scholarly journals Altered Erythropoiesis in Mouse Models of Type 3 Hemochromatosis

2017 ◽  
Vol 2017 ◽  
pp. 1-12 ◽  
Author(s):  
R. M. Pellegrino ◽  
F. Riondato ◽  
L. Ferbo ◽  
M. Boero ◽  
A. Palmieri ◽  
...  

Type 3 haemochromatosis (HFE3) is a rare genetic iron overload disease which ultimately lead to compromised organs functioning. HFE3 is caused by mutations in transferrin receptor 2 (TFR2) gene that codes for two main isoforms (Tfr2αand Tfr2β). Tfr2αis one of the hepatic regulators of iron inhibitor hepcidin. Tfr2βis an intracellular isoform of the protein involved in the regulation of iron levels in reticuloendothelial cells. It has been recently demonstrated that Tfr2 is also involved in erythropoiesis. This study aims to further investigate Tfr2 erythropoietic role by evaluating the erythropoiesis of two Tfr2 murine models wherein either one or both of Tfr2 isoforms have been selectively silenced (Tfr2 KI and Tfr2 KO). The evaluations were performed in bone marrow and spleen, in 14 days’ and 10 weeks’ old mice, to assess erythropoiesis in young versus adult animals. The lack of Tfr2αleads to macrocytosis with low reticulocyte number and increased hemoglobin values, together with an anticipation of adult BM erythropoiesis and an increased splenic erythropoiesis. On the other hand, lack of Tfr2β(Tfr2 KI mice) causes an increased and immature splenic erythropoiesis. Taken together, these data confirm the role of Tfr2αin modulation of erythropoiesis and of Tfr2βin favoring iron availability for erythropoiesis.

2013 ◽  
Vol 26 (2) ◽  
pp. 132
Author(s):  
RedaI Rakha ◽  
SamarM.K. El-Din Fathallah ◽  
FathiaM El-Nemr ◽  
FaridaH El-Rashidi ◽  
SehamM Ragab

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. SCI-1-SCI-1
Author(s):  
Laura Silvestri ◽  
Alessia Pagani ◽  
Antonella Nai ◽  
Clara Camaschella

Abstract Iron, an essential element in mammals, is absorbed by duodenal enterocytes, enters the circulation through the iron exporter ferroportin, (FPN), circulates bound to transferrin and is uptaken through Transferrin Receptor 1. If in excess, iron is stored in macrophages and hepatocytes and released when needed. To maintain systemic iron homeostasis and to avoid the formation of "non transferrin bound iron" (NTBI), a highly reactive form which causes organ damage, the liver synthetizes hepcidin that, binding FPN, blocks iron export to the circulation. Hepcidin integrates signals from body iron, erythropoiesis and inflammatory cytokines. Defective hepcidin production causes iron overload and organ failure in Hereditary Hemochromatosis and Thalassemia; hepcidin excess leads to anemia in Iron Refractory iron Deficiency Anemia (IRIDA) and Anemia of Inflammation (AI). In hepatocytes hepcidin is under the control of the BMP-SMAD pathway, which is activated in a paracrine manner by BMP2 and BMP6 produced by liver sinusoidal endothelial cells. BMP2 maintains hepcidin basal levels, while BMP6 controls its expression in response to iron. The two ligands have different affinity for BMP type I receptors ALK2 and ALK3, suggesting two distinct branches of the hepcidin activation pathway. This possibility is consistent with the non-redundant function of BMP2 and BMP6, the different iron phenotype of hepatocyte-conditional ALK2 and ALK3 KO mice and the residual ability of BMP6 to activate hepcidin in hemochromatosis mice. Moreover ALK2, but not ALK3, is inhibited by the immunophilin FKBP12 in the absence of ligands. The BMP pathway activation depends upon the coreceptor hemojuvelin (HJV), the MHC class I protein HFE and the second transferrin receptor (TFR2). Mutations of all these proteins lead to decreased hepcidin expression in hemochromatosis. Hepcidin expression is inhibited in iron deficiency, hypoxia and when erythropoiesis is increased. Inhibitors are the liver transmembrane serine protease TMPRSS6, whose genetic inactivation causes IRIDA, and the erythroid hormone erythroferrone (ERFE), which is released by erythropoietin-stimulated erythroblasts. The mechanism of hepcidin inhibition by ERFE is unclear; still to allow ERFE function the BMP-SMAD pathway has not to be hyperactive. Intriguingly, both iron deficiency and erythropoiesis require epigenetic modifications at the hepcidin locus with HDAC3-dependent reversible loss of H3K9ac and H3K4me3. Hepcidin also acts as an antimicrobial peptide since its expression, increased by proinflammatory cytokines, such as IL6 through JAK2-STAT3 signaling, restricts iron availability for microbial growth. This first-line of defense against infections negatively influences erythropoiesis since chronic hepcidin activation causes AI. Despite persistent JAK2-STAT3 activation, inhibition of the BMP-SMAD pathway reduces hepcidin activation in AI experimental rodent models, suggesting that hepcidin activation in inflammation requires a functional BMP-SMAD pathway. Independently from hepcidin, inflammation also reduces FPN mRNA levels, favoring macrophage iron sequestration. The identification of hepcidin-ferroportin axis molecular players has translational implications. In primary and secondary iron overload hepcidin agonists (hepcidin peptides or mimics, agents that inhibit the hepcidin inhibitor TMPRSS6 and likely the ALK2-inhibitor FKBP12) and ferroportin inhibitors are potentially useful to prevent iron overload and/or to favor iron redistribution to macrophages. In case of AI, hepcidin antagonists (including anti-hepcidin, anti-HJV and anti-BMP6 monoclonal antibodies, L-enantiomeric oligonucleotides targeting hepcidin, siRNA against hepcidin, non-anticoagulant heparins, the ALK2 inhibitor momelotinib) might improve erythropoiesis increasing iron availability. The effect of some agents that have now entered the clinical phase will become apparent in the coming years. Disclosures Camaschella: vifor Pharma: Honoraria, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. SCI-37-SCI-37
Author(s):  
Elizabeta Nemeth ◽  
Tomas Ganz ◽  
Léon Kautz

For successful expansion of erythropoiesis, the activity of the hormone erythropoietin (EPO) must be coordinated with the supply of iron to erythroid precursors. Increased iron supply for erythropoiesis is ensured by the suppression of hepcidin, the iron-regulatory hormone produced by the liver. Low hepcidin levels allow greater absorption of dietary iron and greater mobilization of iron from the stores in the spleen and the liver. The mechanisms coordinating erythropoietic activity with iron delivery are not well understood. We recently identified erythroferrone as a new mediator of hepcidin suppression during stress erythropoiesis1. Erythroferrone (ERFE) is a member of the C1q/TNF-related protein (CTRP) family of metabolic mediators. ERFE is produced in response to EPO by erythroblasts of the bone marrow and spleen of mice. The induction of ERFE by EPO was dependent on Jak2/Stat5 signaling. Ex vivo treatment of human erythroblasts with EPO also resulted in a dramatic induction of ERFE expression. The essential role of ERFE in acute hepcidin suppression by erythropoiesis was demonstrated in ERFE-deficient mice. In contrast to wild-type mice which suppressed hepcidin ~10-fold within hours after hemorrhage or erythropoietin injection, no hepcidin suppression was observed in ERFE knockout mice within 24 h. As a consequence, ERFE-deficient mice exhibited delayed recovery of hemoglobin after hemorrhage or severe inflammation. Treatment of mice or hepatocytes with recombinant ERFE protein confirmed the hepcidin-suppressive activity of the protein. It remains to be seen whether administration of ERFE protein would be useful for the treatment of anemia of inflammation mediated by elevated hepcidin. In iron-loading anemias including β-thalassemia, hepcidin is chronically suppressed by the exuberant but ineffective erythropoietic activity. This is the cause of iron overload in untransfused thalassemia patients and may contribute to iron loading even in transfused patients. We found that ERFE expression is greatly increased in the bone marrow and spleen of mice with β-thalassemia intermedia (th3 model). Transgenic ablation of ERFE in th3 mice normalized hepcidin and partially corrected their iron overload. Although human studies of the role of ERFE in health and disease are clearly needed, ERFE is a promising candidate for the pathological suppressor of hepcidin in anemias with ineffective erythropoiesis. References: 1. Kautz L, Jung G, Valore EV, et al. Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat Genet. 2014; 46: 678-684. Disclosures Nemeth: Intrinsic LifeSciences: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Merganser Biotech: Equity Ownership. Ganz:Intrinsic LifeSciences: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Keryx Pharma: Consultancy; Merganser Biotech: Consultancy, Equity Ownership.


Author(s):  
Pooja Sharma ◽  
Anshu Palta ◽  
Anita Tahlan ◽  
Manveen Kaur ◽  
Ram Singh

Abstract Objectives Hypocellular bone marrow (BM) disorders comprise heterogeneous entities associated with peripheral cytopenias and decreased production of hematopoietic cells in BM. This study was undertaken to analyze immunohistochemical expression of CD34, CD117, and p53 in morphologically diagnosed patients of hypocellular BM (aplastic anemia [AA], hypocellular myelodysplastic syndrome [h-MDS], and hypocellular acute myeloid leukemia [h-AML]). Materials and Methods BM specimens were obtained from patients presenting with pancytopenia/bicytopenia. On 30 patients diagnosed as hypocellular BM, immunohistochemistry (IHC) for CD34, CD117, and p53 was performed. Results BM cellularity was < 30% in all (100%) patients. Blast count was increased in h-MDS and h-AML. Features of dysplasia were noted in six (20%) patients. Out of these, three patients were diagnosed as h-MDS having bilineage/trilineage dysplasia, and the other three patients were of AA (11.5% patients) displaying only dyserythropoiesis. On IHC, percentage of BM CD34+ cells was increased in h-MDS+ h-AML (3.87 ± 0.86) as compared with AA (0.19 ± 0.15) and controls (0.81 ± 0.21), p = 0.01. Percentage of BM p53+ cells was also increased in h-MDS+ h-AML (2.9 ± 2.07) as compared with AA and controls, which did not show any p53+ cells, p = 0.0. No statistically significant difference was observed in the expression of CD117 in h-MDS+ h-AML (4.95 ± 3.40) compared with AA (4.49 ± 1.07), p = 0.99. Conclusion The study demonstrates the usefulness of CD34 and p53 immunoexpression as an important ancillary method in distinguishing various hypocellular BM disorders, especially h-MDS and AA. However, the role of CD117 remains unclear and needs to be evaluated further by larger studies.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 923-923
Author(s):  
Katie Giger ◽  
Georgios E Christakopoulos ◽  
Lisa Trump ◽  
Clarissa E. Johnson ◽  
Haripriya Sakthivel ◽  
...  

Abstract CDAR (ClinicalTrials.gov Identifier: NCT02964494), a registry for patients with Congenital Dyserythropoietic Anemia (CDA) in North America, was recently created with the goal to provide a longitudinal database and associated biorepository to facilitate natural history studies and research on the molecular pathways involved in the pathogenesis of CDAs. A 2 y.o. female patient with transfusion dependent anemia, pathologic diagnosis of Congenital Dyserythropoietic Anemia type I (CDA-I), and neurodevelopmental delay was enrolled in CDAR. Next Generation sequencing and deletion/duplication assay identified no mutations in the known CDA-associated genes, including CDAN1 and C15orf41, which are causative for CDA-I. Whole-exome sequencing for the patient and her parents (family-trio design) revealed a novel, de novo VPS4A missense variant located in the last codon of exon 8, potentially affecting splicing. VPS4A is an ATPase which, in association with the endosomal sorting complex required for transport (ESCRT), has been shown to play a critical role in cell division of HeLa cells in vitro, concentrating at the spindle poles during mitosis and at the midbody during cytokinesis. The aim of this work is to validate the pathogenetic role of the VPS4A variant for CDA and further investigate the role of VPS4A in erythroblast mitosis and cytokinesis. Central review of the patient's bone marrow aspirate smears revealed bi-nucleated erythroblasts in the range of 3-7%, a criterion compatible with CDA-I. However, cytoplasmic bridges were noted (arrows in Figure 1A) rather than the nuclear bridges typical of CDA-I. Immunofluorescence staining performed on erythroblasts generated ex vivo from normal CD34+ cells verified that VPS4A localizes to the spindle poles during mitosis and the midbody during cytokinesis in dividing human erythroid cells analyzed by Imaging Flow Cytometry (Figure 1B). The level of VPS4A mRNA expression in the patient's reticulocytes was evaluated by qPCR using three different sets of primers and found to be decreased by 55-70% compared to control reticulocytes and knock-down of VPS4A in normal CD34+ cells resulted in erythroid cultures enriched in binucleated cells. Induced pluripotent stem cells (iPSCs) were generated from the patient's peripheral blood mononuclear cells after the family's consent. Erythroblasts produced from these iPSCs exhibited decreased VPS4A localization at the spindle poles and midbody and fail to divide properly, frequently maintaining cytoplasmic bridges as seen in the patient's bone marrow. Additionally, flow cytometry analysis of the patient's peripheral blood cells stained with anti-CD71 for the transferrin receptor and Thiazol Orange (TO) for RNA revealed a unique cell population which is TO negative, yet CD71 positive implying that VPS4A is also involved in reticulocyte maturation, likely participating in vesicle formation and the normal exocytosis of the transferrin receptor. VPS4A appears to play a critical role in erythroblast mitosis and cytokinesis, as well as erythrocyte maturation, and is a novel candidate gene for congenital dyserythropoietic anemia. Figure 1. A) Binucleated erythroblasts and cytoplasmic bridges (arrows) were noted on the patient's bone marrow aspirate smears. B) VPS4A localizes at the spindle poles (upper image) and midbody (lower image) in normal human erythroblasts. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4251-4251
Author(s):  
Pedro Ramos ◽  
Ella Guy ◽  
Robert W Grady ◽  
Maria de Sousa ◽  
Stefano Rivella

Abstract Abstract 4251 A deficient hepcidin response to iron is the principal mechanism responsible for increased iron uptake from the diet leading to iron overload. In hereditary hemochromatosis (HH), mutations in the HFE gene lead to iron overload through abnormally low levels of hepcidin. Interestingly, hepcidin has been shown to respond to a variety of stimuli, including iron, hypoxia, erythropoiesis and inflammation, requiring integration of the respective signals for its regulation. Further studies showed that HFE/Hfe could also modulate cellular iron uptake by associating with the transferrin receptor-1 (Tfrc), a crucial protein for iron uptake by erythroid cells. In addition, some studies have reported altered erythropoietic values in HH patients. Despite these findings, the role of Hfe in erythropoiesis was never explored. We hypothesized that Hfe influences erythropoiesis by two distinct mechanisms: 1) limiting hepcidin expression, thereby increasing iron availability, under conditions of simultaneous iron overload and stress erythropoiesis; 2) participating directly in the control of transferrin-bound iron uptake by erythroid cells. To test this hypothesis we investigated the role of Hfe in erythropoiesis, aiming to uncover the relative contribution of each of the aforementioned mechanisms. When erythropoiesis was challenged by phlebotomy, Hfe-KO animals were able to recover faster from anemia (p≤0.05) than either normal or iron overloaded wt mice. In Hfe-KO mice, despite their increased iron load, downregulation of hepcidin in response to phlebotomy or erythropoietin administration was comparable to that seen in wt mice. In contrast, iron overloaded wt mice showed increased hepcidin expression both at steady state and after erythropoietic stimulation compared to wt or Hfe-KO mice. In phlebotomized mice fed a standard diet, analysis of serum iron and transferrin saturation indicated that wt mice on the standard diet were able to increase their serum iron very rapidly. After 24 hours, both wt and Hfe-KO mice had similar serum iron and transferrin saturation levels. On the other hand, wt mice kept on an iron deficient diet over the course of phlebotomy, were unable to overcome the phlebotomy-induced anemia. In contrast, Hfe-KO mice fed the low iron diet were able to recover from anemia, although at a slower pace than either Hfe-KO or wt mice on a standard diet. These data indicate that gastrointestinal iron absorption in both wt and Hfe-KO mice is a major factor leading to recovery from anemia, although the excess iron in the liver of Hfe-KO mice contributes to restoration of the red blood cell reservoir. Phlebotomy is the main tool utilized to treat iron overload in HH patients. However, our data suggests that this treatment leads to both mobilization of iron from stores and increased gastrointestinal iron absorption. These observations suggest that patients might benefit from a controlled iron diet or from supplementation with hepcidin or an hepcidin agonist to limit iron absorption. Next, we determined that Hfe is expressed in erythroid cells and that it interacts with Tfrc in murine erythroleukemia cells. Moreover, we discovered that the level of Tfrc expression in Hfe-KO cells is 80% of that seen in wt cells, as measured by flow cytometry. This observation, together with measurement of iron uptake using 59Fe-saturated transferrin, indicated that Hfe-KO erythroid cells take up significantly more iron than wt cells. To confirm that Hfe plays a role in erythropoiesis independent from that in the liver, we transplanted Hfe-KO or wt bone marrow cells into lethally irradiated wt recipients and analyzed their recovery from phlebotomy. We observed that recovery from anemia was faster in Hfe→wt than in wt→wt and was associated with increased mean corpuscular hemoglobin levels, suggesting that lack of Hfe in the hematopoietic compartment can lead to increased hemoglobin production. In summary, our results indicate that lack of Hfe enhances iron availability for erythropoiesis by two distinct mechanisms. On the one hand, Hfe plays an important role in maintaining erythroid iron homeostasis by limiting the response of hepcidin to iron, particularly under conditions of erythropoietic stimulation. On the other hand, lack of Hfe contributes directly to increased iron intake by erythroid progenitors, even in the absence of iron overload. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3917-3917
Author(s):  
Sandra Ballester ◽  
Pilar Eroles ◽  
Blanca Navarro ◽  
Anabel Teruel ◽  
Carlos Solano ◽  
...  

Abstract Abstract 3917 INTRODUCTION VEGF is an important pro-angiogenic factor involved in survival and dissemination of chronic lymphocytic leukemia cells. At high concentrations such as those present in bone marrow niches, VEGF reduces MMP9 production and favours bone marrow retention. But on the other hand, it is well known that autologous VEGF and CD49d are required for B-CLL cell migration. In other cancer cell lines, this effect seems to be mediated by CXCR4/SDF-1 axis. In this work we studied the role of exogenous VEGF on B-CLL migration as well its relationship with the G-coupled proteins CXCR4, CXCR7 and CD49d. MATERIALS AND METHODS We obtained peripheral blood mononuclear cells from 33 patients diagnosed of CLL according to established clinical and laboratory protocols at our institution after informed consent. We also used the CLL cell line Mec-1. B-lymphocytes were purified by Ficoll-Hypaque density gradient centrifugation anti-CD19 conjugated Dynabeads and stored in liquid nitrogen. Then we analysed by flow cytometry the expression of several chemokines and adhesion molecules, CXCR4, CXCR7 and CD49d before and after exogenous VEGF exposure (50 ng/mL), VEGF-R2 inhibitor (70 nM) or both. In order to evaluate the role of VEGF in the motility of these cells, we performed an in vitro 24-hours transmigration assay towards a media containing (or not) SDF-1. B-CLL (5 × 105 cells) cells were incubated on the upper chamber of transwell filters coated with human umbilical vein endothelial cells in the presence or absence (control) of vascular endothelial growth factor (VEGF) ([50 ng/ml], 24 hours), VEGFR2/KDR inhibitor [70nM] or both simultaneously. We quantified the migration ratio between those stimulated with VEGF compared with non-stimulated controls, inhibitor-treated cells versus control and both (R2-inh plus VEGF) versus inhibitor-treated cells. Migration ratio was given as mean ± SD. Statistical analysis was performed by non-parametric Wilcoxon test using SPSS statistical software (version 19.0). RESULTS Basal CXCR4, CXCR7 and CD49d expression levels of B-CLL cells were highly variable among the 33 patients analyzed. Mean fluorescent intensity (MFI) of CXCR4 expression was significantly higher on cells treated with VEGF versus untreated cells (average increase, 9.64 ± 95, p=0.028). However, we did not detect a significant difference in the percentage of cells expressing this receptor. On the other hand, VEGF treatment did not influence either the mean fluorescent intensity or the number of CXCR7 and CD49d expressing cells. Exposure to a VEGFR2 inhibitor reduced the percentage of cells expressing CXCR4 and CXCR7, suggesting a potential regulatory role of this receptor in the expression of these chemokines. Concerning B-CLL migration, we observed a significant increase in cell migration of cells treated with exogenous VEGF versus the control ones in both the Mec-1 cell line and the primary cells (27.66 ± 69.97 p= 0.03). Furthermore, the treatment with VEGFR2 inhibitor reduced significantly the migration index (−23.18 ± 33.5, p= 0.001) and the motility was restored by the addition of VEGF to the R2-inhibitor treated cells (36.04 ± 39.33, p=0.001). DISCUSSION This preliminary data suggest that VEGF seems to be involved in B-CLL migration through the regulation of CXCR4 expression levels. Detailed molecular mechanisms implicated in this process should be further studied. New therapeutic strategies focussed in blocking both the SDF1-CXCR4 axis and/or VEGF pathway could have a potential therapeutic implication by decreasing B-CLL cell migration into lymph nodes and bone marrow microenvironment. Disclosures: No relevant conflicts of interest to declare.


Biomolecules ◽  
2021 ◽  
Vol 11 (6) ◽  
pp. 803
Author(s):  
Emma C. Tovey Crutchfield ◽  
Sarah E. Garnish ◽  
Joanne M. Hildebrand

Necroptosis is an inflammatory form of lytic programmed cell death that is thought to have evolved to defend against pathogens. Genetic deletion of the terminal effector protein—MLKL—shows no overt phenotype in the C57BL/6 mouse strain under conventional laboratory housing conditions. Small molecules that inhibit necroptosis by targeting the kinase activity of RIPK1, one of the main upstream conduits to MLKL activation, have shown promise in several murine models of non-infectious disease and in phase II human clinical trials. This has triggered in excess of one billion dollars (USD) in investment into the emerging class of necroptosis blocking drugs, and the potential utility of targeting the terminal effector is being closely scrutinised. Here we review murine models of disease, both genetic deletion and mutation, that investigate the role of MLKL. We summarize a series of examples from several broad disease categories including ischemia reperfusion injury, sterile inflammation, pathogen infection and hematological stress. Elucidating MLKL’s contribution to mouse models of disease is an important first step to identify human indications that stand to benefit most from MLKL-targeted drug therapies.


Hypertension ◽  
2015 ◽  
Vol 66 (suppl_1) ◽  
Author(s):  
Joseph Gigliotti ◽  
Sylvia Cechova ◽  
Thu Le

The immune system plays a major role in animal models of hypertension (HTN) and end-organ damage. However, few studies have assessed the role of lymphoid organs in the pathogenesis of HTN. We have shown previously that prior splenectomy (SPLX) significantly alters tissue inflammation; however the effect of SPLX on HTN remains unclear. Therefore, the objective of the current study is to determine whether prior SPLX influences the development of HTN in 2 different mouse models. Mice underwent SPLX or sham surgery 7 days prior to the induction of HTN using angiotensin-II (AngII, 400ng/kg*min s.c.) or nitric oxide synthase inhibition using L-NAME (30mg/kg*d in drinking water). Systolic blood pressure (SBP) was measured by tail-cuff manometer daily and mice were euthanized 14 days after induction of HTN. Heart weight/body weight (H/BW) ratios were calculated and kidney leukocyte infiltration was analyzed by flow cytometry. Mice with prior SPLX+AngII had significantly lower (P=0.03) SBP at both week 1 (148±7) and week 2 (135±7) as compared to Sham+AngII (174 and 173±7mmHg, n=5). Similarly, SPLX+AngII mice had significantly smaller (P=0.007) H/BW (4.3±0.3) as compared to Sham+AngII treated mice (5.2±0.4mg/g BW). Interestingly, no difference was observed in renal CD45+ (9.8±3 vs 10.6±3x105 cells/g, P=0.64) or CD3+ T-cell infiltration (8.8±0.2 vs 9.6±0.1x104 cells/g , P=0.64) between the Sham+AngII and SPLX+AngII treated mice, respectively (n=4/5). Furthermore, SPLX did not appear to influence the development of L-NAME HTN. SPLX+L-NAME mice had similar (P=0.84) SBP (145±4mmHg) as the Sham+L-NAME group (146±4mmHg, n=6) after 2 weeks. Relative heart weights were also similar (P=0.45) between SPLX+L-NAME (4.9±0.2) and Sham+L-NAME treated mice (4.8±0.3mg/g BW). Our data suggests that the full pressor response to AngII is dependent on the spleen. However, the effect of the spleen appears to be independent of renal inflammation. Moreover, the protective effect of the spleen is specific to AngII-dependent HTN and does not appear to be generalizable to all mouse models of hypertension. Further studies are needed to understand the physiological link between lymphoid organs (such as the spleen), renal inflammation, and the development of chronic HTN.


Sign in / Sign up

Export Citation Format

Share Document