composed of stromal cells. We examined the hematopoietic supportive ability of stromal cells in a 3D culture system using polymer particles with grafted epoxy polymer chains. Umbilical cord blood-derived CD34+ cells were co-cultivated with MS-5 stromal cells. They formed a 3D structure in the culture dish in the presence of particles, and the total numbers of cells and the numbers of hematopoietic progenitor cells, including colony-forming unit (CFU)-Mix, CFU-granulocyte-macrophage, CFU-megakaryocyte and burst-forming unit-erythroid, were measured every seven days. The hematopoietic supportive activity of the 3D culture containing polymer particles and stromal cells was superior to that of 2D culture, and allowed the expansion and maintenance of hematopoietic progenitor cells for more than 12 weeks. Various types of hematopoietic cells, including granulocytes, macrophages and megakaryocytes at different maturation stages, appeared in the 3D culture, suggesting that the CD34+ cells were able to differentiate into a range of blood cell types. Morphological examination showed that MS-5 stromal cells grew on the surface of the particles and bridged the gaps between them to form a 3D structure. Hematopoietic cells slipped into the 3D layer and proliferated within it, relying on the presence of the MS-5 cells. These results suggest that this 3D culture system using polymer particles reproduced the hematopoietic phenomenon in vitro, and might thus provide a new tool for investigating hematopoietic stem cell–stromal cell interactions.


Blood ◽  
1983 ◽  
Vol 61 (6) ◽  
pp. 1045-1053 ◽  
Author(s):  
BB Lozzio ◽  
EA Machado ◽  
J Mitchell ◽  
CB Lozzio ◽  
CJ Wust ◽  
...  

Abstract Six human hematopoetic cell lines were successfully heterotransplanted into athymic (nude) and asplenic-athymic (lasat) neonatal mice. The tumors arising from leukemia and lymphoma cells could then be serially transplanted into adult nude mice. Seven days after the fourth serial mouse passage, each mouse was treated with goat immune gamma globulin against K-562 cells. One control group was treated similarly, but with nonimmune (normal) gamma globulin, while another control group was not treated. The goat gamma globulin was not toxic for nude and lasat mice, and the immune, but not the normal, gamma globulin suppressed local subcutaneous growth of myelosarcomas, lymphosarcomas, and Burkitt lymphoma cells. On the other hand, the growth of lung, breast, and prostatic carcinomas and a melanoma of human origin were not altered by the immune gamma globulin. Since suppression of cell growth occurred equally well in decomplemented mice, a complement-mediated cytotoxicity apparently cannot be considered as responsible for the abrogation. The Fab fragment of the immunoglobulin did not suppress the growth of the myelosarcomas. We conclude that antibody suppression of the in vivo proliferation was specific for malignant hematopoietic cells and that the Fc portion of IgG is necessary for in vivo cytolysis of leukemia cells. The most probable mechanisms are direct antibody cytolysis and antibody-dependent macrophage-mediated cytotoxicity.


Blood ◽  
2002 ◽  
Vol 99 (5) ◽  
pp. 1585-1593 ◽  
Author(s):  
Anna Jetmore ◽  
P. Artur Plett ◽  
Xia Tong ◽  
Frances M. Wolber ◽  
Robert Breese ◽  
...  

Differences in engraftment potential of hematopoietic stem cells (HSCs) in distinct phases of cell cycle may result from the inability of cycling cells to home to the bone marrow (BM) and may be influenced by the rate of entry of BM-homed HSCs into cell cycle. Alternatively, preferential apoptosis of cycling cells may contribute to their low engraftment potential. This study examined homing, cell cycle progression, and survival of human hematopoietic cells transplanted into nonobese diabetic severe combined immunodeficient (NOD/SCID) recipients. At 40 hours after transplantation (AT), only 1% of CD34+ cells, or their G0(G0CD34+) or G1(G1CD34+) subfractions, was detected in the BM of recipient mice, suggesting that homing of engrafting cells to the BM was not specific. BM of NOD/SCID mice receiving grafts containing approximately 50% CD34+ cells harbored similar numbers of CD34+ and CD34− cells, indicating that CD34+ cells did not preferentially traffic to the BM. Although more than 64% of human hematopoietic cells cycled in culture at 40 hours, more than 92% of cells recovered from NOD/SCID marrow were quiescent. Interestingly, more apoptotic human cells were detected at 40 hours AT in the BM of mice that received xenografts of expanded cells in S/G2+M than in recipients of G0/G1 cells (34.6% ± 5.9% and 17.1% ± 6.3%, respectively; P < .01). These results suggest that active proliferation inhibition in the BM of irradiated recipients maintains mitotic quiescence of transplanted HSCs early AT and may trigger apoptosis of cycling cells. These data also illustrate that trafficking of transplanted cells to the BM is not selective, but lodgment of BM-homed cells may be specific.


2005 ◽  
Vol 73 (11) ◽  
pp. 7718-7726 ◽  
Author(s):  
Annemarie M. C. van Rossum ◽  
Elena S. Lysenko ◽  
Jeffrey N. Weiser

ABSTRACT Nasopharyngeal colonization is the first step in the interaction between Streptococcus pneumoniae (the pneumococcus) and its human host. Factors that contribute to clearance of colonization are likely to affect the spread of the pneumococcus and the rate of pneumococcal disease in the population. To identify host and bacterial factors contributing to this process, we examined the time course of colonization using genetically modified mice and pneumococci. Severe combined immunodeficient mice remained persistently colonized (>6 weeks). Major histocompatibility complex II-deficient mice, but not μMT mice, were unable to clear colonization and showed a diminished T helper 1 response. Thus, CD4+ T cells, rather than the generation of specific antibody, appear to be required for effective Th1-mediated clearance. In addition, the microbial pattern recognition receptor toll-like receptor 2 (TLR2), but not TLR4, was necessary for efficient clearance of colonization. In contrast, no role of complement component 3, inducible nitric oxide synthetase, interleukin 12 (IL-12), or IL-4 could be demonstrated. Expression of the pneumococcal toxin pneumolysin enhanced acute localized inflammatory responses and promoted clearance of colonization in a TLR4-independent manner. We conclude that both innate and CD4+ T-cell-mediated immunity and proinflammatory bacterial factors, rather than a humoral adaptive immune response, are important for clearance of S. pneumoniae from the murine nasopharynx.


Sign in / Sign up

Export Citation Format

Share Document