Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Data Brief ; 51: 109784, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38053599

ABSTRACT

A crucial attribute of potted ornamental plants is compactness characterized by well branched plants with rather short stems bearing numerous flowers. To gain plant compactness, producers use plant growth regulators (PGRs), in particular growth retardants during culture. However, due to their negative environmental impacts, growth retardants are progressively withdrawn from the market. As a response, eco-friendly alternative methods to chemicals need to be developed. One method consists in mimicking mechanical stimulation (MS) imposed by wind on plants which causes reduction in stem elongation, an increase in stem diameter and an increase in branching, all contributing to plant compactness. So far, few plant species were studied under MS and little is known on molecular response mechanisms to MS. This first transcriptomic data after MS in Hydrangea macrophylla will contribute unravelling how plants respond to mechanical stimuli. RNAseq data were obtained from total mRNA of stems collected 15 min before MS and 1, 3, 24 and 72 h after MS treatment. RNA from non-MS treated plants were used as control. MS treatment consisted in 12 consecutive bendings (i.e. 6 forth and 6 back) applied at 9 a.m. during 1 h and for a single day. From RNAseq data a de novo assembly of the transcriptome was produced and 78,398 transcripts functionally annotated. These transcriptomic data also contribute to a better knowledge of how outdoor crop respond to the increasing frequency of strong harmful winds under climate change.

3.
Transfusion ; 63(10): 1937-1950, 2023 10.
Article in English | MEDLINE | ID: mdl-37615493

ABSTRACT

BACKGROUND: Pathogen reduction of platelet concentrates (PCs) using amotosalen and broad-spectrum UVA illumination contributes to the safety of platelet transfusion by reducing the risk of transfusion-transmitted infections. We evaluated the in vitro quality of stored buffy-coat (BC) PCs treated with amotosalen and a prototype light-emitting diode (LED) illuminator. METHODS: Double-dose BC-PCs collected into PAS-III/plasma or SSP+ /plasma (55/45%) were treated with amotosalen in combination with either conventional UVA lamps (INT100 Illuminator 320-400 nm) or LED illuminators at 350 nm. Platelet quality and function were evaluated over 7 days. RESULTS: Platelet counts were conserved during storage in all groups, as was platelet swirling without appearance of macroscopic aggregates. Integrin αIIbß3 and glycoprotein (GP) VI expression remained stable, whereas GPIbα and GPV declined similarly in all groups. UV lamp- and LED-treated PCs displayed similar glucose consumption, lactate generation, and pH variation. Comparable spontaneous and residual P-selectin and phosphatidylserine exposure, activated αIIbß3 exposure, mitochondrial membrane potential, lactate dehydrogenase release, and adhesive properties under flow conditions were observed during storage. The use of SSP+ /plasma compared with PAS-III/plasma better preserved most of these parameters, especially during late storage, irrespective of the type of illuminator. CONCLUSION: Replacing the UVA lamp for photochemical treatment by LED illuminators had no impact on platelet metabolism, spontaneous activation, apoptosis or viability, or on the in vitro function of BC-PCs stored for 7 days in SSP+ or PAS-III/plasma. These findings support improved procedures for the pathogen reduction and storage of PCs, to ensure transfusion safety and retention of platelet functional properties.


Subject(s)
Furocoumarins , Ultraviolet Rays , Humans , Furocoumarins/pharmacology , Blood Platelets/metabolism , Platelet Transfusion , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Blood Preservation/methods
4.
Blood Adv ; 7(15): 4003-4018, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37171626

ABSTRACT

Megakaryocytes (MKs) are the precursor cells of platelets, located in the bone marrow (BM). Once mature, they extend elongated projections named proplatelets through sinusoid vessels, emerging from the marrow stroma into the circulating blood. Not all signals from the microenvironment that regulate proplatelet formation are understood, particularly those from the BM biomechanics. We sought to investigate how MKs perceive and adapt to modifications of the stiffness of their environment. Although the BM is one of the softest tissue of the body, its rigidification results from excess fibronectin (FN), and other matrix protein deposition occur upon myelofibrosis. Here, we have shown that mouse MKs are able to detect the stiffness of a FN-coated substrate and adapt their morphology accordingly. Using a polydimethylsiloxane substrate with stiffness varying from physiological to pathological marrow, we found that a stiff matrix favors spreading, intracellular contractility, and FN fibrils assembly at the expense of proplatelet formation. Itgb3, but not Itgb1, is required for stiffness sensing, whereas both integrins are involved in fibrils assembly. In contrast, soft substrates promote proplatelet formation in an Itgb3-dependent manner, consistent with the ex vivo decrease in proplatelet formation and the in vivo decrease in platelet number in Itgb3-deficient mice. Our findings demonstrate the importance of environmental stiffness for MK functions with potential pathophysiological implications during pathologies that deregulate FN deposition and modulate stiffness in the marrow.


Subject(s)
Fibronectins , Megakaryocytes , Animals , Mice , Blood Platelets/metabolism , Bone Marrow , Fibronectins/metabolism , Megakaryocytes/metabolism , Platelet Count
5.
Res Pract Thromb Haemost ; 7(1): 100006, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36970736

ABSTRACT

Background: The platelet population is heterogeneous, with different subsets that differ on the basis of their function and reactivity. An intrinsic factor participating in this difference of reactivity could be the platelet age. The lack of relevant tools allowing a formal identification of young platelets prevents so far to draw solid conclusions regarding platelet reactivity. We recently reported that human leukocyte antigen-I (HLA-I) molecules are more expressed on human young platelets. Objectives: The aim of this study was to assess platelet reactivity according to their age based on HLA-I expression level. Methods: Platelet activation was assessed by flow cytometry (FC) for different platelet subsets based on their HLA-I expression. These populations were further cell sorted and their intrinsic properties were determined by FC and electron microscopy (EM). Statistical analyses were performed with GraphPad Prism 5.02 software using two-way ANOVA followed by a Tukey post hoc test. Results: HLA-I expression level allowed the identification of 3 platelet subpopulations regarding to their age (HLA low, dim, and high). HLA-I was reliable to guide platelet cell sorting and highlighted the features of young platelets in the HLA-Ihigh population. In response to different soluble agonists, HLA-Ihigh platelets were the most reactive subset as shown by the level of P-selectin secretion and fibrinogen binding assessed by flow cytometry. Moreover, the highest capacity of HLA-Ihigh platelets to simultaneously express annexin-V and von Willebrand factor or activated αIIbß3 after coactivation with TRAP and CRP indicated that the procoagulant feature of platelets was age-related. Conclusion: The young HLA-Ihigh population is the most reactive and prone to become procoagulant. These results open up new perspectives to investigate deeply the role of young and old platelets.

6.
Front Plant Sci ; 14: 1268272, 2023.
Article in English | MEDLINE | ID: mdl-38293622

ABSTRACT

Environmental prejudices progressively lead to the ban of dwarfing molecules in agriculture, and alternatives are urgently required. Mechanical stimulation (MS) is a promising, eco-friendly, and economical technique, but some responses to mechanical stimulation vary from one plant species to another. Additionally, as more frequent and violent wind episodes are forecasted under global climate change, knowledge of plant responses to stimuli mimicking wind sways is decisive for agriculture. However, little is known about plant mechanosensitive responses after long-term, recurrent MS. Here, the effects of 3-week, recurrent, symmetrical bendings (1 or 12 per day) in Hydrangea macrophylla stems are examined. Bendings repressed internode elongation and leaf area development, whereas the diametrical growth of the basal internode is increased. Responses were dose-dependent, and no desensitization was observed during the 3 weeks of treatment. MS was almost as efficient as daminozide for plant dwarfing, and it improved stem robustness. Histological and molecular responses to MS were spatially monitored and were concordant with ongoing primary or secondary growth in the internodes. Our molecular data provide the first knowledge on the molecular paths controlled by mechanical loads in Hydrangea and revealed for the first time the involvement of XYP1 in thigmomorphogenetic responses. MS still had a transcriptional impact 48 h after the last bending session, promoting the expression of XYP1, FLA11, and CAD1 while repressing the expression of EXP3 and XTH33 homologs in accordance with xylogenesis, cell wall thickening, and lignin deposition in the xylem of basal internodes. In upper elongating internodes, repression of XYP1, CAD1, SAMS1, and CDC23 homologs is correlated with ongoing primary, even though stunted, growth. For producers, our findings highlight the potential of MS as a sustainable and economical option for controlling plant compactness in Hydrangea and show valuable reinforcement of stem strength.

7.
J Vis Exp ; (171)2021 05 20.
Article in English | MEDLINE | ID: mdl-34096917

ABSTRACT

Bone marrow megakaryocytes are large polyploid cells that ensure the production of blood platelets. They arise from hematopoietic stem cells through megakaryopoiesis. The final stages of this process are complex and classically involve the bipotent Megakaryocyte-Erythrocyte Progenitors (MEP) and the unipotent Megakaryocyte Progenitors (MKp). These populations precede the formation of bona fide megakaryocytes and, as such, their isolation and characterization could allow for the robust and unbiased analysis of megakaryocyte formation. This protocol presents in detail the procedure to collect hematopoietic cells from mouse bone marrow, the enrichment of hematopoietic progenitors through magnetic depletion and finally a cell sorting strategy that yield highly purified MEP and MKp populations. First, bone marrow cells are collected from the femur, the tibia, and also the iliac crest, a bone that contains a high number of hematopoietic progenitors. The use of iliac crest bones drastically increases the total cell number obtained per mouse and thus contributes to a more ethical use of animals. A magnetic lineage depletion was optimized using 450 nm magnetic beads allowing a very efficient cell sorting by flow cytometry. Finally, the protocol presents the labeling and gating strategy for the sorting of the two highly purified megakaryocyte progenitor populations: MEP (Lin-Sca-1-c-Kit+CD16/32-CD150+CD9dim) and MKp (Lin- Sca-1-c-Kit+CD16/32-CD150+CD9bright). This technique is easy to implement and provides enough cellular material to perform i) molecular characterization for a deeper knowledge of their identity and biology, ii) in vitro differentiation assays, that will provide a better understanding of the mechanisms of maturation of megakaryocytes, or iii) in vitro models of interaction with their microenvironment.


Subject(s)
Megakaryocyte Progenitor Cells , Megakaryocytes , Animals , Bone Marrow Cells/cytology , Cell Differentiation/physiology , Cell Separation/methods , Hematopoietic Stem Cells/cytology , Megakaryocyte Progenitor Cells/cytology , Megakaryocytes/cytology , Mice
8.
Methods Mol Biol ; 2002: 13-27, 2019.
Article in English | MEDLINE | ID: mdl-30671734

ABSTRACT

The development, differentiation, and maturation of hematopoietic cells are regulated by the intrinsic and extrinsic regulation. Intrinsic activity is affected by cell autonomous gene expression and extrinsic factors originate from the so-called niche surrounding the hematopoietic cells. It remains unclear why the hematopoietic sites are shifted during embryogenesis. Flow cytometry and immunohistochemistry enable us to study embryonic regulation of hematopoietic niche in the mouse embryo.


Subject(s)
Cell Differentiation , Embryo, Mammalian/cytology , Embryonic Development , Hematopoietic Stem Cells/cytology , Stem Cell Niche/physiology , Animals , Mice
9.
Blood Adv ; 1(21): 1854-1866, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-29296832

ABSTRACT

The fetal liver is the site of a major expansion of the hematopoietic stem cell (HSC) pool and is also a privileged organ to study megakaryocyte progenitor differentiation. We identified in the mouse fetal liver at day 13.5 a discrete stromal cell population harboring a CD45-TER119-CD31-CD51+VCAM-1+PDGFRα- (V+P-) phenotype that lacked colony-forming unit fibroblast activity and harbored an hepatocyte progenitor signature. This previously undescribed V+P- population efficiently supported megakaryocyte production from mouse bone marrow HSC and human peripheral blood HSC-myeloid progenitors cultured in the presence of limited cytokine concentrations. Megakaryocytes obtained in V+P- cocultures were polyploid, positive for CD41/CD42c, and efficiently produced proplatelets. Megakaryocyte production appeared to be mediated by an expansion of the progenitor compartment through HSC-stromal cell contact. In conclusion, the fetal liver contains a unique cellular microenvironment that could represent a platform for the discovery of regulators of megakaryopoiesis.

10.
Plant Physiol ; 172(1): 489-509, 2016 09.
Article in English | MEDLINE | ID: mdl-27462085

ABSTRACT

Bud outgrowth is controlled by environmental and endogenous factors. Through the use of the photosynthesis inhibitor norflurazon and of masking experiments, evidence is given here that light acts mainly as a morphogenic signal in the triggering of bud outgrowth and that initial steps in the light signaling pathway involve cytokinins (CKs). Indeed, in rose (Rosa hybrida), inhibition of bud outgrowth by darkness is suppressed solely by the application of CKs. In contrast, application of sugars has a limited effect. Exposure of plants to white light (WL) induces a rapid (after 3-6 h of WL exposure) up-regulation of CK synthesis (RhIPT3 and RhIPT5), of CK activation (RhLOG8), and of CK putative transporter RhPUP5 genes and to the repression of the CK degradation RhCKX1 gene in the node. This leads to the accumulation of CKs in the node within 6 h and in the bud at 24 h and to the triggering of bud outgrowth. Molecular analysis of genes involved in major mechanisms of bud outgrowth (strigolactone signaling [RwMAX2], metabolism and transport of auxin [RhPIN1, RhYUC1, and RhTAR1], regulation of sugar sink strength [RhVI, RhSUSY, RhSUC2, and RhSWEET10], and cell division and expansion [RhEXP and RhPCNA]) reveal that, when supplied in darkness, CKs up-regulate their expression as rapidly and as intensely as WL Additionally, up-regulation of CKs by WL promotes xylem flux toward the bud, as evidenced by Methylene Blue accumulation in the bud after CK treatment in the dark. Altogether, these results suggest that CKs are initial components of the light signaling pathway that controls the initiation of bud outgrowth.


Subject(s)
Cytokinins/metabolism , Gene Expression Regulation, Developmental/radiation effects , Gene Expression Regulation, Plant/radiation effects , Light , Meristem/genetics , Plant Shoots/genetics , Cytokinins/pharmacology , Darkness , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Plant/drug effects , Meristem/growth & development , Meristem/metabolism , Models, Biological , Plant Growth Regulators/metabolism , Plant Growth Regulators/pharmacology , Plant Proteins/genetics , Plant Proteins/metabolism , Plant Shoots/growth & development , Plant Shoots/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rosa/genetics , Rosa/growth & development , Rosa/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/radiation effects , Time Factors , Xylem/genetics , Xylem/growth & development , Xylem/metabolism
11.
PLoS One ; 11(3): e0151848, 2016.
Article in English | MEDLINE | ID: mdl-27010197

ABSTRACT

Microenvironment and activation signals likely imprint heterogeneity in the lymphatic endothelial cell (LEC) population. Particularly LECs of secondary lymphoid organs are exposed to different cell types and immune stimuli. However, our understanding of the nature of LEC activation signals and their cell source within the secondary lymphoid organ in the steady state remains incomplete. Here we show that integrin alpha 2b (ITGA2b), known to be carried by platelets, megakaryocytes and hematopoietic progenitors, is expressed by a lymph node subset of LECs, residing in medullary, cortical and subcapsular sinuses. In the subcapsular sinus, the floor but not the ceiling layer expresses the integrin, being excluded from ACKR4+ LECs but overlapping with MAdCAM-1 expression. ITGA2b expression increases in response to immunization, raising the possibility that heterogeneous ITGA2b levels reflect variation in exposure to activation signals. We show that alterations of the level of receptor activator of NF-κB ligand (RANKL), by overexpression, neutralization or deletion from stromal marginal reticular cells, affected the proportion of ITGA2b+ LECs. Lymph node LECs but not peripheral LECs express RANK. In addition, we found that lymphotoxin-ß receptor signaling likewise regulated the proportion of ITGA2b+ LECs. These findings demonstrate that stromal reticular cells activate LECs via RANKL and support the action of hematopoietic cell-derived lymphotoxin.


Subject(s)
Endothelial Cells/immunology , Lymph Nodes/cytology , Platelet Membrane Glycoprotein IIb/immunology , RANK Ligand/immunology , Animals , Cells, Cultured , Endothelial Cells/cytology , Fibronectins/immunology , Lymph Nodes/immunology , Lymphotoxin-beta/immunology , Mice, Inbred C57BL , Signal Transduction
12.
Blood ; 127(18): 2231-40, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26966088

ABSTRACT

The mechanisms regulating megakaryopoiesis and platelet production (thrombopoiesis) are still incompletely understood. Identification of a progenitor with enhanced thrombopoietic capacity would be useful to decipher these mechanisms and to improve our capacity to produce platelets in vitro. Differentiation of peripheral blood CD34(+) cells in the presence of bone marrow-human mesenchymal stromal cells (MSCs) enhanced the production of proplatelet-bearing megakaryocytes (MKs) and platelet-like elements. This was accompanied by enrichment in a MK precursor population exhibiting an intermediate level of CD41 positivity while maintaining its expression of CD34. Following sorting and subculture with MSCs, this CD34(+)CD41(low) population was able to efficiently generate proplatelet-bearing MKs and platelet-like particles. Similarly, StemRegenin 1 (SR1), an antagonist of the aryl hydrocarbon receptor (AhR) transcription factor known to maintain CD34 expression of progenitor cells, led to an enriched CD34(+)CD41(low) fraction and to an increased capacity to generate proplatelet-producing MKs and platelet-like elements ultrastructurally and functionally similar to circulating platelets. The effect of MSCs, like that of SR1, appeared to be mediated by an AhR-dependent mechanism because both culture conditions resulted in repression of its downstream effector CYP1B1. This newly described isolation of a precursor exhibiting strong MK potential could be exploited to study normal and abnormal thrombopoiesis and for in vitro platelet production.


Subject(s)
Megakaryocyte Progenitor Cells/cytology , Receptors, Aryl Hydrocarbon/physiology , Thrombopoiesis/physiology , Antigens, CD34/analysis , Blood Platelets/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free , Cytochrome P-450 CYP1B1/physiology , Humans , Immunophenotyping , Platelet Count , Platelet Membrane Glycoprotein IIb/analysis , Purines/pharmacology , Signal Transduction
13.
Stem Cells Dev ; 24(9): 1066-72, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25674667

ABSTRACT

CD34(+) cell dose provides a measure of hematopoietic tissue that predicts the rate of engraftment upon transplant. It is positively correlated with multiple measures of hematopoietic recovery, including platelet engraftment. Here we identify a subpopulation of CD34(+) cells that coexpress a surface antigen--MA6, which is more positively correlated with platelet engraftment in a clinical setting than CD34(+) alone. The specific identity and function of MA6 remain to be determined, however, it is expressed by primitive megakaryocyte (MK) progenitors, but is lost with differentiation and is not expressed by platelets. Commitment of CD34(+)MA6(+) cells to the MK lineage was confirmed by in vitro assays and their significance in hematopoietic transplantation explored by flow cytometric analysis of cryopreserved samples of granulocyte colony stimulating factor-mobilized peripheral blood progenitor cell (PBPC) products along with a retrospective analysis of platelet engraftment data. Platelet engraftment by day 21 was predicted by receipt of ≥ 6 × 10(6) CD34(+) cells/kg or ≥ 0.3 × 10(6) CD34(+)MA6(+) cells/kg. Subsequent analysis of cord blood (CB) CD34(+) cells revealed <0.2% coexpressed MA6(+), compared to 8% of PBPC CD34(+) cells. This low proportion of CD34(+)MA6(+) cells may be responsible, at least in part, for the delayed platelet engraftment associated with CB transplantation. However, platelet engraftment is markedly improved in recipients of ex vivo-expanded CB. This may be a consequence of an increased proportion of CD34(+)MA6(+) cells present in the ex vivo-expanded product and also suggests that optimizing ex vivo culture conditions to generate CD34(+)MA6(+) cells might further improve platelet engraftment in CB recipients.


Subject(s)
Antigens, CD34/metabolism , Blood Platelets/metabolism , Fetal Blood/transplantation , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Platelet Transfusion , Antigens, CD34/genetics , Blood Platelets/cytology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Humans
14.
Exp Hematol ; 40(6): 445-56, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22306295

ABSTRACT

Delayed engraftment remains a major hurdle after cord blood (CB) transplantation. It may be due, at least in part, to low fucosylation of cell surface molecules important for homing to the bone marrow microenvironment. Because fucosylation of specific cell surface ligands is required before effective interaction with selectins expressed by the bone marrow microvasculature can occur, a simple 30-minute ex vivo incubation of CB hematopoietic progenitor cells with fucosyltransferase-VI and its substrate (GDP-fucose) was performed to increase levels of fucosylation. The physiologic impact of CB hematopoietic progenitor cell hypofucosylation was investigated in vivo in NOD-SCID interleukin (IL)-2Rγ(null) (NSG) mice. By isolating fucosylated and nonfucosylated CD34(+) cells from CB, we showed that only fucosylated CD34(+) cells are responsible for engraftment in NSG mice. In addition, because the proportion of CD34(+) cells that are fucosylated in CB is significantly less than in bone marrow and peripheral blood, we hypothesize that these combined observations might explain, at least in part, the delayed engraftment observed after CB transplantation. Because engraftment appears to be correlated with the fucosylation of CD34(+) cells, we hypothesized that increasing the proportion of CD34(+) cells that are fucosylated would improve CB engraftment. Ex vivo treatment with fucosyltransferase-VI significantly increases the levels of CD34(+) fucosylation and, as hypothesized, this was associated with improved engraftment. Ex vivo fucosylation did not alter the biodistribution of engrafting cells or pattern of long-term, multilineage, multi-tissue engraftment. We propose that ex vivo fucosylation will similarly improve the rate and magnitude of engraftment for CB transplant recipients in a clinical setting.


Subject(s)
Fetal Blood/transplantation , Fucose/metabolism , Interleukin Receptor Common gamma Subunit/genetics , Animals , Antigens, CD34/immunology , Bone Marrow Cells/metabolism , Cell Lineage , Fetal Blood/cytology , Fetal Blood/immunology , Flow Cytometry , Humans , Membrane Glycoproteins/physiology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Spleen/cytology , Spleen/metabolism , Transplantation, Heterologous
15.
Blood ; 119(11): e86-95, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22262767

ABSTRACT

The low incidence of CFU-F significantly complicates the isolation of homogeneous populations of mouse bone marrow stromal cells (BMSCs), a common problem being contamination with hematopoietic cells. Taking advantage of burgeoning evidence demonstrating the perivascular location of stromal cell stem/progenitors, we hypothesized that a potential reason for the low yield of mouse BMSCs is the flushing of the marrow used to remove single-cell suspensions and the consequent destruction of the marrow vasculature, which may adversely affect recovery of BMSCs physically associated with the abluminal surface of blood vessels. Herein, we describe a simple methodology based on preparation and enzymatic disaggregation of intact marrow plugs, which yields distinct populations of both stromal and endothelial cells. The recovery of CFU-F obtained by pooling the product of each digestion (1631.8 + 199) reproducibly exceeds that obtained using the standard BM flushing technique (14.32 + 1.9) by at least 2 orders of magnitude (P < .001; N = 8) with an accompanying 113.95-fold enrichment of CFU-F frequency when plated at low oxygen (5%). Purified BMSC populations devoid of hematopoietic contamination are readily obtained by FACS at P0 and from freshly prepared single-cell suspensions. Furthermore, this population demonstrates robust multilineage differentiation using standard in vivo and in vitro bioassays.


Subject(s)
Bone Marrow Cells/cytology , Endothelium, Vascular/cytology , Stem Cells/cytology , Stromal Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Colony-Forming Units Assay , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID
16.
Stem Cell Res ; 5(1): 65-75, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20537607

ABSTRACT

During the course of studies to investigate whether MPC circulate in response to G-CSF, the agent most frequently used to induce mobilization of hematopoietic progenitors, we observed that while G-CSF failed to increase the number of MPC in circulation (assayed in vitro as fibroblast colony-forming cells, CFU-F), G-CSF administration nevertheless resulted in a time-dependent increase in the absolute number of CFU-F within the BM, peaking at Day 7. Treatment of BM cells from G-CSF-treated mice with hydroxyurea did not alter CFU-F numbers, suggesting that the increase in their numbers in response to G-CSF administration is not due to proliferation of existing CFU-F. Given previous studies demonstrating that G-CSF potently induces bone turnover in mice, we hypothesized that the increase in CFU-F may be triggered by the bone resorption that occurs following G-CSF administration. In accord with this hypothesis, administration of an inhibitor of osteoclast differentiation, osteoprotegerin (OPG), prevented the increase of CFU-F numbers induced by G-CSF. In conclusion, these data indicate that the cytokine treatment routinely used to mobilize hematopoietic stem cells could provide a readily applicable method to induce in vivo expansion of MPC for clinical applications.


Subject(s)
Bone Marrow/drug effects , Bone Resorption/pathology , Granulocyte Colony-Stimulating Factor/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteoclasts/drug effects , Animals , Cell Adhesion/drug effects , Cell Movement , Cell Proliferation/drug effects , Male , Mice , Mice, Inbred BALB C , Osteogenesis/drug effects
17.
J Clin Invest ; 119(9): 2795-806, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19652362

ABSTRACT

The cellular and molecular microenvironment of epithelial stem and progenitor cells is poorly characterized despite well-documented roles in homeostatic tissue renewal, wound healing, and cancer progression. Here, we demonstrate that, in organotypic cocultures, dermal pericytes substantially enhanced the intrinsically low tissue-regenerative capacity of human epidermal cells that have committed to differentiate and that this enhancement was independent of angiogenesis. We used microarray analysis to identify genes expressed by human dermal pericytes that could potentially promote epidermal regeneration. Using this approach, we identified as a candidate the gene LAMA5, which encodes laminin alpha5, a subunit of the ECM component laminin-511/521 (LM-511/521). LAMA5 was of particular interest as we had previously shown that it promotes skin regeneration both in vitro and in vivo. Analysis using immunogold localization revealed that pericytes synthesized and secreted LAMA5 in human skin. Consistent with this observation, coculture with pericytes enhanced LM-511/521 deposition in the dermal-epidermal junction of organotypic cultures. We further showed that skin pericytes could also act as mesenchymal stem cells, exhibiting the capacity to differentiate into bone, fat, and cartilage lineages in vitro. This study suggests that pericytes represent a potent stem cell population in the skin that is capable of modifying the ECM microenvironment and promoting epidermal tissue renewal from non-stem cells, a previously unsuspected role for pericytes.


Subject(s)
Pericytes/physiology , Regeneration/physiology , Skin Physiological Phenomena , Base Sequence , Cell Differentiation , Cells, Cultured , Coculture Techniques , Epidermal Cells , Epidermis/metabolism , Gene Expression , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Laminin/genetics , Laminin/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Oligonucleotide Array Sequence Analysis , Pericytes/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regeneration/genetics
18.
Stem Cell Res ; 3(2-3): 126-41, 2009.
Article in English | MEDLINE | ID: mdl-19700382

ABSTRACT

WNT and bone morphogenetic protein (BMP) signaling are known to stimulate hemogenesis from pluripotent embryonic stem (ES) cells. However, osteochondrogenic mesoderm was generated effectively when BMP signaling is kept to a low level, while WNT signaling was strongly activated. When mesoderm specification from ES cells was exogenous factor dependent, WNT3a addition supported the generation of cardiomyogenic cells expressing lateral plate/extraembryonic mesoderm genes, and this process involved endogenous BMP activities. Exogenous BMP4 showed a similar effect that depended on endogenous WNT activities. However, neither factor induced robust chondrogenic activity. In support, ES cell differentiation in the presence of either WNT3a or BMP4 was associated with elevated levels of both Bmp and Wnt mRNAs, which appeared to provide sufficient levels of active BMPs and WNTs to promote the nonchondrogenic mesoderm specification. The osteochondrogenic mesoderm expressed PDGFRalpha, which also expressed genes that mark somite and rostral presomitic mesoderm. A strong WNT signaling was required for generating the mesodermal progeny, while approximately 50- to 100-fold lower concentration of WNT3a was sufficient for specifying axial mes(end)oderm. Thus, depending on the dose and cofactor (BMP), WNT signaling stimulates the generation of different biological activities and specification of different types of mesodermal progeny from ES cells.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Embryonic Stem Cells/cytology , Mesoderm/cytology , Wnt Proteins/metabolism , Antigens, CD/metabolism , Bone Morphogenetic Protein 4/pharmacology , Cadherins/metabolism , Embryonic Stem Cells/metabolism , Humans , Mesoderm/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wnt Proteins/pharmacology , Wnt3 Protein , Wnt3A Protein
19.
Stem Cells ; 27(8): 1932-40, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19544439

ABSTRACT

The use of umbilical cord blood (UCB) grafts for hematopoietic stem cell transplantation (HSCT) is a promising technique that permits a degree of human leukocyte antigen mismatch between the graft and the host without the concomitant higher rate of graft-versus-host disease that would be observed between an adult marrow graft and a mismatched host. A disadvantage to the use of UCB for HSCT is that immune reconstitution may be significantly delayed because of the low stem cell dose available in the graft. Ex vivo expansion of UCB CD34 cells would provide a greater number of stem cells; however, there are persistent concerns that ex vivo-expanded CD34 cells may lose pluripotency and the ability to contribute meaningfully to long-term engraftment. To address this issue, we transduced CD34-selected UCB cells with a lentiviral construct expressing luciferase, and determined homing and engraftment patterns in vivo by noninvasive bioluminescent imaging in sublethally irradiated NOD/SCID/IL-2Rgamma(-/-) (NSG) mice. Graft contribution to multilineage commitment was also confirmed by analysis of primary and secondary transplants by flow cytometry and immunohistochemistry. Our results demonstrate that, other than a mild delay at the onset of engraftment, there were no significant differences in lineage repopulation or in long-term or secondary engraftment between culture-expanded and unexpanded UCB CD34-selected cells. The results suggest that multipotent stem cells can be expanded ex vivo and can contribute meaningfully to long-term hematopoietic engraftment.


Subject(s)
Antigens, CD34/analysis , Cord Blood Stem Cell Transplantation/methods , Fetal Blood/cytology , Mesenchymal Stem Cells/cytology , Animals , Cell Lineage , Flow Cytometry , Humans , Immunohistochemistry , Luciferases, Firefly/chemistry , Luminescent Agents/chemistry , Luminescent Measurements/methods , Mice , Mice, Inbred NOD , Mice, SCID , Transduction, Genetic
20.
Methods Mol Biol ; 482: 259-68, 2009.
Article in English | MEDLINE | ID: mdl-19089361

ABSTRACT

Bone marrow from numerous species, including rodents and man, has been shown to contain a rare population of cells known as marrow stromal cells or mesenchymal stem cells (MSC). Given the innate ability of these cells to give rise to multiple tissue types including bone, fat and cartilage, there is considerable interest in utilizing MSC in a broad repertoire of cell-based therapies for the treatment of human disease. In order for such therapies to be realized, a preclinical animal model in which to refine strategies utilizing MSC is required.We have described methodology allowing for the prospective isolation by fluorescence activated cell sorting (FACS) of a highly purified population of MSC from murine compact bone (CB). These cells are multipotent and capable of extensive proliferation in vitro and thus represent an ideal source of cells with which to explore both the fundamental biology of MSC and their efficacy in a variety of cellular therapies.


Subject(s)
Bone and Bones/cytology , Cell Separation/methods , Mesenchymal Stem Cells/cytology , Animals , Cell Lineage , Cell Shape , Cells, Cultured , Flow Cytometry , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...