β-Thalassemia Mouse Model: Macrophages and the Induction of Iron Overload.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2699-2699
Author(s):  
Yelena Z. Ginzburg ◽  
Radma Mahmood ◽  
Steven Brunnert ◽  
Mary E. Fabry ◽  
Ronald L. Nagel

Abstract Despite the use of transfusion and iron chelation therapy, patients with β-thalassemia major have a shortened life expectancy. Many of those deaths are attributable to cardiac iron overload. Nevertheless, the process by which cardiac iron overload occurs is not well understood. We have used the homozygous βmajor deletion [Hbbth-1] (THL) mouse model to assess hepatic and cardiac iron load. RBC indices for 3 THL mice and 2 C57BL/6 wildtype control mice prior to and post therapy with parenteral iron were evaluated with Advia. Intraperitoneal iron dextran injection at 10mg/25gm body weight daily 5 days per week for 12 days was performed and then switched to 1.25mg/25gm body weight of iron injection for another 10 days for a total of 4 weeks. Histological samples of liver and heart were stained with Prussian blue in mice prior to and post administration of parenteral iron. Immunohistochemistry with antibody to F4/80, specific for macrophages, was performed and counterstained with Prussian blue in livers and hearts of THL and C57 mice. The RBC indices in THL mice reveal an anemia (HCT 29.5±2.3 vs 45±2.1%, P=0.005) and reticulocytosis (2218±501 vs 406±101 x 109 cells, P=0.018) prior to therapy relative to the C57 mice (values presented as mean ± standard deviation). In THL mice after parenteral iron, HCT (41.8±6.8 vs 29.5±2.3%, P=0.04) and reticulocyte counts (2218±501 vs 3760±633 x 109 cells, P=0.03) increased significantly from pre-treatment values while in C57 mice, the HCT (53.8±6 vs 45±2.1%, NS) and reticulocyte count (406±101 vs 210±49 x109 cells, NS) did not change appreciably from baseline. Prior to therapy, the liver of THL mice exhibit 20–25% Kupffer cells staining with Prussian blue, with no Prussian blue staining in hepatocytes. The hearts of THL mice have no macrophages and no iron deposition at baseline. Prior to therapy, the livers of C57 mice had similar numbers of Kupffer cells compared to THL mice though none stain with Prussian blue. After treatment with parenteral iron, the livers of THL and C57 mice became significantly iron loaded (75–80% of Kupffer cells are positive for Prussian blue), the number of Kupffer cells increased 4-fold, and the majority of the Prussian blue staining was limited to Kupffer cells (90–95%). After treatment with parenteral iron, the hearts of THL and C57 mice became significantly iron loaded as well, but unlike the liver, most (90%) of the Prussian blue positive cells were myocytes. Only a small fraction of the myocytes in the heart was involved (5%). THL mice appear to be iron deficient and show bone marrow reserve with reticulocytosis significantly above baseline when excess iron is administered. Iron overload secondary to intraperitoneal iron dextran administration affects THL mice as well as C57 mice. In the liver of THL mice, Kupffer cells normally resident in the liver become laden with iron; little iron is deposited in hepatocytes. In the heart, an organ without resident macrophages and few macrophages migrating into the tissue during parenteral iron administration, both THL and C57 mice reveal myocyte deposition of iron. In conclusion, parenteral iron administration leads to a noticeable increase in RBCs in THL mice. Furthermore, both the livers and hearts of THL mice accumulate iron. Finally, these findings correlate well with the natural history of cardiac iron overload in human β-thalassemia major, leading to the conclusion that THL mice are a suitable model for the study of cardiac iron overload in thalassemia.

2018 ◽  
Vol 86 (6) ◽  
pp. 1707-1712
Author(s):  
DINA Sh.M. MANAA, M.Sc.; MOHAMMED R. EL-SHANSHORY, M.D. ◽  
OSAMA ABD RAB EL-RASSOL, M.D.; RASHA A. EL-SHAFEY, M.D.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3819-3819
Author(s):  
Elisabetta Volpato ◽  
Elena Cassinerio ◽  
Maria Rosaria Fasulo ◽  
Paola Pedrotti ◽  
Stefano Pedretti ◽  
...  

Abstract Introduction: cardiac failure due to secondary iron overload remains the main cause of death in patients with b-Thalassemia Major (TM). Cardiovascular Magnetic Resonance Imaging (CMR) T2* technique is a new tool to assess myocardial iron concentration that allows to tailor the optimal iron chelation treatment for each patient. Aim of the study: to assess left ventricular function and myocardial iron overload in a cohort of TM patients, cared at Hereditary Anemia Center in Milan, Italy. Methods and Results: In 91 TM patients (33 males/58 females, mean age 32 ± 6 yrs) myocardial iron loading was assessed with the use of CMR T2* measurements (CMR Tools, Cardiovascular Imaging Solutions, London, UK). Left ventricular ejection fraction (LVEF) was also assessed with CMR. In the overall group hemoglobin levels were 9.0 ± 1.0 g/dl; the mean serum ferritin levels and iron intake during the six months before CMR evaluation were 1507 ± 1884 ng/ml and 0.34 ± 0.08 mg/kg/die respectively. T2* was significantly different between females and males (24 ± 11 and 32 ± 12 ms, respectively; p < 0.0001), with significant differences in diabetes mellitus prevalence (17% vs 8%, p<0.01) but not in age, serum ferritin, iron intake and hemoglobin levels (Table 1). Seven (7.6%) asymptomatic females showed a severe cardiac iron overload (T2* ≤ 10 ms), 9 patients (9.9%) moderate (T2* between 10.1 and 14 ms), 15 patients (16.4%) mild cardiac iron overload (T2* between 14.1 and 20 ms) and 60 patients (65.9%) had normal T2* (> 20 ms). LVEF was significantly different between females and males (35% vs 57%, p<0.001) with evidence of a significant relationship between iron overload severity and LVEF impairment (r=0.92). Conclusions: CMR cardiac function and T2* assessment allow to detect pre-symptomatic cardiac iron overload. Females are more at risk for severe iron overload and left ventricular impairment. The prevalence of diabetes mellitus and compliance to chelation therapy could be relevant in explaining the gender differences. Clinical parameters and T2* values in men and women with thalassemia major Men p Women SD: standard deviation Number of patients (n. of pts) 33 - 58 Age ± SD (years) 33 ± 6 ns 32 ± 6 Hemoglobin levels ± SD (g/dl) 9.0 ± 1.7 ns 9.0 ± 0.8 Ferritin levels ± SD (ng/ml) 964 ± 891 ns 1821 ± 2216 Iron intake ± SD (mg/Kg/die) 0.30 ± 0.07 ns 0.36 ± 0.09 Mean T2* value ± SD (ms) 32 ± 12 <0.0001 24 ± 11 T2*< 10 ms (n. of pts) 0 - 7 T2* between 10.1 and 14 ms (n. of pts) 1 - 8 T2* between 14.1 and 20 ms (n. of pts) 7 - 8 T2* > 20 ms (n. of pts) 25 - 35 T2*< 10 ms (n. of pts) plus LVEF≤ 57 % 0/0 (0%) - 6/7 (85.7%) T2* between 10.1 and 14 ms (n. of pts) plus LVEF≤ 57 % 1/1 (100%) - 3/8 (37.5%) T2* between 14.1 and 20 ms (n. of pts) plus LVEF≤ 57 % 3/7 (42.8%) - 1/8 (12.5%) T2* > 20 ms (n. of pts) plus LVEF≤ 57 % 7/25 (28%) - 5/35 (14.3%)


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5415-5415
Author(s):  
Sandra Regina Loggetto ◽  
Mônica Veríssimo ◽  
Antônio Fabron Júnior ◽  
Giorgio Roberto Baldanzi ◽  
Nelson Hamerschlak ◽  
...  

Abstract Introduction: Cardiac failure is a main cause of morbidity and mortality in patients with thalassemia major (TM) who are receiving regular blood transfusion due to iron overload. So, effective and adequate iron chelation is extremely important. Deferoxamine (DFO), the most widely used iron chelator, has poor compliance. Combined therapy with Deferiprone (DFP) increases chelation efficacy, decreases iron-induced complications, improves compliance increasing survival in thalassemia. Objectives: Assessment of efficacy and safety in combined chelation with DFP and DFO in thalassemic patients with iron overload. Methods and results: We have 50 thalassemia major patients in 4 Brazilian Centers (Boldrini Hospital, Sao Paulo Hematology Center, HEMEPAR and FAMEMA) receiving combined chelation therapy with follow up to three years. DFP (75–100 mg/kg/daily) and DFO (30–60 mg/kg, 4–7 days/week) are being administered during one to three years. Median age of this group is 21,5 y/o (range 8–35), with 48% female. Median age to start regular transfusions was 12 months (range 2–140) and to begin chelation therapy was 57 months (range 17–216). All patients were screened for Hepatitis C and 26% had positive sorology and/or PCR. Statistical analysis were made with Spearman test and Fisher test. All patients, except two, did cardiac and liver MRI in the initial phase of the study, resulting in 60,5% with cardiac iron overload (T2*<20ms), being severe in 31,2%. Assessment of liver iron concentration (LIC) showed 95,7% with liver iron overload (>3ug/g dry weight), being severe in 17,4%. During follow up, only 43 patients (86%) was screened with MRI. From these, 67,4% had cardiac iron overload (severe in 32,5%) and 78,6% had liver iron overload (severe in 11,9%). Mean serum ferritin before and after three years were 3095,7 ±1934,5 ng/ml and 2373,9±1987,6 ng/ml, respectively. Our data showed positive correlation between serum ferritin, LIC and ALT, even in initial data and after combined chelation therapy (p<0,001), but there is no correlation between cardiac T2* and LIC and between cardiac T2* and ferritin. DFP adverse events included 8% agranulocytosis, 22% neutropenia, 20% arthralgia and 38% gastric intolerance. DFO adverse events were 2,6% deafness, 2,0% cataract and 12% growth deficit. Hepatic toxicity was found in 6%, but without necessity to stop treatment. Compliance in this group was excellent in 48%, good in 22% and poor in 30%. Conclusions: This is the first multicenter study to evaluate combined chelation therapy in Brazil based on cardiac MRI and LIC. Most patients had cardiac and hepatic iron overload probably because they began iron chelation lately, due to difficult access to iron chelators in the past. Cardiac iron overload didn’t have correlation with ferritin and LIC and these data need more understanding. Age of initial regular blood transfusion, increased transfusional requirement, inadequate chelation or delayed chelation may play a role in this question. Combined therapy with DFO and DFP is effective to decrease serum ferritin and LIC. Follow up and improving compliance may decrease cardiac iron overload. Adverse events are similar to literature. Combined therapy is safety in TM patients with transfusional iron overload.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1013-1013
Author(s):  
Antonella Meloni ◽  
Mammen Puliyel ◽  
Alessia Pepe ◽  
Massimo Lombardi ◽  
Vasilios Berdoukas ◽  
...  

Abstract Introduction Chronically transfused sickle cell disease (SCD) patients have lower risk of endocrine and cardiac iron overload load than comparably transfused thalassemia major patients. The mechanisms for this protection remain controversial but likely reflects lower transferrin saturation and circulating labile iron pools because of chronic inflammation and regeneration of apotransferrin through erythropoiesis. However, cardioprotection is incomplete; we have identified 6 patients out of the 201 patients (3%) followed at our Institution who have prospectively developed cardiac iron. We present the clinical characteristics of these patients to identify potential risk factors for cardiac iron accumulation. Methods Cardiac, hepatic, and pancreatic iron overload were assessed by R2* Magnetic Resonance Imaging (MRI) techniques as extensively described by our laboratory. The medical records of the selected patients were reviewed for demographic data, for transfusion and chelation history and for hematologic and biochemical parameters. Results Table 1 describes clinical characteristics of the six patients at the time they developed detectable cardiac iron (R2* ≥ 50 ms). Patient 6 was included because he showed a R2* of 49 Hz that was increasing rapidly. Five of the six patients were managed on simple transfusions. Five patients had been on chronic transfusion for more than 11 years. The three patients who developed cardiac iron the earliest (3.7 – 14 years of transfusions) had more efficient suppression of endogenous red cell production (HbS levels 2-5%) compared with patients who required longer transfusional exposure (HbS levels 13.3 – 41%). All patients had qualitatively poor chelation compliance (<50%), based upon their prescription refill rate. All patients had serum ferritin levels exceeding 4600 and liver iron concentration (LIC) greater than 22 mg/g. Pancreatic R2* was greater than 100 Hz in every patient studied (5/6). Figure 1 shows the longitudinal relationship between iron overload in the heart and in the other organs for each patient; initial iron levels are shown in black. Cardiac R2* appears increase dramatically once a critical LIC “threshold” is reached, qualitatively similar to the 18 mg/g threshold observed in thalassemia major patients. Cardiac R2* rose proportionally to pancreas R2*, similar to thalassemia major patients, with all of the patients having pancreas R2* > 100 Hz at the time cardiac iron was detected. Conclusions Cardiac iron overload occurs in a small percentage of chronically transfused SCD patients and is only associated with exceptionally poor control of total body iron stores. Duration of chronic transfusion is clearly important but other factors, such as levels of effective erythropoiesis, may also contribute to cardiac risk. The relationship between cardiac iron and pancreas R2* suggests that pancreas R2* can serve as a valuable screening tool for cardiac iron in SCD patients. Disclosures: Berdoukas: ApoPharma inc: Consultancy. Coates:ApoPharma inc, Novartis, Shire: Consultancy. Wood:Novartis: Consultancy, Honoraria; Shire: Consultancy, Research Funding; ApoPharma: Consultancy, Honoraria, Use of deferiprone in myocardial infarction, Use of deferiprone in myocardial infarction Patents & Royalties.


Hematology ◽  
2015 ◽  
Vol 21 (1) ◽  
pp. 46-53 ◽  
Author(s):  
Galila M. Mokhtar ◽  
Eman M. Sherif ◽  
Nevin M. Habeeb ◽  
Abeer A. Abdelmaksoud ◽  
Eman A. El-Ghoroury ◽  
...  

2020 ◽  
Vol 21 (8) ◽  
Author(s):  
Yazdan Ghandi ◽  
Danial Habibi ◽  
Aziz Eghbali

Background: Cardiac involvement in beta-thalassemia major patients is an important cause of mortality. Therefore, in these patients, timely diagnosis of cardiac disorder is essential. Objectives: The present study aimed at determining the association between cardiac iron overload and fragmented QRS (fQRS). Methods: This cross-sectional study was conducted on 40 β-TM patients, aged 5 - 40 years. The presence of fQRS was evaluated in 12-lead surface electrocardiograms. Cardiac T2* MRI was performed to determine the iron overload. The patients were divided into four groups of chelation therapy. Results: The mean age of patients was reported to be 22.50 ± 6.75 years. The groups showed no significant difference regarding gender, age, or left ventricular ejection fraction. The presence of fQRS was detected in 10 patients (25%), while T2* value was lower than 20 ms in 10 patients (25%). The mean age of patients with and without fQRS was 26.23 ± 2.71 and 19.40 ± 2.61 years, respectively (P = 0.001). The univariate analysis indicated that fQRS had a significant relationship with cardiac iron overload (OR = 5; 95% CI: 1.04 - 23.99; P < 0.044). The multiple logistic regression analysis represented a significant association between iron overload and fQRS (OR = 5.556; 95% CI: 1.027 - 30.049). The sensitivity and specificity of the fQRS against MRI were equal to 50% and 83.3% respectively. Conclusions: The absence of fQRS on ECGs could be a good predictor of the lack of cardiac iron overload in β-TM patients. The results showed that fQRS might indicate the no need for close monitoring for cardiac overload with cardiac MRI and aggressive chelation therapy.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 2766-2766
Author(s):  
Jon A. Detterich ◽  
Janelle Miller ◽  
Thomas D. Coates ◽  
John C. Wood

Abstract Electrocardiographic abnormalities are common in Thalassemia Major patients as well as in animal models of iron overload. The relationship between these ECG abnormalities and the degree of cardiac iron overload has not been well characterized. Hypothesis: Standard 12-lead ECG identifies preclinical cardiac iron deposition. Methods: Fifty-four patients with Thalassemia Major underwent MRI quantification of cardiac iron within one month of a standard 12-lead ECG. Cardiac T2* measurements to quantify iron were performed using a validated multiecho gradient-echo sequence performed on a 1.5 Tesla General Electric CVi scanner. The PR, QRS, QT, and QTc durations as well as P-wave, T-wave and QRS axes and heart rates were measured. Normative data was derived from 20 control patients without detectable cardiac iron (T2* > 20 ms) and age-appropriate norms were created for each ECG criteria. Z-scores for the ECG parameters were then calculated for 34 patients having detectable cardiac iron (T2* < 20ms). Results: ECG parameters varied significantly with age, with correlation coefficients ranging from 0.39 (QTc interval) to 0.66 (PR interval). Cardiac iron was associated with lower heart rates (Z=−0.44, p=0.06), QTc prolongation (Z=0.48, p=0.04) and leftward shift of the P and T wave axes [P-Axis (Z = −1.53, p<0.001); T-Axis (Z= −1.91, p<0.001)]. In total, 31 of 34 pts with cardiac iron had one or more ECG abnormalities (|Z|>2), compared to only 7 of 20 control pts (p<0.0001). Subjective ECG review was consistent with Z-score. The most common abnormalities associated with cardiac iron were non-specific ST-T wave changes (n = 15), sinus bradycardia (n=4), symmetric T-wave inversions (n = 4) and LVH (n = 4). The presence of any of these findings on ECG predicts detectable cardiac iron with a sensitivity and specificity of 68% and 85%. A combined metric of PR prolongation (Z>2), left shift of P or T axis (Z<–2), bradycardia, LVH, ST changes, or symmetric T-wave inversions produced a sensitivity of 94% and specificity of 75% for detectable cardiac iron. Conclusions: Cardiac iron overload produces characteristic abnormalities in atrial conduction and ventricular repolarization. ECG may be an effective and inexpensive screening modality for cardiac iron overload. Figure Figure Figure Figure


2013 ◽  
Vol 126 (9) ◽  
pp. 834-837 ◽  
Author(s):  
Juliano Lara Fernandes ◽  
Erika Fontana Sampaio ◽  
Kleber Fertrin ◽  
Otavio Rizzi Coelho ◽  
Sandra Loggetto ◽  
...  

Haematologica ◽  
2010 ◽  
Vol 96 (1) ◽  
pp. 48-54 ◽  
Author(s):  
D. J. Pennell ◽  
J. B. Porter ◽  
M. D. Cappellini ◽  
L. L. Chan ◽  
A. El-Beshlawy ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document