Educating the Innate Immune System

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. SCI-28-SCI-28
Author(s):  
Scott Worthen ◽  
Junjie Mei ◽  
Yuhong Liu ◽  
Hitesh Deshmukh

Abstract The microbiome regulates the development of innate immunity G. Scott Worthen, Junjie Mei, Yuhong Liu, Ning Dai, Hitesh Deshmukh Circulating granulocytes are maintained within a fairly narrow window for each individual during homeostasis. This regulatory system is nonetheless capable of dramatic shifts during stress. The mechanisms that govern control over granulocyte number remain incompletely understood. Recent information, however, provides clues to the feedback control systems that regulate homeostatic and emergency granulopoiesis. The first clues have come from adult mice with abnormalities in granulocyte function. Defects in Leukocyte Integrins or chemokine receptors result in marked increases in circulating (?2 integrin) or bone marrow (Cxcr2) neutrophils, associated with increased circulating IL-17 and G-CSF. This result appears to be due to a functional inability to arrive at sentinel site(s) whence they are attracted by (among others) Cxcl5. Strikingly, a single infusion of normal WT neutrophils resets this feedback loop. One signal for expression of IL-17 and G-CSF appears to be the gut microbiome. Antibiotic treatment also resets the system, reducing neutrophil numbers and cytokine expression. Thus, at sentinel site(s), the supply of functional neutrophils is balanced against perceived threat from the microbiome. In neonates, exposure to commensal organisms is an immediate result of birth from a sterile environment into a dirty one. We have described postnatal granulopoiesis in murine neonates, a dramatic increase in circulating and bone marrow neutrophils, that lasts for 5-7 days. Human infants similarly have been shown to demonstrate a postnatal increase in circulating neutrophils for 72 hrs after birth. Antibiotic-exposed mouse pups fail to develop postnatal granulopoiesis, as do mice deficient in IL17ra, MyD88, TLR4, or G-CSF even if not exposed to antibiotics, indicating a pathway that requires exposure to LPS that induces IL17 and G-CSF, as in adults. In contrast to adults, however, commensal bacteria are required for host defense. Antibiotic-exposed pups are exquisitely sensitive to E. coli sepsis, which can be partially reversed by transfer of cecal contents or exogenous G-CSF. Furthermore, small quantities of LPS, fed by gavage, can also partially protect. Thus, neonates use exposure to commensal bacteria and their products to trigger the rapid expansion and functional maturation of granulocyte development, and in so doing, prepare the neonate for potential exposure to pathogens. Adults maintain this system, where it is at least partially responsible for maintenance of granulocyte homeostasis. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1988-1988
Author(s):  
Jadwiga Gasiorek ◽  
Gregory Chevillard ◽  
Zaynab Nouhi ◽  
Volker Blank

Abstract Abstract 1988 Poster Board I-1010 The NF-E2 transcription factor is a heterodimer composed of a large hematopoietic-specific subunit called p45 and widely expressed 18 to 20-kDa small Maf subunits. In MEL (mouse erythroleukemia) cells, a model of erythroid differentiatin, the absence of p45 is inhibiting chemically induced differentiation, including induction of globin genes. In vivo, p45 knockout mice were reported to show splenomegaly, severe thrompocytopenia and mild erythroid abnormalities. Most of the mice die shortly after birth due to haemorrhages. The animals that survive display increased bone, especially in bony sites of hematopoiesis. We confirmed that femurs of p45 deficient mice are filled with bone, thus limiting the space for cells. Hence, we observed a decrease in the number of hematopoietic cells in the bone marrow of 3 months old mice. In order to analyze erythroid progenitor populations we performed flow cytometry using the markers Ter119 and CD71. We found that p45 deficient mice have an increased proportion of early erythroid progenitors (proerythroblasts) and a decreased proportion of late stage differentiated red blood cells (orthochromatic erythroblasts and reticulocytes) in the spleen, when compared to wild-type mice. We showed that the liver of p45 knockout adult mice is also becoming a site of red blood cell production. The use of secondary sites, such as the spleen and liver, suggests stress erythropoiesis, likely compensating for the decreased production of red blood cells in bone marrow. In accordance with those observations, we observed about 2 fold increased levels of erythropoietin in the serum of p45 knockout mice.Overall, our data suggest that p45 NF-E2 is required for proper functioning of the erythroid compartment in vivo. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1760-1760 ◽  
Author(s):  
Mariela Sivina ◽  
Robert J. Kreitman ◽  
Amnon Peled ◽  
Farhad Ravandi ◽  
Jan A. Burger

Abstract Abstract 1760 Hairy cell leukemia (HCL) has a unique pattern of tissue infiltration by the HCL cells, causing splenomegaly and marrow infiltration, but rarely affecting the lymph nodes, or causing a significant leukemic involvement. Chemokine receptors and adhesion molecules play key roles in normal B cell trafficking and homing to different tissue microenvironments, and expression of these molecules on hairy cells is thought to function in a similar fashion, regulating the trafficking of hairy cells between the blood, spleen, and bone marrow. In this study, we profiled the expression and function of chemokine receptors (CXCR4, CXCR5, CXCR3, CCR7) and adhesion molecules (CD49d/VLA-4, CD54/ICAM-1, CD44, and CD62L) in primary HCL cells and HCL cell lines (Bonna-12, HC-1 and ESKOL). Primary HCL cells from 9 different patients displayed high levels of CD49d/VLA-4, CD54/ICAM-1, CXCR4, CD44, CD103 and CD11c; only in few cases we found a small sub-population of cells expressing CXCR5. Cell adhesion molecules like CD62L and the chemokine receptors CCR7 that are critical for entry and positioning in lymph nodes were not expressed in any of the analyzed samples. Similar immunophenotypic profiles were found in the HCL cell lines, with the exception of CD103 and CD11c, which were not expressed on the HCL cell lines. Bone marrow stroma cells (BMSC) constitutively secrete CXCL12 and provide ligands for VLA-4 integrins, and thereby may attract and retain HCL cells in the marrow. Also, these two molecules can be clinically targeted using small molecule antagonists or monoclonal antibodies (mAbs). Therefore, we analyzed HCL cell migration and adhesion towards BMSC (R-15C and Tst-4) after 4 hours of co-culture. As documented via phase contrast microscopy and quantified by flow cytometry, HCL cell lines and primary HCL cells showed abundant adhesion and migration beneath BMSC. Next, we examined whether blocking of CXCR4 using the CXCR4 antagonists AMD3100 and BKT-140 or blocking of VLA-4 integrins using CS-1 peptides affects this adhesion of HCL cells to BMSC. Pre-incubation with AMD3100 significantly reduced BMSC adhesion of hairy cells to levels that were 70.6 ±7.8% (p<0.05) of controls using Tst-4 BMSC. Similar results were found when hairy cells were pre-incubated with the CXCR4 antagonist BKT-140 (65.4 ± 10.6%, p<0.05, n=7). Interestingly, when hairy cells were pretreated with a cyclic peptide inhibitor with a minimal VLA-4 binding motif “LDV” (CS-1 peptide), a significant reduction of HCL cell adhesion was also found, reducing HCL cell adhesion to 53.0 ± 9.7% of controls (p< 0.01) with Tst-4 BMSC; similar results were found using R-15C BMSC, as shown in the Figure. In a preliminary study, with 4 cases analyzed, we also found that co-culture of primary HCL cells with BMSC improved HCL cell survival, as assessed by staining with DiOC6 and PI. BMSC co-culture increased the proportion of viable HCL cells to 134 ± 13% (n=4) of the control (HCL in culture without BMSC) on day 2 and to 252 ± 72% (n=2) on day 4. Collectively, our studies provide insight into the molecular interactions between HCL cells and BMSC and provide an explanation for the distinct pattern of tissue infiltration in HCL. Also, they provide a rationale for further preclinical development of CXCR4 and VLA-4 antagonists in HCL. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5123-5123
Author(s):  
Sandrine Susini ◽  
Séverine Mouraud ◽  
Elodie Elkaim ◽  
Julien Roullier ◽  
Sonia Luce ◽  
...  

Abstract To generate T cells throughout adult life, the thymus must import hematopoietic progenitor cells from the bone marrow via the blood. The cellular and molecular mechanisms governing the circulation of thymus-seeding progenitor cells are well characterized in mice but not in humans. The aim of the present study was to characterize the molecular mechanisms and cellular components involved in thymus colonization by lymphoid progenitors (CD34+/CD10+/CD7-/CD24-) and the early steps of thymopoiesis under physiological conditions in humans. Our results demonstrate that circulating lymphoid progenitor cells express CCR9 and CXCR4 chemokine receptors, VLA-4, VLA-5 and VLA-6 integrins and PSGL-1 and CD44 adhesion molecules. We used in vitro migration and adhesion assays to validate the functional status of these markers. As in the mouse, human circulating progenitor cells enter the thymus at the corticomedullary junction (CMJ). Once in the thymus, crosstalk with thymic epithelial cells causes the circulating progenitors to commit to the T-cell differentiation pathway. In order to characterize thymic niches and interactions between circulating progenitors and the thymic stroma, we undertook a chemokine/chemokine-receptor-focused gene expression analysis of sorted lymphoid progenitor cells and CMJ epithelial cells (based on the expression of EpCAM and Delta-like-4). We observed an unexpected gene expression profile for chemokines and chemokine regulators in thymus-seeding CD34+/CD10+/CD7-/CD24- cells and epithelial cells at the CMJ. The present results should help us to highlight candidate genes involved in the early steps of human thymopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1506-1506
Author(s):  
Min Ye ◽  
Hong Zhang ◽  
Pu Zhang ◽  
Daniel G. Tenen

Abstract Abstract 1506 Poster Board I-529 During ontogeny, hematopoietic stem cells (HSCs) undergo a change from rapid dividing cells with high reconstitution ability to mainly quiescent cells with lower repopulation capacity. However, little is known about how this switch is regulated. Here we report that levels of C/EBPα, a transcription factor that is frequently disrupted in human acute myeloid leukemia, regulate the proliferation and self-renewal transition of HSCs during development. Loss of C/EBPα in adult mice resulted in a profound expansion of phenotypic HSCs and elevated proliferation rates. Limiting dilution transplantation to measure the frequency of competitive repopulation units (CRU) revealed an increase in the number of functional HSC in C/EBPα-/- mice. Serial transplantation of C/EBPα-/- bone marrow showed advanced reconstitution ability, indicating enhanced self-renewal ability. Interestingly, levels of C/EBPα in HSCs were significantly up-regulated 3 weeks after birth during which HSCs change from an actively cycling state to quiescence in bone marrow. When we conditionally inactivated C/EBPα in mice of different age, we observed a tight correlation between the age-specific levels of C/EBPα expression and the expansion of HSCs. Gene profiling analysis of C/EBPα-/- adult HSCs showed the up-regulation of oncogenes c-myc and n-myc, whose expression can regulate pluripotency and self-renewal of stem cells, as shown by recent induced pluripotent stem cell studies. Knocking down n-myc and c-myc expression in C/EBPα-/- HSCs using shRNA, we observed reduced proliferation and decreased colony formation in serial replating assay, which assesses the preservation of “self-renewal” in the progenitor cell compartment. Consistently, we observed down-regulation of n-myc in HSCs during the transition time, which is reciprocal to C/EBPα expression. Together, our data indicate C/EBPα as a key regulator of HSC self-renewal and proliferation during development, whose levels of expression might control the fetal to adult switch of HSC properties through regulating myc genes. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4153-4153
Author(s):  
Noelia Purroy ◽  
Pau Abrisqueta ◽  
Eva Calpe ◽  
Cecilia Carpio ◽  
Carles Palacio ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) is characterized by the accumulation and proliferation of monoclonal CD5+ mature B-cells in peripheral blood (PB), lymph nodes (LN), and bone marrow (BM). The microenvironment found in BM and LN, where CLL cells interact with different accessory cells, induces active proliferation of CLL cells and protects them from spontaneous and chemotherapy-induced apoptosis. This may explain why the BM and secondary lymphoid organs are preferential sites for minimal residual disease persistence and for relapses observed in all patients after treatment. Therefore, further characterization of primary CLL cells found in the proliferative niches can facilitate the discovery and study of new targets specifically expressed by this proliferative and resistant subset of primary CLL cells. In order to further characterize the proliferative subset of primary CLL cells we analyzed the characteristics of proliferating subclones of CLL cells found in PB and compared it to the phenotype of primary CLL cells co-cultured ex vivo in conditions mimicking the microenvironment of the proliferative niches. Firstly, we compared actively proliferating CLL cells from 40 patients diagnosed with CLL with their quiescent counterpart. For this, we analyzed by flow cytometry (FC) the differential expression of ZAP-70, CD38, and the chemokine receptors CXCR4, CXCR5 and CCR7 in Ki-67 positive vs. negative CLL cells. Ki-67 positive CLL cells had higher expression levels of ZAP-70 and CD38, while the expression levels of all the chemokine receptors analyzed were significantly lower (Figure 1). This phenotype indicates that these cells may have an increased capability to respond to different survival and migration signals provided by the cellular microenvironment, since high expression of ZAP-70 and CD38 has been related to increased response to this kind of signaling. Moreover, downregulation of chemokine receptors would indicate recent stimulation by chemoatracting cytokines. Furthermore, with the aim of ex vivo mimic the microenvironment found in the proliferative centers, we co-cultured primary CLL cells from 27 patients with the BM-derived stromal cell line UE6E7T-2, 1μg/mL soluble CD40L and 1.5μg/mL CpG ODN, and analyzed the effects in terms of proliferation, modulation of surface molecules and development of chemoresistance. Of note, already 24 hours after co-culture, CLL cells increased their size, resembling stimulated B cells, and tended to be located in sparse clusters that included dividing cells, as observed after immunocytochemistry studies. In this setting, proliferative responses assessed by Ki-67 expression analyzed by FC were already observed after 24 hours (mean % Ki-67 positive cells: 1.48±0.28 in co-culture vs. 0.56±0.11 in suspension, P<0.01) and further increased after 48 hours (mean % Ki-67 positive cells: 2.89±0.51 in co-culture vs. 0.25±0.10 in suspension, P<0.01) and 72 hours (mean % Ki-67 positive cells: 7.68±2.11 in co-culture vs. 0.81±0.24 in suspension, P<0.001). Next, we analyzed the expression levels of ZAP-70, CD38 and the chemokine receptors CXCR4, CXCR5 and CCR7. Interestingly, the modulation of the expression of these molecules in co-cultured CLL cells correlated with the previously mentioned differential expression observed between the Ki-67 positive vs. negative compartments, indicating that the ex vivo co-culture highly resembles the conditions found in proliferative niches (Figure 1). Finally, in order to assess the role of microenvironment in chemoresistance, CLL cells were cultured in suspension and in co-culture for 48 hours and subsequently treated with increasing doses of fludarabine for 24 hours. Interestingly, the co-culture of CLL cells inhibited at such extend the capacity of fludarabine to induce apoptosis that it was not possible to calculate its lethal dose 50, whereas lethal dose 50 for fludarabine in CLL cells in suspension was 416μM (95%CI 125.5-1379). In conclusion, these results suggest that culturing CLL cells in the previously described conditions promotes proliferation and the acquisition of a phenotype which bears many similarities to the one found in actively proliferating CLL cells circulating in PB. This study provides a model for a co-culture system which might serve as a basis for the development and testing of new drugs that target the proliferative and drug resistant CLL cell compartment. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2226-2226
Author(s):  
Tai-Chung Huang ◽  
Yi Yang ◽  
Cory Brayton ◽  
Wen-Chien Chou ◽  
Akhilesh Pandey

Abstract Abstract 2226 Background: MicroRNAs (miRs) play important roles in normal hematopoiesis as well as in hematopoietic malignancies. In generating mature miRs, Dicer-1, an RNase III in the cytosol, excises the hairpin-loop of precursor miRs and assembles mature miRs onto an RNA-Induced Silencing Complex. Among 27 exons of Dicer1, exons 21 and 22 comprise the RNase IIIa domain. Knockout of this domain was shown to be lethal in mouse embryos. In this study, we used tamoxifen to induce Cre-mediated exon deletion and our goal was to investigate the impact of Dicer1 knockout on hematopoiesis in adult mice. Material and Methods: Two C57BL/6 transgenic mice were crossed: ROSA26-CreERT2 and floxed Dicer1 exons 21 & 22. At the age of 8 week, three female mice were treated with tamoxifen (1 mg/day, Day 1 to 5) and two with placebo. On Day 8, mice were sacrificed for examination. DNA genotyping and qRT-PCR were used to confirm Dicer1 genomic deletion and to quantify the transcription copy number of Dicer1. Cardiocentesis was done for complete peripheral blood count. Femoral and tibial bone marrow, spleen and inguinal lymph nodes were evaluated by flow cytometry whereas formalin-perfused sternum and thymus were subjected to histopathological examination. Results: Two tamoxifen-treated mice presented with unkempt appearance, hunched posture and diarrhea from Day 7. Vehicle-treated control mice remained clinically normal. qRT-PCR showed lower copy number of Dicer1 exons 21 & 22 in tamoxifen-treated group than in control (0.09 vs. 1.00). In tamoxifen-treated group, the spleen weights were lower, white blood cell and platelet counts were lower and bone marrow histopathology revealed hypocellularity, with markedly reduced and immature myeloid elements, consistent with arrested or abrogated development of myelomonocytic and megakaryocytic elements. Conclusions: The RNase III domain of Dicer-1 is crucial for the normal differentiation of marrow myeloid series in adult mice, probably at the stage of common myeloid progenitor. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2320-2320
Author(s):  
Per Holmfeldt ◽  
Jennifer Pardieck ◽  
Shannon McKinney-Freeman

Abstract Abstract 2320 Hematopoietic stem cells (HSCs) are both necessary and sufficient to sustain the complete blood system of vertebrates. Specified at several locations during fetal development, they ultimately congregate in the fetal liver for rapid expansion. Around birth HSCs relocate to the bone marrow (BM) and enter a state of cellular quiescence, cycling intermittently to supply progenitor cells, which differentiate into the distinct blood lineages. Due to their regenerative potential, HSCs are heavily utilized in the clinic for bone marrow transplants (BMTs) to treat a variety of diseases. However, a lack of suitable donors and inefficiency in recipient engraftment currently limit this life saving therapy. To improve BMT regimens, a better understanding of regulators of HSC BM engraftment is required. Recently, we examined the gene expression patterns of HSCs as they emerge throughout murine ontogeny (McKinney-Freeman et al., Cell Stem Cell, in press). We observed that the transcription factor Nfix, a member of the nuclear factor I (NFI) family of transcription factors never before linked to HSC biology, was highly expressed by both fetal liver and BM HSCs. These data suggest that Nfix may play a novel role in regulating HSC function in the BM and/or fetal liver. To test this hypothesis, HSCs were enriched from E14.5 fetal liver or adult BM (Lineage-, c-kit+, Sca-1+ (LSK) cells) and then transduced with lentiviruses carrying shRNAs targeting Nfix. Twenty-four hours post-transduction, cells were injected into lethally irradiated mice along with untransduced BM LSK competitor cells congenic at the CD45 allele. The peripheral blood of recipient mice was then analyzed periodically over 16 weeks for engraftment of the Nfix-depleted cells. Depletion of Nfix by two independent shRNA (confirmed by Western blot analysis to deplete NFIX protein levels to <20% of baseline) resulted in a significant decrease in the repopulating activity of BM LSK cells relative to LSK cells transduced with either of two independent control shRNAs. As early as two weeks post-transplant, a 22% +/− 5% (p=0.03) reduction in repopulating activity was observed. By 16 weeks post-transplant, this reduction in repopulating potential had gradually increased to 55% +/−8% (p<0.0001) in four independent experiments. Depletion of Nfix in fetal liver-derived LSK cells resulted in a similar loss of repopulating potential. Critically, in vitro analysis of BM LSK expansion in liquid culture and differentiative potential, as analyzed by the methylcellulose based colony forming assay, revealed no differences in the activity of LSK cells transduced with Nfix-specific shRNAs compared to controls. Thus, it is unlikely that the observed decrease in BM repopulating activity is due to general cytotoxicity resulting from Nfix depletion or a block in differentiation. Concordantly, lineage analysis of peripheral blood of recipients showed no significant differences in the percentage of the major blood lineages derived from LSK cells transduced with Nfix-specific shRNAs compared to controls. Thus, the observed decrease in repopulating activity likely occurs at the level of HSCs and multipotent progenitors. In agreement with this conclusion, when BM of recipients transplanted with Nfix-depleted LSK cells are examined 4 and 16 weeks post transplant, a loss of phenotypic HSCs (LSK/CD150+/CD48-) relative to controls is evident. The loss of repopulating potential by Nfix-depleted cells as early as two weeks post-transplant suggests that Nfix may be involved in either the homing/lodgment of HSCs in the BM or their ability to expand soon after incorporation into the stem cell niche. We are presently teasing out the molecular mechanism behind this phenomenon. Furthermore, to examine a role for Nfix during HSC homeostasis, we are currently analyzing mice in which Nfix has been conditionally ablated in the hematopoietic compartment. Finally, functional analysis of two other members of the NFI gene family shown by our array analysis to be expressed by BM HSC, Nfia and Nfic, will further assess a role for this gene family in the regulation of HSCs. In summary, we have for the first time established a role for a member of the NFI gene family, Nfix, in HSC biology, as evident by a decrease in BM repopulating activity in Nfix-depleted HSCs. By dissecting the precise role of Nfix in HSC biology, we will glean insights that could improve our understanding of graft failure in clinical BMTs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2692-2692
Author(s):  
Jillian Jafek ◽  
Arvind Shakya ◽  
Pei-Yi Tai ◽  
Gerald J Spangrude ◽  
Todd W Kelley ◽  
...  

Abstract Oct1, encoded by the Pou2f1 gene, is a widely expressed transcription factor that is associated with metabolic control, stress response, stem cell phenotypes and malignant transformation. Oct1 associates with a number of cofactors including the activating cofactor OCA-B encoded by Pou2af1. OCA-B is normally expressed specifically in B cells, but is misexpressed in about 30% of AML cases in myeloid cells and is a poor prognostic indicator. We observed a conserved Oct1 binding site in the promoter of Cdx2, a homeodomain transcription factor that is normally expressed solely in the gastrointestinal tract of adult humans, but has been shown to misexpressed in blood cells in almost all cases of AML. Our study looks at the interaction of Oct1 with Cdx2, and the effects of Oct1 and OCAB-B knockout in a mouse model of AML. Using ChIP we determined that Oct1 is bound to the Cdx2 promoter in normal patient samples and AML patient samples. However, in normal patient samples a repressive cofactor (the NuRD complex) is also bound. In AML patient samples an activating cofactor (Jmjd1a) is bound. This suggests that in AML Oct1 is bound to Cdx2 and is associating with Jmjd1a, which can occur through ERK signaling or OCA-B expression, and activating expression of Cdx2. We next wanted to determine the effect of Oct1 knockout in AML using a MLL-AF9 mouse model. Our lab has developed a conditional Oct1 allele, as Oct1 germline knockout is embryonic lethal. We harvested bone marrow from Oct1+/fl;MX1-Cre, Oct1fl/D;MX1-Cre, and Oct1+/+;MX1-Cre mice and transduced the bone marrow with virus expressing MLL-AF9. Recipient mice were lethally irradiated and transplanted with the transduced bone marrow. Mice were injected with Poly IC at 5 weeks post-transplant to delete Oct1 foxed alleles. Mice that were wildtype for Oct1 developed leukemia around 7 weeks. Mice with one allele of Oct1 survived longer but still developed leukemia. Mice with no Oct1 survived longer still, but eventually suffered from bone marrow failure. In order to assess whether the lethality in Oct1 deficient mice with MLL-AF9-induced leukemia was due to the presence of the MLL-AF9 oncoprotein or if Oct1 deletion is lethal in adult mice under normal conditions, we treated non-leukemic mice that were Oct1+/fl;MX1-Cre or Oct1fl/D;MX1-Cre with Poly IC to delete Oct1 at 5 weeks. Following Poly IC injection, mice with and without Oct1 appeared normal and healthy by examination and by blood counts. At 20 weeks, all mice were injected with 5-flurouracil (5-FU). We observed that mice that were deficient in Oct1, namely Oct1fl/D;MX1-Cre mice injected with Poly IC, died within 2 weeks of 5-FU treatment. Put together, our data suggests that Oct1 is not necessary for normal hematopoiesis, as adult mice with no Oct1 appear normal under homeostatic conditions, but Oct1 is necessary for the hematopoietic proliferative stress response. The protective effect of Oct1 deletion against leukemia in mice suggested an important role for Oct1 in regulating the leukemic phenotype. However, since Oct1 deletion is also lethal in the leukemic context, we looked at the effect of deletion of OCA-B in leukemia. OCA-B germline deficient mice are viable and the only detectable phenotype is the lack of germinal center formation. We harvested OCA-B germline deficient and wildtype control bone marrow and followed the same protocol as outlined above with the transduction of the bone marrow with MLL-AF9. Mice with no OCA-B did not develop leukemia and had 100% survival. This striking result suggests that OCA-B could be a therapeutic target in MLL-AF9 AML. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 12 (7) ◽  
Author(s):  
Chao Liu ◽  
An-Song Liu ◽  
Da Zhong ◽  
Cheng-Gong Wang ◽  
Mi Yu ◽  
...  

AbstractBone marrow-derived mesenchymal stem cells (BM-MSCs), the common progenitor cells of adipocytes and osteoblasts, have been recognized as the key mediator during bone formation. Herein, our study aim to investigate molecular mechanisms underlying circular RNA (circRNA) AFF4 (circ_AFF4)-regulated BM-MSCs osteogenesis. BM-MSCs were characterized by FACS, ARS, and ALP staining. Expression patterns of circ_AFF4, miR-135a-5p, FNDC5/Irisin, SMAD1/5, and osteogenesis markers, including ALP, BMP4, RUNX2, Spp1, and Colla1 were detected by qRT-PCR, western blot, or immunofluorescence staining, respectively. Interactions between circ_AFF4 and miR-135a-5p, FNDC5, and miR-135a-5p were analyzed using web tools including TargetScan, miRanda, and miRDB, and further confirmed by luciferase reporter assay and RNA pull-down. Complex formation between Irisin and Integrin αV was verified by Co-immunoprecipitation. To further verify the functional role of circ_AFF4 in vivo during bone formation, we conducted animal experiments harboring circ_AFF4 knockdown, and born samples were evaluated by immunohistochemistry, hematoxylin and eosin, and Masson staining. Circ_AFF4 was upregulated upon osteogenic differentiation induction in BM-MSCs, and miR-135a-5p expression declined as differentiation proceeds. Circ_AFF4 knockdown significantly inhibited osteogenesis potential in BM-MSCs. Circ_AFF4 stimulated FNDC5/Irisin expression through complementary binding to its downstream target molecule miR-135a-5p. Irisin formed an intermolecular complex with Integrin αV and activated the SMAD1/5 pathway during osteogenic differentiation. Our work revealed that circ_AFF4, acting as a sponge of miR-135a-5p, triggers the promotion of FNDC5/Irisin via activating the SMAD1/5 pathway to induce osteogenic differentiation in BM-MSCs. These findings gained a deeper insight into the circRNA-miRNA regulatory system in the bone marrow microenvironment and may improve our understanding of bone formation-related diseases at physiological and pathological levels.


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.


Sign in / Sign up

Export Citation Format

Share Document