scholarly journals Studies of the Hemopoietic Microenvironment. II. Effect of Erythropoietin on the Splenic Microvasculature of Polycythemic CF1 Mice

Blood ◽  
1972 ◽  
Vol 39 (6) ◽  
pp. 809-813 ◽  
Author(s):  
Robert S. McCuskey ◽  
Howard A. Meineke ◽  
Stephen M. Kaplan

Abstract The effect of erythropoietin on the splenic microvascular system of polycythemic CF1 mice was studied using in vivo microscopic methods. Administration of a single dose (3 U) of erythropoietin resulted in an increase in the linear velocity of blood flow through the splenic sinusoids and a reduction in the number of sinusoids storing blood. This response was first seen 4-6 hr after injection; it persisted for 48 hr and was reduced markedly by 72 hr. By 120 hr the spleens were indistinguishable from controls. The response was specific for erythrogenic tissue, since no response was seen in the adjacent nonerythropoietic pancreatic tissue. The results suggest that the splenic microvascular response to erythropoietin may be indirect and may be mediated by the release of a vasoactive metabolite from the erythrogenic tissues surrounding the sinusoids. Erythropoietin-sensitive stem cells are suggested to be the source of such a metabolite.

Blood ◽  
1972 ◽  
Vol 39 (5) ◽  
pp. 697-712 ◽  
Author(s):  
Robert S. McCuskey ◽  
Howard A. Meineke ◽  
Samuel F. Townsend

Abstract Specific alterations in the microvascular and connective tissue compartments of the hemopoietic microenvironment have been examined during erythropoietic regeneration and suppression in the murine spleen and bone marrow using in vivo microscopic and histochemical methods. The results have confirmed the concept of specific hemopoietic microenvironments and have demonstrated specific alterations in the microenvironment during erythropoietic stimulation and repression. Elevated erythropoiesis in the splenic red pulp is accompanied by an elevation in blood flow through the microvascular system. Both the linear velocity of flow and the number of sinusoids with blood flow in them increased significantly. In contrast, erythropoietic repression was accompanied by a decreased linear velocity of blood flow, as well as a marked increase in the amount of blood being stored in the splenic sinusoids. This also was the picture when diffuse granulopoiesis was present in the red pulp, or when granuloid or undifferentiated colonies were present. The chemical composition of the stroma in the spleen and bone marrow also varied during states of hemopoietic activity and, in addition, there were differences in the composition of the stroma between these two organs. In both organs, foci of early proliferating cells were enveloped by a coating of sulfated acid mucopolysaccharide. This coat persisted on cells in later stages of granulopoiesis but not on cells in the later stages of erythropoiesis. The latter were enveloped with a coating of neutral mucopolysaccharide. A tentative hypothesis to explain the mechanisms involved in producing these changes is discussed.


1993 ◽  
Vol 73 (4) ◽  
pp. 855-868 ◽  
Author(s):  
J. M. Kelly ◽  
B. G. Southorn ◽  
C. E. Kelly ◽  
L. P. Milligan ◽  
B. W. McBride

The effect of level of nutrition on in vitro and in vivo O2 consumption by the gastrointestinal tract in four nonlactating, nonpregnant ewes catheterized in the anterior mesenteric vein, hepatic portal vein and mesenteric artery with duodenal cannulae was investigated. Animals were fed a pelleted ration at maintenance (M) or twice maintenance (2M) or fasted (F) subsequent to the M measurement. Duodenal in vitro O2, ouabain-sensitive O2 (OSO2) and cycloheximide-sensitive O2 (CSO2) consumption was determined polarographically using a YSI O2 monitor; whole-gut O2 consumption was determined as (arterio-venous difference of O2 concentration) × (blood flow through the PV). Whole-body O2 consumption was determined using indirect calorimetry. Ewes fed 2M exhibited higher (P < 0.10) whole-body O2 consumption than either M or F ewes. Ewes fed M and 2M had higher (P < 0.10) duodenal in vitro O2 and ouabain-insensitive O2 (OIO2) consumption than F ewes. Hepatic portal blood flow was directly proportional to level of intake (P < 0.10): it was lowest for F ewes (81.0 L h−1), intermediate for M ewes (97.7 L h−1) and highest for 2M ewes (122.5 L h−1). Ouabain inhibition of O2 consumption by portal-drained viscera (PDV) was highest in M ewes and lowest in 2M ewes (P < 0.10). CSO2 consumption by the entire PDV was not affected by level of intake, corresponding to no change in OIO2 consumption by the PDV. As a proportion of whole-body O2 consumption, total O2, OSO2 and cycloheximide-insensitive O2 consumption by the PDV was higher in F ewes than in 2M ewes (P < 0.10). Fasted ewes expended a greater proportion of whole-body O2 consumption on gastrointestinal energetics than did 2M ewes. Key words: Sheep, gastrointestinal oxygen consumption, sodium–potassium ATPase, protein synthesis


Author(s):  
Dooyoung Lee ◽  
Kuldeepsinh Rana ◽  
Karin Lee ◽  
Lisa A. DeLouise ◽  
Michael R. King

In previous work, we have described the adhesive capture of circulating stem cells to surfaces coated with adhesive selectin protein, both in vitro and in vivo. Here we describe PDMS surfaces microfabricated to contain an array of square 80 × 80 × 80 micron cavities. These cavities are intended to provide a local bioreactor environment to culture stem cells over extended periods of time, while sheltered from the higher shear stresses of the surrounding blood flow external of the cavities. In this paper we present in vitro flow experiments with polymeric, blood cell-sized microspheres, showing the creation of stable vortices within the microscale cavities. Computational fluid dynamics (CFD) was performed to predict the velocity field within the cavity, and for comparison with experimentally determined microsphere velocities. Future work will establish the ability to place local chemoattract molecules within the cavity interior, and the ability to accumulate viable stem cells within these cavities.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1790-1790
Author(s):  
M.R. Finney ◽  
L.R. Fanning ◽  
P.J. Vincent ◽  
D.G. Winter ◽  
M.A. Hoffman ◽  
...  

Abstract Recent reports have utilized a variety of cell types for cellular therapy in mediating therapeutic angiogenesis in response to ischemia. We sought to assess the vasculogeneic potential of selected CD133+ hematopoietic stem cells (HSC) from umbilical cord blood (UCB) utilizing in vitro functional assays and an in vivo murine hind-limb ischemia model. Methods & Results: Mononuclear cells (MNC) from UCB or bone marrow (BM) were incubated with CD133+ conjugated magnetic beads, followed by automated sorting through magnetic columns (Miltenyi). Routine yield of CD133+ cells was 0.5±0.2% of UCB MNC and 0.7±0.3% of BM MNC, with a purity of 79±2% (UCB, n=30) and 84±5% (BM, n=12). Surface expression in the UCB CD133+ population was 3.6±1.5% KDR(VEGFR2), 8.7± 3.8% CXCR4 and 22.7±2.8% CD105 compared to 9.2±1.8% KDR, 14.4±1.3% CXCR4 and 23.7±2.3% CD105 in the BM CD133+ population. We measured chemotactic migration of cells towards SDF-1 (100ng/mL) compared to control wells containing media alone. The fold increase over control was 4.9±2.9 UCB MNC, 1.8±0.7 UCB CD133+ and 8.3±1.7 BM CD133+ (n=3). Angiogenic protein assays of CD133+ cells demonstrated elevated levels of IL-8 production as compared to MNC (103+/−380 pg/mL greater in CD133+ than MNC from the same UCB unit) when cultured for 24h in basal media. NOD/SCID mice underwent ligation of the right femoral artery and were given cells or vehicle control via intracardiac injection immediately following injury. Mice were given 1 x 106 MNC or 0.5 x 106 CD133+ cells. Laser Doppler flow measurements were obtained from both limbs each week for 6 weeks and the ratio of perfusion in the ischemic/healthy limb was calculated. At 28 days, perfusion ratios were statistically higher in study groups receiving UCB CD133+ cells, 0.55±0.06 (n=9), BM CD133+ cells 0.47±0.07 (n=8), BM MNC 0.48±0.8 (n=6) compared to cytokine controls 0.37±0.03 (n=12, p<0.05). Mice receiving UCB MNC did not show statistically significant improvement in measured blood flow over control animals 0.42±0.05 (n=8, p=0.34). At sacrifice, bone marrow was harvested to assess engraftment of human cells by flow cytometric analysis. Mice injected with UCB CD133+ cells showed 19±4.9% positive huCD45 cells compared to 2.5±0.6% for UCB MNC, 1.6±0.4% for BM CD133+ cells and 2.3±0.3% for BM MNC (n=3). Histological studies from day 42 tissue samples of muscle distal to arterial ligation were evaluated for capillary density. Control animals had capillary density of 131±6.9 cells/mm2. Capillary density was statistically higher that controls in animals receiving UCB CD133+ (320±18; p<0.0001), BM CD133++ (183±9.3; p<0.0001), and UCB MNC (164±10.5; p=0.011). Mice treated with BM MNC (135±9.4) did not have a statistically significant increase in capillary density from controls (p=0.73). In addition, animals treated with either UCB or BM-derived CD133+ cells had statistically higher capillary density than unselected MNC (p=<0.0001 and p=0.0004, respectively). Conclusions: In vitro functional assays showed that UCB-derived CD133+ HSC demonstrate enhanced homing capability (migration) as well as the potential for cellular recruitment (via IL-8 production) for angiogenesis in response to ischemia. Furthermore, UCB derived CD133+ HSC mediate significantly improved blood flow in an in vivo murine hind-limb injury model of ischemia, indicating the greater vasculogenic potential of selected CD133+ cells from of this stem cell source.


2021 ◽  
Vol 7 (30) ◽  
pp. eabh0101
Author(s):  
Thomas A. Longden ◽  
Amreen Mughal ◽  
Grant W. Hennig ◽  
Osama F. Harraz ◽  
Bo Shui ◽  
...  

Healthy brain function depends on the finely tuned spatial and temporal delivery of blood-borne nutrients to active neurons via the vast, dense capillary network. Here, using in vivo imaging in anesthetized mice, we reveal that brain capillary endothelial cells control blood flow through a hierarchy of IP3 receptor–mediated Ca2+ events, ranging from small, subsecond protoevents, reflecting Ca2+ release through a small number of channels, to high-amplitude, sustained (up to ~1 min) compound events mediated by large clusters of channels. These frequent (~5000 events/s per microliter of cortex) Ca2+ signals are driven by neuronal activity, which engages Gq protein–coupled receptor signaling, and are enhanced by Ca2+ entry through TRPV4 channels. The resulting Ca2+-dependent synthesis of nitric oxide increases local blood flow selectively through affected capillary branches, providing a mechanism for high-resolution control of blood flow to small clusters of neurons.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3568-3568
Author(s):  
Claus Sondergaard ◽  
Fernando Antonio Fierro ◽  
Jeannine McGee ◽  
Jan A. Nolta

Abstract Abstract 3568 Poster Board III-505 Both mesenchymal stem cells (MSC) and platelet derived growth factor beta (PDGFB) have been shown to promote angiogenesis in vivo and are therefore interesting therapeutic agents for the restoration of blood flow to ischemic tissues encountered in peripheral artery disease and critical limb ischemia. Careful evaluation of the mechanisms responsible for neovessel formation in stem cell-based therapeutic angiogenesis is critical for developing efficient treatment strategies. Previous studies have shown that MSCs correspond to pericytes, which are lost in PDGFB-null mice, suggesting that MSCs strongly rely on PDGFB signaling in vivo. Our major aim was to establish a highly sensitive method to assess whether overexpression of PDGFB in MSCs could modulate expression of pro-angiogenic cytokines and improve the restoration of bloodflow in an established murine xenograft model of hind limb ischemia. Human bone marrow derived MSCs (passage 2-5) were transduced with either a lentiviral vector containing the coding sequence of PDGFB or the same vector without PDGFB (control). As expected, overexpression of PDGFB strongly enhanced the proliferation of MSCs, reducing the doubling time from 51 to 20 hours (n=4, p<0.05). This effect correlated with a shift of 15% of cells from the G0/G1 to the S/G2/M phase, as compared to controls, when measured by flow cytometry of propidium iodide stained cells. Next, we induced a unilateral hind limb ischemia in NODSCIDbeta2null mice by excision of the femoral artery and performed intravenous transplantation of 2.5×10e5 MSCs (control MSC vs. PDGFB-MSC) the day following surgery. Recovery of blood flow to the affected limb was assessed at days 0 ((before transplantation), n=11), 4 (n=11), 7 (n=9), 10 (n=6), and 14 (n=6) by laser doppler imaging and values were normalized to the non-ischemic contralateral leg. Treated animals of both groups (control and PDGFB overexpressing MSC) exhibited a steadily progressive recovery in blood flow in the region proximal to the primary ligation site, reaching near normal levels by day 10. Surprisingly, no significant difference was seen in the blood flow recovery in animals receiving the PDGF over expressing MSCs. Donor MSCs (control and PDGFB) homed to the site of injury, as assessed by immunofluorescent staining of the ischemic muscle in the regions proximal and distal to the primary ligation site at days 4 and 7. However, the rapid recovery in blood flow did not appear to be attributed to a direct differentiation or integration of MSC into the vasculature, but rather through the expression of paracrine angiogenic factors. Interestingly, overexpression of PDGF reduced FGF2 mRNA expression levels in MSC (n=4, p=0.056), suggesting that high levels of PDGFB may indeed lead to a reduction of other key angiogenic factors, explaining the mild effect of PDGFB in vivo. Our results show that, although overexpression of PDGFB in bone marrow-derived MSCs enhanced the proliferation of MSC in vitro, it did not impact the speed or magnitude of blood flow recovery to the ischemic hind limb as compared to native MSCs, likely due to down modulation of other key angiogenic signals including FGF2. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 728-728 ◽  
Author(s):  
Trista E. North ◽  
Wolfram Goessling ◽  
Marian Peeters ◽  
Pulin Li ◽  
Allegra M. Lord ◽  
...  

Abstract During vertebrate embryogenesis, definitive hematopoietic stem cells (HSC) arise in the aorta-gonads-mesonephros (AGM). Based on the functional conservation of AGM hematopoiesis from fish to man, an evolutionary advantage for the production of stem cells within the aorta must exist. The identification of the signals that induce HSCs at this developmental stage is of significant interest. Through a chemical genetic screen in zebrafish, a diverse group of compounds that regulate blood flow were found to affect the production of runx1/cmyb+ HSCs. These compounds represented modulators of the adrenergic and renin/angiotensin pathways, and Ca+, Na+ and nitric oxide (NO) signaling. In general, we determined that compounds which increased blood flow enhanced HSC number, whereas chemicals that decreased blood flow diminished runx1/cmyb expression. The conserved physiological mechanism of action of each compound on the vasculature was confirmed in vivo by confocal microscopy of transgenic fli1:GFP reporter fish. In the zebrafish, the step-wise initiation of heartbeat, establishment of vigorous circulation and onset of definitive hematopoiesis in the aorta-gonad-mesonephros region (AGM) suggests that blood flow may trigger HSC formation. silent heart (sih) embryos that lack a heartbeat and fail to establish blood circulation exhibit severely reduced numbers of runx1+ HSCs in the AGM. Blood flow modifying agents primarily exerted their effects after the onset of the heartbeat (>24 hpf), however, only compounds that increase NO production (L-Arginine, S-nitroso-N-acetyl-penicillamine (SNAP)) could induce HSC formation prior to the initiation of circulation (5 somites to 22 hpf). Furthermore, SNAP rescued HSC production in sih mutant zebrafish, whereas other drugs that increased blood flow could not. Treatment with the NO synthase (NOS) inhibitor, N-nitro-L-arginine methyl ester (L-NAME), and morpholino-oligonucleotide (MO)-knockdown of nos1 (nnos/enos) blocked HSC development. Additionally, modulation of downstream components of the NO pathway affected HSC production in the zebrafish embryo. Together these data indicate that NO signaling is the downstream effector of blood flow on AGM HSC induction. To document that NO-mediated regulation of HSC formation was conserved across vertebrate species, we examined definitive HSC production in the murine AGM. Nos3 (eNos) was found to be expressed in the AGM endothelium and aortic hematopoietic clusters. Additionally, Nos3 expression specifically marks the population of HSCs with long-term adult bone marrow repopulating activity. Intrauterine NOS inhibition with L-NAME resulted in a lack of hematopoietic clusters in the AGM and a failure to generate transplantable hematopoietic progenitors. Our work provides a direct link between the initiation of circulation and the onset of AGM hematopoiesis, and identifies NO signaling as a conserved downstream regulator of HSC development. ^TEN and WG contributed equally to this work


1990 ◽  
Vol 18 (4) ◽  
pp. 283-292 ◽  
Author(s):  
N.P. Rowell ◽  
M.A. Flowers ◽  
V.R. McCready ◽  
B. Cronin ◽  
A. Horwich

Sign in / Sign up

Export Citation Format

Share Document