162 Dysregulated NO Signaling in the Lower Genitourinary Tract: A Common Mechanism of Erection and Micturition Dysfunction in a Mouse Model of Sickle Cell Disease

2018 ◽  
Vol 15 (2) ◽  
pp. S47
Author(s):  
B. Musicki ◽  
S. Karakus ◽  
U. Anele ◽  
F. Silva ◽  
A. Burnett
Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 29-30
Author(s):  
Yuanbin Song ◽  
Rana Gbyli ◽  
Liang Shan ◽  
Wei Liu ◽  
Yimeng Gao ◽  
...  

In vivo models of human erythropoiesis with generation of circulating mature human red blood cells (huRBC) have remained elusive, limiting studies of primary human red cell disorders. In our prior study, we have generated the first combined cytokine-liver humanized immunodeficient mouse model (huHepMISTRG-Fah) with fully mature, circulating huRBC when engrafted with human CD34+ hematopoietic stem and progenitor cells (HSPCs)1. Here we present for the first time a humanized mouse model of human sickle cell disease (SCD) which replicates the hallmark pathophysiologic finding of vaso-occlusion in mice engrafted with primary patient-derived SCD HSPCs. SCD is an inherited blood disorder caused by a single point mutation in the beta-globin gene. Murine models of SCD exclusively express human globins in mouse red blood cells in the background of murine globin knockouts2 which exclusively contain murine erythropoiesis and red cells and thus fail to capture the heterogeneity encountered in patients. To determine whether enhanced erythropoiesis and most importantly circulating huRBC in engrafted huHepMISTRG-Fah mice would be sufficient to replicate the pathophysiology of SCD, we engrafted it with adult SCD BM CD34+ cells as well as age-matched control BM CD34+ cells. Overall huCD45+ and erythroid engraftment in BM (Fig. a, b) and PB (Fig. c, d) were similar between control or SCD. Using multispectral imaging flow cytometry, we observed sickling huRBCs (7-11 sickling huRBCs/ 100 huRBCs) in the PB of SCD (Fig. e) but not in control CD34+ (Fig. f) engrafted mice. To determine whether circulating huRBC would result in vaso-occlusion and associated findings in SCD engrafted huHepMISTRG-Fah mice, we evaluated histological sections of lung, liver, spleen, and kidney from control and SCD CD34+ engrafted mice. SCD CD34+ engrafted mice lungs showed an increase in alveolar macrophages (arrowheads) associated with alveolar hemorrhage and thrombosis (arrows) but not observed control engrafted mice (Fig. g). Spleens of SCD engrafted mice showed erythroid precursor expansion, sickled erythrocytes in the sinusoids (arrowheads), and vascular occlusion and thrombosis (arrows) (Fig. h). Liver architecture was disrupted in SCD engrafted mice with RBCs in sinusoids and microvascular thromboses (Fig. i). Congestion of capillary loops and peritubular capillaries and glomeruli engorged with sickled RBCs was evident in kidneys (Fig. j) of SCD but not control CD34+ engrafted mice. SCD is characterized by ineffective erythropoiesis due to structural abnormalities in erythroid precursors3. As a functional structural unit, erythroblastic islands (EBIs) represent a specialized niche for erythropoiesis, where a central macrophage is surrounded by developing erythroblasts of varying differentiation states4. In our study, both SCD (Fig. k) and control (Fig. l) CD34+ engrafted mice exhibited EBIs with huCD169+ huCD14+ central macrophages surrounded by varying stages of huCD235a+ erythroid progenitors, including enucleated huRBCs (arrows). This implies that huHepMISTRG-Fah mice have the capability to generate human EBIs in vivo and thus represent a valuable tool to not only study the effects of mature RBC but also to elucidate mechanisms of ineffective erythropoiesis in SCD and other red cell disorders. In conclusion, we successfully engrafted adult SCD patient BM derived CD34+ cells in huHepMISTRG-Fah mice and detected circulating, sickling huRBCs in the mouse PB. We observed pathological changes in the lung, spleen, liver and kidney, which are comparable to what is seen in the established SCD mouse models and in patients. In addition, huHepMISTRG-Fah mice offer the opportunity to study the role of the central macrophage in human erythropoiesis in health and disease in an immunologically advantageous context. This novel mouse model could therefore serve to open novel avenues for therapeutic advances in SCD. Reference 1. Song Y, Shan L, Gybli R, et. al. In Vivo reconstruction of Human Erythropoiesis with Circulating Mature Human RBCs in Humanized Liver Mistrg Mice. Blood. 2019;134:338. 2. Ryan TM, Ciavatta DJ, Townes TM. Knockout-transgenic mouse model of sickle cell disease. Science. 1997;278(5339):873-876. 3. Blouin MJ, De Paepe ME, Trudel M. Altered hematopoiesis in murine sickle cell disease. Blood. 1999;94(4):1451-1459. 4. Manwani D, Bieker JJ. The erythroblastic island. Curr Top Dev Biol. 2008;82:23-53. Disclosures Xu: Seattle Genetics: Membership on an entity's Board of Directors or advisory committees. Flavell:Zai labs: Consultancy; GSK: Consultancy.


2019 ◽  
Vol 187 (2) ◽  
pp. 246-260
Author(s):  
Katelyn E. Sadler ◽  
Sarah N. Langer ◽  
Anthony D. Menzel ◽  
Francie Moehring ◽  
Ashley N. Erb ◽  
...  

2000 ◽  
Vol 279 (6) ◽  
pp. R1949-R1955 ◽  
Author(s):  
K. A. Nath ◽  
V. Shah ◽  
J. J. Haggard ◽  
A. J. Croatt ◽  
L. A. Smith ◽  
...  

We investigated a transgenic mouse model of sickle cell disease, homozygous for deletion of mouse β-globin and containing transgenes for human βSand βS-antillesglobins linked to the transgene for human α-globin. In these mice, basal cGMP production in aortic rings is increased, whereas relaxation to an endothelium-dependent vasodilator, A-23187, is impaired. In contrast, aortic expression of endothelial nitric oxide synthase (NOS) is unaltered in sickle mice, whereas expression of inducible NOS is not detected in either group; plasma nitrate/nitrite concentrations and NOS activity are similar in both groups. Increased cGMP may reflect the stimulatory effect of peroxides (an activator of guanylate cyclase), because lipid peroxidation is increased in aortae and in plasma in sickle mice. Despite increased vascular cGMP levels in sickle mice, conscious systolic blood pressure is comparable to that of aged-matched controls; sickle mice, however, evince a greater rise in systolic blood pressure in response to nitro-l-arginine methyl ester, an inhibitor of NOS. Systemic concentrations of the vasoconstrictive oxidative product 8-isoprostane are increased in sickle mice. We conclude that vascular responses are altered in this transgenic sickle mouse and are accompanied by increased lipid peroxidation and production of cGMP; we suggest that oxidant-inducible vasoconstrictor systems such as isoprostanes may oppose nitric oxide-dependent and nitric oxide-independent mechanisms of vasodilatation in this transgenic sickle mouse. Destabilization of the vasoactive balance in the sickle vasculature by clinically relevant states may predispose to vasoocclusive disease.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 898-898
Author(s):  
Enrico M Novelli ◽  
Mingyi Yao ◽  
Xiaojun Huang ◽  
Jeffrey Baust ◽  
Hunter Champion ◽  
...  

Abstract Abstract 898 In sickle cell disease (SCD), mutant hemoglobin S polymerizes when deoxygenated, driving red blood cell (RBC)-dependent vaso-occlusion and hemolysis. These processes lead to platelet and hemostatic activation, pulmonary hypertension (PH) and vascular disease. Transgenic-knockout sickle (BERK) mice that express exclusively human α- and βS-globins mimic SCD in humans by displaying reduced nitric oxide (NO) bioavailability, impaired NO-mediated vascular reactivity and PH. Recently, the platelet α-granule protein thrombospondin-1 (TSP1) was found to be elevated in the plasma of patients with SCD and to potently inhibit physiologic NO signaling, via binding to the cell surface receptor CD47. We hypothesized that blocking the TSP1-CD47 interaction may restore NO signaling and prevent PH in BERK mice. To test this hypothesis we conducted a transplantation experiment to explore the repopulating potential of BERK bone marrow (BM) in lethally myeloablated CD47KO recipients and the impact of the CD47 null milieu on the PH phenotype. We harvested the BM from 5–6 months old BERK mice and transplanted it into irradiated (10 Gy) 8–9 weeks old CD47KO mice (n=9). All recipients survived transplantation and were terminally evaluated 4 months post transplantation. Mice underwent blood sampling for determination of engraftment by hemoglobin electrophoresis, evaluation of endothelial dependent arterial vasodilation by myography, full pulmonary hemodynamic assessment and measurement of right ventricular hypertrophy (RVH) using the Fulton Index (ratio of ventricular weights (right ventricle/left ventricle including septum). The chimeras had 98.3% (SD 0.6%) hemoglobin S, thereby demonstrating full donor chimerism. Segments of thoracic aortas from the chimeras were mounted on a myograph system and exposed to acetylcholine, a physiologic vasodilator that stimulates endothelial nitric oxide synthase (eNOS) activation. Concentration-response curves showed that the arterial segments from chimeras that lacked tissue CD47 had improved endothelial-dependent vasodilation, as evaluated by % relaxation in response to acetylcholine, as compared to arterial segments from BERK mice (P < 0.05). Hemodynamic data showed that the tissue CD47KO chimeras had lower right ventricular end systolic pressure (RV ESP) as compared to BERK mice (22 vs. 31 mm Hg, p<0.05). Conversely, their RV ESP did not significantly differ from historical control C57BL/6 mice (22 vs. 20 mm Hg, NS, panel A). Measurement of RVH (Fulton Index) similarly revealed that the chimeras were protected from RVH (p<0.05, panel B). Thus, despite the presence of sickle RBC, the absence of the TSP1-CD47 signaling axis improved endothelial-eNOS-NO signaling and reduced pulmonary pressures and RVH responses. These data demonstrate that BM from BERK mice successfully engrafts CD47KO mice, and that in the absence of the TSP1-CD47 axis endothelial and vascular function improves and PH is ameliorated. We now plan to validate these results in controlled experiments where BM from BERK mice is transplanted in CD47KO and C57BL mice as controls. We expect that unlike C57BL mice transplanted with BERK BM, CD47KO mice will be protected from the vascular complications of SCD, including PH.Figurelegend: CD47KO mice transplanted with BERK BM (chimeras) show improved hemodynamics (Panel A) and less right ventricular (RV) hypertrophy as measured by the Fulton Index as compared to BERK mice (Panel B). * = statistically significant, NS = non significant, RV ESP = right ventricle end systolic pressure.Figure. legend: CD47KO mice transplanted with BERK BM (chimeras) show improved hemodynamics (Panel A) and less right ventricular (RV) hypertrophy as measured by the Fulton Index as compared to BERK mice (Panel B). * = statistically significant, NS = non significant, RV ESP = right ventricle end systolic pressure. Disclosures: Isenberg: Vasculox, Inc.: Equity Ownership.


Blood ◽  
2016 ◽  
Vol 127 (10) ◽  
pp. 1371-1373 ◽  
Author(s):  
Erica M. Sparkenbaugh ◽  
Pichika Chantrathammachart ◽  
Kasemsiri Chandarajoti ◽  
Nigel Mackman ◽  
Nigel S. Key ◽  
...  

2012 ◽  
Vol 181 (5) ◽  
pp. 1725-1734 ◽  
Author(s):  
Steven M. Szczepanek ◽  
Jeffrey T. McNamara ◽  
Eric R. Secor ◽  
Prabitha Natarajan ◽  
Linda A. Guernsey ◽  
...  

Blood ◽  
2014 ◽  
Vol 123 (11) ◽  
pp. 1747-1756 ◽  
Author(s):  
Erica M. Sparkenbaugh ◽  
Pichika Chantrathammachart ◽  
Jacqueline Mickelson ◽  
Joanne van Ryn ◽  
Robert P. Hebbel ◽  
...  

Key Points Inhibition of FXa or thrombin might be considered to reduce thrombotic complications and vascular inflammation in sickle cell patients. PAR-2 could be a potential target to inhibit vascular pathology associated with sickle cell disease.


2013 ◽  
Vol 162 (1) ◽  
pp. 120-129 ◽  
Author(s):  
Hui Wang ◽  
Wei Luo ◽  
Jintao Wang ◽  
Chiao Guo ◽  
Stephanie L. Wolffe ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document