Serotonin and Its Metabolism Enhances Megakaryopoiesis and Proplatelet Formation

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4753-4753
Author(s):  
Jie yu Ye ◽  
Fan yi Meng ◽  
Qianli Jiang ◽  
Su yi Li ◽  
En yu Liang ◽  
...  

Serotonin (5-HT) has been recently identified as a novel growth factor. We previously demonstrated that 5-HT enhances murine megakaryopoiesis via 5-HT2 receptors and has promotor effect on hematopoiesis(Yang M et al, Stem Cells, 2007). However, the molecular mechanism remains under explored. In the terminal stage of mammalian megakaryocyte development, platelets are released from proplatelet protruding from megakaryocytes via cytoskeleton reorganization. 5-HT is shown to modulate cell migration and remolding by activating cytoskeleton reorganization, but the effects of 5-HT on proplatelet formation have not been investigated. Our results showed that 5-HT significantly promoted human CFU-MK formation and reduced apoptosis on human megakaryocytes through phosphorylation of Akt. These effects were attenuated by addition of ketanserin, a 5HT2 receptor inhibitor. 5-HT also stimulated proplatelet formation through activating the p-Erk1/2 expression and F-actin reorganization. Melatonin, the metabolism of 5-HT, promoted the recovery of platelets and the formation of bone marrow colony forming units in irradiated mice. Our findings suggested that 5-HT and melatonin plays an important role in human megakaryopoiesis. Interaction of 5-HT and 5-HTR2B induced downstream activation of PI3-k/Akt signal pathway leading to human MK cell proliferation. In addition, activation of 5-HTR2B also induced Erk1/2 phosphorylation, which then promoted cytoskeleton reorganization and subsequent proplatelet formation. We also proved that melatonin exerts a protective effect on MK and platelets in the irradiation mice model. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 218-218 ◽  
Author(s):  
Qiu-Sha Huang ◽  
Jing Xue ◽  
Chen-Cong Wang ◽  
Ya-Zhen Qin ◽  
Lan-Ping Xu ◽  
...  

Introduction Sphingosine-1-phosphate (S1P) is now emerging as a vital lipid mediator. Activation of sphingosine kinase (SphK) produces intracellular S1P, which in turn can be secreted out of the cell and act extracellularly by binding to S1P receptors (S1PR). Recent studies suggest that the "inside-out" signaling by S1P in megakaryocytes (MKs) plays a critical role in proplatelet formation (PPF) (Blood, 2013; J EXP MED, 2012). PPF requires a profound reorganization of the MK actin and tubulin cytoskeleton. Rho GTPases which can be activated by S1P, including Rac1 and Cdc42, have been shown to be master regulators of cytoskeletal rearrangements. The pathogenesis mechanisms of immune thrombocytopenia (ITP) are not entirely understood. Our previous data indicated that impaired PPF contributed to the development of thrombocytopenia in ITP. To further explore the underlying mechanism of impaired PPF in ITP, we found that S1P-mediated microtubule reorganization is defective in PPF of ITP. All-trans retinoic acid (ATRA), which has demonstrated to be a promising option for ITP patients in our previous study (Lancet Haematology, 2017), could correct the altered microtubule reorganization and promote PPF. Methods Thirty consecutive patients with primary ITP and 20 healthy donors were enrolled in our study. MKs were isolated from bone marrow samples, and they were collected again after ITP patients received ATRA therapy. MK mRNA sequencing by microarray was used to assess the difference of gene expression between ITP and controls. Microtubule regrowth assay was performed to observe microtubule dynamic behavior. In this assay nocodazole was first used to induce complete depolymerization of microtubule network, followed by drug washout to allow microtubule regrowth over time. ATRA was added to the culture medium of MKs to determine the mechanism of ATRA in correcting impaired PPF. Additionally, ITP mice model was established to observe the therapeutic effects of ATRA in PPF. Pf4-Cre/loxP system was used to specifically knock down gene of MKs. Results S1P concentration in bone marrow from ITP patients was lower compared to healthy donors. MKs mRNA sequencing demonstrated that S1P synthetase SphK2 and S1P receptor S1PR1 gene were downregulated while S1P lyase (SPL) gene was upregulated in ITP patients, which caused abnormal S1P signaling. Furthermore, we observed that PPF capacity of MKs in patients with ITP was reduced. Pharmacological disruption of S1PR1 blocked PPF, exogenous S1P corrected impaired PPF. Collectively, deregulation of S1P signaling was associated with impaired PPF in ITP. To verify the downstream role of S1P in regulating PPF, the Rho GTPases detection of MKs revealed a decrease in Cdc42 and Rac1 levels from ITP patients. Immunofluorescence of the differentiated MKs showed that the expression and distribution of β1 tubulin were abnormal from ITP patients. Early PPs from MKs of healthy donors displayed a well-organized tubulin bundles resembling bunches of grapes. In contrast, in MKs from ITP patients, tubulin was disorganized in thick bundles. In addition, TEM analysis of the MKs showed an irregular distribution of granules, tortuous membranes and impaired proplatelet structure. In microtubule regrowth assay, MKs from ITP patients had significantly lower microtubule regrowth at 10 min post-nocodazole washout compared with controls. Together, microtubule alteration resulted in impaired PPF in ITP. We tested whether S1P pathway were required for microtubule reorganization, both SphK2-/- and S1PR1-/- mice displayed significantly reduced S1P, Cdc42 and Rac1, altered microtubule architecture and defective PPF. Taken together, abnormal S1P pathway accounted for impaired microtubule reorganization in ITP. Next, we explored the effect of ATRA on microtubules reorganization in ITP patients, our data showed that in vitro treatment with ATRA restored microtubules structure by upregulating S1P and activating Rho GTPases. In vivo studies showed that ARTA could rescue the impaired PPF in both patients and mice model with ITP. Conclusions The MKs of ITP patients displayed defective cytoskeletal reorganization regulated by S1P pathway. ATRA restored cytoskeletal structure and corrected impaired PPF by upregulating S1P and activating Rho GTPases. It sheds light on a novel mechanism of ITP pathogenesis and provides a basis for the therapeutic potential of ARTA in ITP patients. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3347-3347
Author(s):  
Mo Yang ◽  
Fanyi Meng ◽  
Jie Yu Ye ◽  
Yue Xu ◽  
Bin Xiao ◽  
...  

Abstract Abstract 3347 We have reported that serotonin (5-HT) show a promoting effect on cord blood CD34+ stem/progenitor cells (Yang et al, Stem Cells 2007). We also demonstrated that serotonin enhances murine megakaryopoiesis via 5-HT2 receptors (Yang et al. Blood Coagul Fibrinol 1996). In this present study, we explored how serotonin regulated human megakaryocytopoiesis, proplatelet formation, and thrombopoietin (TPO) production. Our results indicated that serotonin significantly promoted human CFU-MK formation and reduced apoptosis in megakaryocytes through phosphorylation of Akt. These effects were attenuated by addition of ketanserin, a 5-HT2 receptor inhibitor. In addition, serotonin was able to stimulate the F-actin reorganization in megakaryocytes through activating the p-Erk1/2 expression. Bone marrow mesenchymal stromal cells (MSCs) are important in regulating megakaryocytopoiesis through stimulating release of thrombopoietic growth factor, such as TPO. Our studies suggested that when activated by serotonin, bone marrow MSCs were induced to release significant amount of TPO by q-PCR, ELISA and cytokine-array assays. Our findings demonstrated an important role of serotonin played on megakaryocytopoiesis. This effect was likely mediated via 5HT2 receptors with subsequent activation of Akt and Erk 1/2 phosphorylation, which led to survival of megakaryocytes and proplatelet formation. Serotonin also stimulated TPO released from MSCs, which indirectly promoted megakaryopoiesis. In present studies, we have demonstrated a positive “feed-back” control loop between MK-derived granule- serotonin and megakaryocytopoiesis. These findings improved our knowledge on megakaryocytopoiesis regulation and provided new clues on identifying novel thrombopoietic agents. It also deepened our understandings on how TPO production is regulated. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 14-14
Author(s):  
Mo Yang ◽  
Enyu Liang ◽  
Jieyu Ye ◽  
Beng H Chong ◽  
Liang Li

Background: Our previous work confirmed that serotonin (5-HT) promotes the proliferation of hemopoietic stem cells and megakaryocytes (Yang M et al, Stem Cells, 2007; 2014). However, the mechanisms remain indefinite. Methods: Q-PCR, Flow Cytometry, Western Blot, or Immunofluorescence microscope were used in the receptor and TPO study. MTT/CCK-8, Proplatelet assay, and Flow Cytometry were also used in cell proliferation and apoptosis study. The relationship between 5-HT and TPO was studied in a traumatic stress mice model. Results: In-vitro study, there was a stimulating effect of 5-HT on proplatelet formation in human bone marrow megakaryocytes. Human BM MK progenitors cultured in serum-free medium with either 5-HT (200nM) or TPO (100 ng/ml) had more proplatelet bearing MKs than the control group (5-HT (12.3 ± 5.0)% vs. Control (6.2 ± 3.5)%, P=0.025; TPO (15.6 ± 2.5)% vs. Control, P=0.04; n=4). The 5-HT treatment group showed more mature and more in the final stage MK cells as compared to the TPO group. 5-HT2A, 2B, 2C receptors were detected in the surface of megakaryocytes. The effect of 5-HT on proplatelet formation in MK cells was via 5-HT2 receptors and this effect was reduced by 5-HT2 receptor inhibitor ketanserin. 5-HT acted on cytoskeleton reorganization in MKs via 5-HT2 receptors and ERK1/2 pathway. Using an immunofluorescence microscope with F-actin specific binder rhodamine-phalloidin staining, the polymerized actin level was lower in the control group than the 5-HT group and actin distributed diffusely throughout the cytoplasm. In contrast, the polymerization actin level was higher in the 5-HT group. Adding ketanserin and ERK1/2 inhibitor PD98059 to 5-HT treatment, the fluorescence intensity was correspondingly reduced. Our data also demonstrated that ERK1/2 was activated in MKs treated with 5-HT for 30 minutes. In a traumatic stress mice model, both of 5-HT and TPO were increased, but the increasing of TPO is posterior to 5-HT. After added LX1606, the synthesis inhibitor of 5-HT, 5-HT was reduced markedly, as well as TPO. The expression of TPO mRNA and the production of TPO protein were increased as compared with the control in this model. Conclusions: This study suggests that 5-HT promotes thrombopoiesis from two aspects: one is the direct effect on megakaryocytes. 5-HT could promote the proplatelet formation from megakaryocytes. The second is the indirect effect by promoting the production of TPO, which is a paracrine secretion to influence thrombopoiesis. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3348-3348
Author(s):  
Mo Yang ◽  
Fanyi Meng ◽  
Jie yu Ye ◽  
Yue Xu ◽  
Bin Xiao ◽  
...  

Abstract Abstract 3348 Platelet-derived growth factor (PDGF), a platelet alpha-granule molecule, imply their potential effect in the regulation of megakaryocytopoiesis and thrombopoiesis, which also intimates the existence of an autocrine and/or paracrine loop constructed by megakaryocytes/platelets and their granular constituents. Our previous studies demonstrated the presence of functional PDGF receptors (PDGFR) on human megakaryocytes and platelets (Yang et al, Thromb Haemastasis, 1997) and CD34+ cells, and their ability to mediate a mitogenic response. PDGF promoted the ex vivo expansion of human hematopoietic stem (CD34+) and progenitor (CD41+ CD61+) cells. More significantly, PDGF enhanced the engraftment of human CD45+ cells and their myeloid subsets (CD33+, CD14+ cells) in NOD/SCID mice. PDGF stimulated in vitro megakaryocytopoiesis via PDGFR and/or the indirect effect on bone marrow microenvironment to produce TPO and other cytokines. It also showed a direct stimulatory effect of PDGF on c-Fos, GATA-1 and NF-E2 expressions in megakaryocytes. We speculate that these transcription factors might be involved in the signal transduction of PDGF on the regulation of megakaryocytopoiesis. PDGF also enhanced platelet recovery in mice model with radiation-induced thrombocytopenia. Studies showed that PDGF, like thrombopoietin (TPO), significantly promoted platelet recovery and the formation of bone marrow colony-forming unit-megakaryocyte (CFU-MK) in this irradiated-mouse. An increased number of hematopoietic stem/progenitor cells and a reduction of apoptosis were found in the bone marrow histology sections. In the M-07e apoptotic model, PDGF had a similar anti-apoptotic effect as TPO on megakaryocytes. We also demonstrated that PDGF activated the PI3k/Akt signaling pathway, while addition of imatinib mesylate reduced p-Akt expression. Our findings suggested that the PDGF-initiated radioprotective effect is likely to be mediated via PDGF receptors with subsequent activation of the PI3k/Akt pathway. The study provides a possible explanation that blockage of PDGFR may reduce thrombopoiesis and play a role in imatinib mesylate-induced thrombocytopenia in the treatment of CML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 755-755 ◽  
Author(s):  
Maria Feola ◽  
Andrea Zamperone ◽  
Weili Bao ◽  
Tenzin Choesang ◽  
Huihui Li ◽  
...  

Abstract Erythropoiesis is a process during which multipotent hematopoietic stem cells proliferate, differentiate and ultimately produce enucleated reticulocytes. Terminal erythroid differentiation begins at the morphologically recognizable pro-erythroblast (pro-E) stage and is completed when orthochromatic erythroblasts (ortho-E) expel their nuclei to produce reticulocytes. Progressive differentiation between these stages occurs in homologous cell division progressively doubling proportions of pro-E, basophilic (baso-E), polychromatophilic (poly-E), and ortho-E, and multiple signaling pathways are involved in the generation of enucleated erythroid cells, including multiple steps requiring actin cytoskeleton reorganization. We have previously shown that β-thalassemic mice (th1/th1) demonstrate a disordered progression from pro-E to baso-E and that exogenous transferrin therapy restores normal proportion of early stage erythroid precursors in th1/th1 mice (Liu Blood 2013). To identify genes that play novel function in different stages of terminal erythropoiesis, we performed RNA seq analysis of sorted bone marrow pro-E from WT, th1/th1, and transferrin-treated th1/th1 mice. We identify pleckstrin-2 (plek2) as a gene of interest with a 15-fold increase in plek2 mRNA expression in th1/th1 relative to WT mice, normalized in transferrin-treated th1/th1 mice. Plek2 is an actin binding protein, like pleckstrin-1, contains a central DEP domain known to bind RacGTPase, is Epo dependent, and is expressed in all stages of terminal erythropoiesis. We evaluate plek2 mRNA and protein expression in sorted bone marrow erythroid precursors from WT, th1/th1, and transferrin-treated th1/th1 mice. Our data demonstrates a statistically significant increase in plek2 mRNA in th1/th1 relative to WT mice, with the highest expression of plek2 in poly-E, normalized in transferrin-treated th1/th1 mice (Figure 1A). A similar pattern of increased protein concentration in th1/th1 relative to WT mice and normalization in transferrin-treated th1/th1 mice is evident in sorted bone marrow samples (Figure 1B). Prior in vitro studies demonstrate that membrane localization of plek2 is required for erythroid differentiation. Thus, we performed sub-cellular fractionation in bone marrow erythroid precursors and determined for the first time that in sorted erythroblasts from WT bone marrow, plek2 is found exclusively in the cytoplasm in pro-E and in both cytoplasm and membrane from baso-E to ortho-E (Figure 2), co-localized with actin filaments in the membrane (data not shown). In contrast, sorted erythroblasts from th1/th1 bone marrow reveal membrane-associated plek2 starting from pro-E, demonstrating earlier co-localization with actin filaments (data not shown) and suggesting an earlier activation of plek2 and consequent actin cytoskeleton reorganization during erythroid differentiation in th1/th1 mice, normalized in transferrin-treated th1/th1 mice (Figure 2). Erythropoiesis involves a complicated and incompletely understood set of potentially related molecular signals influencing cell survival, differentiation, enucleation, and release into the circulation. For example, although Epo increases survival, Epo signaling also activates RacGTPases, inhibiting enucleation. Recent in vitro data demonstrates that knockdown of plek2 affected enucleation with significantly lower reticulocyte count. Although the involvement of RacGTPase in plek2-mediated erythroid differentiation has not been explored, we hypothesize that plek2 activation triggers RacGTPase and prevents enucleation in th1/th1 mice. Our data demonstrates that RacGTPase concentration is increased in sorted bone marrow erythroid precursors from th1/th1 relative to WT mice and normalized in transferrin-treated th1/th1 mice (Figure 1B). These results suggest that plek2 plays an important role in erythropoiesis likely as a key factor in the improved enucleation of transferrin-treated th1/th1 mice. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2144-2144 ◽  
Author(s):  
Xiaoli Cao ◽  
Mingfeng Zhao ◽  
Deguan Li ◽  
Yi Xing ◽  
Jie Chen ◽  
...  

Abstract Iron overload (IO) is a kind of disease with an excess of iron depositing in the body's organs and tissues, resulting in the dysfunction of these organs and tissues, which contains hereditary hemochromatosis and secondary IO such as ineffective hematopoiesis in aplastic anemia and myelodysplastic syndrome (MDS) and blood transfusion. Numerous researches and our previous study found that IO can damage the hematopoietic function in bone marrow. MDS is one of the most common IO diseases and IO can further injure the hematopoietic function especially on erythropoiesis in MDS.However, it is indeterminacy on the mechanism behind this stiuation. This study is to explore the possible mechanism of IO on the injury of MDS erythropoiesis though establishing an IO MDS mice model. In this study, we first established an IO mice model by administering iron dextran(25 mg/ml) by intraperitoneal injection every 3 days for 2 months in wild and MDS mouse(NUP98-HOXD13 transgenic mouse). The mouse were devided into four grups: the wild mouse without IO(Ctrl group), the wild mouse with IO (IO group), MDS mouse without IO(MDS group), the MDS mouse with IO(MDS+IO group). After 6 months, the establishment of IO mice models were confirmed by both the iron deposits staining of bone marrow(BM), liver, spleen and the labile iron pool level of bone marrow mononuclear cells( BMMNCs). We next found that IO damage the erythropoiesis in MDS mice. We detected the proportion of different stages BM erythroblasts by flow cytometry plots of Ter119+ CD71+ cells (R1, basophilic erythroblasts, poly red cells, immature red blood cells) and Ter119+ CD71- cells (R2, late erythroblast and reticulocyte ). It was found that the hematopoietic colony-forming unit (CFU-E, BFU-E) in MDS+IO group were less than that in other groups. The proportion of R1(10.39±1.43) and R2(1.58±0.54) in MDS+IO group were the least compared with Ctrl group(R1 17.36±3.45,R2 4.59±1.57),IO group(R1 14.53±2.65,R2 2.56±0.76) and MDS group(R1 13.64±3.27,R2 2.35±0.78). And the reticulocytes proportion in MDS+IO group(11.36±3.65) was the highest compared with Ctrl group(5.159±1.537), IO group(7.61±1.36)) and MDS group(7.35±1.39). Then we further explored the mechanism of IO damage on MDS erythropoiesis. It was reported that TGF-β family and oxidative stress (ROS) played an important role on erythropoiesis. Our previous studies also had shown that the ROS and GDF11(a member of TGF-β family) levels were raised in BMMNCs on C57BL/6 mice with IO. Similarly, we detected them on the BM erythroblast in this study. Firstly, we sorted the R1 and R2 erythroblasts,then we performed RT-PCR to analyze mRNA levels of the TGF-β family. We found that the mRNA levels of GDF11, GDF15, Activin B, Acvr2b and ALK5 in MDS+IO group were the highest compared with that of other groups, while the mRNA levels of Activin A, ALK4 and BMP8 had no significant change in these groups. As we found that the change of GDF11 level was the most significant in MDS+IO group compared with other groups, we focused on the study of GDF11 in the following experiment. The concentration of GDF11 in peripheral serum was detected by ELISA method. It was found that the concentration of GDF11 was the highest in MDS+IO group compared with other groups. And also, the GDF11 related signal pathway proteins (Smad 2, Smad 3, p-Smad 2 and p-Smad 3) were significantly elevated in R1 and R2 erythroblast in MDS+IO group compared with other groups,which indicated that GDF11 related signal pathway was involved in the deficient of BM erythropoiesis in MDS+IO mice. Furthermore, We found that the ROS levels in Ter119+ cells in MDS+IO group, MDS group and IO group were increased 4.15 folds, 2.16 folds , 2.63 folds compared with Ctrl group. RT-PCR was used to detect the mRNA expression levels of the gene NOX4 related to ROS generation and GPX1 related with ROS clearance. It was found that the NOX4 expression levels were the highest in MDS+IO group compared with other groups,while the GPX1 mRNA level was the least in MDS+IO group. In conclusion, our study indicated that chronic IO damage the erythropoiesis in MDS mouse by increasing GDF11 and ROS levels which would provide an theoretical basis to find new targets for the treatment of IO in MDS patients with erythropoiesis dysfunction. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2108-2108
Author(s):  
Yuan Shan Cheng ◽  
Yuan Sheng Liu ◽  
Godfrey ChiFung Chan ◽  
Jie Yu Ye ◽  
Yu Lung Lau ◽  
...  

Abstract We previously reported that serotonin (5-HT) is a growth factor for hematopoietic stem cells and megakaryocytic progenitor (Yang et al, Stem Cells, 2007). We further proposed a possible role of serotonin on megakaryocyte differentiation and platelet formation. The effect of serotonin on proplatelet formation and F-actin reorganization in human megakaryocytes (MKs) was investigated in this study. Our results showed that: There was a stimulating effect of serotonin on proplatelet formation in human bone marrow megakaryocytes. Human BM MK progenitors cultured in serum free medium with either 5-HT (200nM) or TPO (50 ng/ml) had more proplatelet bearing MKs than the control group (5-HT(11.33% ± 4.93) vs. control (6% ± 3.60), P=0.026; TPO (14.66% ± 1.53) vs. control, P=0.043; n=3). The 5-HT treatment group showed more mature and more in the final stage MK cells as compared to TPO group; The effect of serotonin on proplatelet formation in Meg-01 cells were via 5-HT2 receptors. Meg-01 cells strongly expressed 5-HT 2A, 2B, 2C receptors by using western blot method. 5-HT also promoted proplatelet formation in these cells and this effect was reduced by 5-HT2 receptor inhibitor ketanserin (KE); and Serotonin acted on cytoskeleton reorganization in human megakaryocytes via 5-HT2 receptors and ERK1/2 pathway. Using an immunofluorescence microscope with F-actin specific binder rhodamine-phalloidin staining, the polymerized actin level was lower in the control group (serum free) than the 5-HT group and actin distributed diffusely throughout the cytoplasm. In contrast, polymerization actin level was higher in 5-HT group. Adding ketanserin and ERK1/2 inhibitor PD98059 to 5-HT treatment, the fluorescence intensity was correspondingly reduced (5HT vs. Control, P=0.006; 5-HT vs.5-HT plus KE, P=0.014; n=6). Our data also demonstrated that ERK1/2 was activated in MK cells treated with 5-HT for 30 minutes (21.76% ± 7.42). Our studies showed that serotonin had a stimulating effect on proplatelet formation and F-actin reorganization in human megakaryocytes and this effect involved the 5-HT2 receptors and the activation of ERK1/2 pathway.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2300-2300
Author(s):  
Shugo Kowata ◽  
Sumio Isogai ◽  
Kazunori Murai ◽  
Shigeki Ito ◽  
Koujiro Tohyama ◽  
...  

Abstract Abstract 2300 Introduction: The derivation of blood platelets from bone marrow (BM) MKs has been known since 1906, and it evoked long dispute for the process of platelet production between proplatelet model and platelet territory model. The proplatelet model, in which MKs extend long thin protrusion into the sinusoidal lumen and the tip of proplatelet then bud off, is supported by the images from in vitro cultured MK and fixed BM tissue. The platelet territory model, in which MK cytoplasm is divided by the membrane system into future platelet territories, is supported by the images by electron microscopy from fixed BM tissue. However, the process of platelet production in vivo remains still enigmatic, because no one could visualize clearly the platelet production from MK in intact BM. Recently, Junt T et al.(1) revealed that MKs routinely release heterogeneous large particles into sinusoids in BM, but their images did not capture the typical long thin proplatelets in sinusoidal lumen using intra-vital microscopy. In this study, we visualized the fine structure of MK protrusions in sinusoidal lumen to reveal the detailed process of platelet production in intact BM of mice skull using intra-vital microscopy. Materials and methods: Two- photon microscope was used to focus on the MK protrusions in sinusoidal lumen of anesthetized green fluorescence protein-transgenic mice to visualize the dynamic platelet production in details. The obtained sequential images were reconstituted as the time-lapse movie, and analyzed in the shape and size of protrusion with the relation to dynamic velocity. To confirm the phenomena captured with the live images, scanning electron microscopy (SEM) was used in fixed BM samples. Results: The time-lapse live images clearly revealed that MK released two types of protrusion into the sinusoidal lumen. One was elliptically-shaped protrusion, followed by typical proplatelets in the sinusoidal lumen (Figure 1). The proplatelet was released as a thin string into sinusoid, but not discoid individual platelet. The other was a thick string, originated from thick protrusions from MK, into sinusoidal lumen (Figure 2). The thick protrusion clearly differed from the typical proplatelet in the size, releasing point, and dynamic movement to the blood flow. Using SEM, we could confirm morphologically two types of platelet production, elliptically-shaped protrusion, followed by typical proplatelets and platelet territory structure in the cytoplasm of thick protrusion. To evaluate the effect of hydrodynamics on proplatelet formation, we evaluated the elongation velocity of proplatelet in sinusoidal lumen by intra-vital imaging. The mean elongating velocity of proplatelet in sinusoidal lumen was 17ƒÊm/min, which was faster than that in static culture condition (0.8ƒÊm/min), implying that sinusoidal flow accelerated the elongation growth of proplatelet. Conclusion: Differed from the previous models based on the live imaging of BM in mice (1), our images captured that the MKs extended and released two types of protrusion into sinusoidal lumens, typical proplatelet without platelet territory and thick protrusion with platelet territory. The structure of thick protrusion with platelet territory suggests a possibility that they have a great advantage for rapid and massive production of circulating platelets in the case of increasing demand for platelets. Proplatelet formation (Figure 1) and thick protrusion (Figure 2) in sinusoid from intra-vital imaging of intact bone marrow in EGFP Tg-mice (megakaryocyte is colored blue and proplatelet is colored green). Disclosures: No relevant conflicts of interest to declare.


Pathogens ◽  
2020 ◽  
Vol 9 (5) ◽  
pp. 372 ◽  
Author(s):  
Jaishree Garhyan ◽  
Surender Mohan ◽  
Vinoth Rajendran ◽  
Rakesh Bhatnagar

One-third of the world’s population is estimated to be latently infected with Mycobacterium tuberculosis (Mtb). Recently, we found that dormant Mtb hides in bone marrow mesenchymal stem cells (BM-MSCs) post-chemotherapy in mice model and in clinical subjects. It is known that residual Mtb post-chemotherapy may be responsible for increased relapse rates. However, strategies for Mtb clearance post-chemotherapy are lacking. In this study, we engineered and formulated novel bone-homing PEGylated liposome nanoparticles (BTL-NPs) which actively targeted the bone microenvironment leading to Mtb clearance. Targeting of BM-resident Mtb was carried out through bone-homing liposomes tagged with alendronate (Ald). BTL characterization using TEM and DLS showed that the size of bone-homing isoniazid (INH) and rifampicin (RIF) BTLs were 100 ± 16.3 nm and 84 ± 18.4 nm, respectively, with the encapsulation efficiency of 69.5% ± 4.2% and 70.6% ± 4.7%. Further characterization of BTLs, displayed by sustained in vitro release patterns, increased in vivo tissue uptake and enhanced internalization of BTLs in RAW cells and CD271+BM-MSCs. The efficacy of isoniazid (INH)- and rifampicin (RIF)-loaded BTLs were shown using a mice model where the relapse rate of the tuberculosis was decreased significantly in targeted versus non-targeted groups. Our findings suggest that BTLs may play an important role in developing a clinical strategy for the clearance of dormant Mtb post-chemotherapy in BM cells.


Sign in / Sign up

Export Citation Format

Share Document