Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Casey Brewer is active.

Publication


Featured researches published by Casey Brewer.


Journal of Magnetic Resonance Imaging | 2009

Spleen R2 and R2* in iron‐overloaded patients with sickle cell disease and thalassemia major

Casey Brewer; Thomas D. Coates; John C. Wood

To evaluate the magnetic properties of the spleen in chronically transfused, iron‐overloaded patients with sickle cell disease (SCD) and thalassemia major (TM) and to compare splenic iron burdens to those in the liver, heart, pancreas, and kidneys.


Acta Haematologica | 2008

Safety and Efficacy of Combined Chelation Therapy with Deferasirox and Deferoxamine in a Gerbil Model of Iron Overload

Maya Otto-Duessel; Casey Brewer; Ignacio Gonzalez; Hanspeter Nick; John C. Wood

Introduction: Combined therapy with deferoxamine (DFO) and deferasirox (DFX) may be performed empirically when DFX monotherapy fails. Given the lack of published data on this therapy, the study goal was to assess the safety and efficacy of combined DFO/DFX therapy in a gerbil model. Methods: Thirty-two female Mongolian gerbils 8–10 weeks old were divided into 4 groups (sham chelated, DFO, DFX, DFO/DFX). Each received 10 weekly injections of 200 mg/kg iron dextran prior to initiation of 12 weeks of chelation. Experimental endpoints were heart and liver weights, iron concentration and histology. Results: In the heart, there was no significant difference among the treatment groups for wet-to-dry ratio, iron concentration and iron content. DFX-treated animals exhibited lower organ weights relative to sham-chelated animals (less iron-mediated hypertrophy). DFO-treated organs did not differ from sham-chelated organs in any aspects. DFX significantly cleared hepatic iron. No additive effects were observed in the organs of DFO/DFX-treated animals. Conclusions: Combined DFO/DFX therapy produced no detectable additive effect above DFX monotherapy in either the liver or heart, suggesting competition with spontaneous iron elimination mechanisms for chelatable iron. Combined therapy was well tolerated, but its efficacy could not be proven due to limitations in the animal model.


Translational Research | 2011

Interdependence of cardiac iron and calcium in a murine model of iron overload

Maya Otto-Duessel; Casey Brewer; John C. Wood

Iron cardiomyopathy in ß-thalassemia major patients is associated with a vitamin D deficiency. Stores of 25-OH-D3 are markedly reduced, whereas the active metabolite, 1-25-(OH)-D3, is normal or increased. Interestingly, the ratio of 25-OH-D3 to 1-25-(OH)-D3 (a surrogate for parathyroid hormone [PTH]) is the strongest predictor of cardiac iron. Increased PTH and 1-25-OH-D3 levels have been shown to up-regulate L-type voltage-gated calcium channels (LVGCC), the putative channel for cardiac iron uptake. Therefore, we postulate that a vitamin D deficiency increases cardiac iron by altering LVGCC regulation. Hemojuvelin knockout mice were calcitriol treated, PTH treated, vitamin D-depleted, or untreated. Half of the animals in each group received the Ca(2+)-channel blocker verapamil. Mn(2+) was infused to determine LVGCC activity. Hearts and livers were harvested for iron, calcium, and manganese measurements as well as histology. Cardiac iron did not differ among the treatment groups; however, liver iron was increased in vitamin D-depleted animals (P < 0.0003). Cardiac iron levels did not correlate with manganese uptake but were proportional to cardiac calcium levels (r(2) = 0.6; P < 0.0001). Verapamil treatment reduced both cardiac (P < 0.02) and hepatic (P < 0.003) iron levels significantly by 34% and 28%, respectively. The association between cardiac iron and calcium levels was maintained after verapamil treatment (r(2) = 0.3; P < 0.008). Vitamin D depletion is associated with an increase in liver, but not cardiac, iron accumulation. Cardiac iron uptake was strongly correlated with cardiac calcium stores and was significantly attenuated by verapamil, suggesting that cardiac calcium and iron are related.


Experimental Hematology | 2014

mRNA regulation of cardiac iron transporters and ferritin subunits in a mouse model of iron overload

Casey Brewer; Ruth I. Wood; John C. Wood

Iron cardiomyopathy is the leading cause of death in iron overload. Men have twice the mortality rate of women, though the cause is unknown. In hemojuvelin-knockout mice, a model of the disease, males load more cardiac iron than females. We postulated that sex differences in cardiac iron import cause differences in cardiac iron concentration. Reverse transcription polymerase chain reaction was used to measure mRNA of cardiac iron transporters in hemojuvelin-knockout mice. No sex differences were discovered among putative importers of nontransferrin-bound iron (L-type and T-type calcium channels, ZRT/IRT-like protein 14 zinc channels). Transferrin-bound iron transporters were also analyzed; these are controlled by the iron regulatory element/iron regulatory protein (IRE/IRP) system. There was a positive relationship between cardiac iron and ferroportin mRNA in both sexes, but it was significantly steeper in females (p < 0.05). Transferrin receptor 1 and divalent metal transporter 1 were more highly expressed in females than males (p < 0.01 and p < 0.0001, respectively), consistent with their lower cardiac iron levels, as predicted by IRE/IRP regulatory pathways. Light-chain ferritin showed a positive correlation with cardiac iron that was nearly identical in males and females (R(2) = 0.41, p < 0.01; R(2) = 0.56, p < 0.05, respectively), whereas heavy-chain ferritin was constitutively expressed in both sexes. This represents the first report of IRE/IRP regulatory pathways in the heart. Transcriptional regulation of ferroportin was suggested in both sexes, creating a potential mechanism for differential set points for iron export. Constitutive heavy-chain-ferritin expression suggests a logical limit to cardiac iron buffering capacity at levels known to produce heart failure in humans.


Experimental Hematology | 2012

Ascorbate status modulates reticuloendothelial iron stores and response to deferasirox iron chelation in ascorbate-deficient rats

Casey Brewer; Maya Otto-Duessel; Jens Lykkesfeldt; Hanspeter Nick; John C. Wood

Iron chelation is essential to patients on chronic blood transfusions to prevent toxicity from iron overload and remove excess iron. Deferasirox (DFX) is the most commonly used iron chelator in the United States; however, some patients are relatively refractory to DFX therapy. We postulated that vitamin C supplementation would improve the availability of transfusional iron to DFX treatment by promoting irons redox cycling, increasing its soluble ferrous form and promoting its release from reticuloendothelial cells. Osteogenic dystrophy rats (n = 54) were given iron dextran injections for 10 weeks. Cardiac and liver iron levels were measured after iron loading (n = 18), 12 weeks of sham chelation (n = 18), and 12 weeks of DFX chelation (n = 18) at 75 mg/kg/day. Ascorbate supplementation of 150 ppm, 900 ppm, and 2250 ppm was used in the chow to mimic a broad range of ascorbate status; plasma ascorbate levels were 5.4 ± 1.9, 8.2 ± 1.4, 23.6 ± 9.8 μM, respectively (p < 0.0001). The most severe ascorbate deficiency produced reticuloenthelial retention, lowering total hepatic iron by 29% at the end of iron loading (p < 0.05) and limiting iron redistribution from cardiac and hepatic macrophages during 12 weeks of sham chelation. Most importantly, ascorbate supplementation at 2250 ppm improved DFX efficiency, allowing DFX to remove 21% more hepatic iron than ascorbate supplementation with 900 ppm or 150 ppm (p < 0.05). We conclude that vitamin C status modulates the release of iron from the reticuloendothelial system and correlates positively with DFX chelation efficiency. Our findings suggest that ascorbate status should be probed in patients with unsatisfactory response to DFX.


Translational Research | 2014

Sex Differences and Steroid Modulation of Cardiac Iron in a Mouse Model of Iron Overload

Casey Brewer; Maya Otto-Duessel; Ruth I. Wood; John C. Wood

Iron cardiomyopathy is the leading cause of death in transfusional iron overload, and men have twice the mortality of women. Because the prevalence of cardiac iron overload increases rapidly during the second decade of life, we postulated that there are steroid-dependent sex differences in cardiac iron uptake. To test this hypothesis, we manipulated sex steroids in mice with constitutive iron absorption (homozygous hemojuvelin knockout); this model mimics the myocyte iron deposition observed in humans. At 4 weeks of age, female mice were ovariectomized (OVX) and male mice were castrated (OrchX). Female mice received an estrogen implant (OVX + E) or a cholesterol control (OVX), whereas male mice received an implant containing testosterone (OrchX + T), dihydrotestosterone (OrchX + DHT), estrogen (OrchX + E), or cholesterol (OrchX). All animals received a high-iron diet for 8 weeks. OrchX, OVX, and OVX + E mice all had similar cardiac iron loads. However, OrchX + E males had a significant increase in cardiac iron concentration compared with OrchX mice (P < 0.01), whereas the OrchX + T and OrchX + DHT groups only trended higher (P < 0.06 and P < 0.15, respectively). Hormone treatments did not impact liver iron concentration in either sex. When data were pooled across hormone therapies, liver iron concentration was 25% greater in males than females (P < 0.01). In summary, we found that estrogen increased cardiac iron loading in male mice, but not in females. Male mice loaded 25% more hepatic iron than female mice regardless of the hormone treatment.


Journal of Zoo and Wildlife Medicine | 2014

Dose titration of deferasirox iron chelation therapy by magnetic resonance imaging for chronic iron storage disease in three adult red bald-headed uakari (Cacajao calvus rubicundus)

Casey Brewer; J. Michael Tyszka; Cynthia K. Stadler; Michael M. Garner; Janet Baer; John C. Wood

Abstract: Iron overload is common in lemurs and some New World nonhuman primates raised in captivity, but there is no such documentation in the red bald-headed uakari (Cacajao calvus rubicundus). This study describes postmortem documentation of severe iron storage disease in one red bald-headed uakari and the use of iron chelation with oral deferasirox in the three surviving members of the colony. Magnetic resonance imaging was used to quantify pretreatment iron burden and to follow the response to therapy in two females, 22 and 28 yr of age, and one male 33 yr of age. Baseline liver iron concentrations ranged from 16 to 23 mg/g dry weight. In humans, a liver iron concentration greater than 15 mg/g is considered severe and associated with endocrine and cardiac toxicity. The uakaris were otherwise asymptomatic, generally healthy, nonpregnant, and on a stable, low-iron diet. Quantitative magnetic resonance imaging indicated that dosage escalations up to 100 mg/kg were needed to produce meaningful reductions in iron stores. After 5 yr of therapy, two animals continue at a dosage of 100 mg/kg per day, and the third was transitioned to twice-weekly maintenance dosing because of successful de-ironing. The animals tolerated iron chelation therapy well, having stable hematologic, renal, and hepatic function profiles before, during, and after treatment. Deferasirox monotherapy may represent a therapeutic option in primates with iron storage disease when dietary measures are ineffective and phlebotomy is logistically challenging.


Cell Reports | 2016

Transplantation Dose Alters the Differentiation Program of Hematopoietic Stem Cells

Casey Brewer; Elizabeth Chu; Mike Chin; Rong Lu


Experimental Hematology | 2018

A Mathematical Model of Mirna Dynamics Predicts State Transition and Identifies Mirna-126 as an Onco-Mir Promoting Acute Myeloid Leukemia Driven by CBFB-MYH11

Russell C. Rockne; Sergio Branciamore; Jing Qi; Lianjun Zhang; Wei-Kai Hua; Casey Brewer; Le Xuan Nguyen; Bin Zhang; Guerry J. Cook; Emily Carnahan; Dijiong Wu; Maria Ramirez; Man Li; Ayelet Marmon; Herman Wu; Davide Maestrini; Xiwei Wu; Denis OMeally; Yate-Ching Yuan; Nadia Carlesso; Stephen J. Forman; Guido Marcucci; Ya-Huei Kuo


Blood | 2010

The Effect of Supplemental Ascorbate on Defersirox Iron Chelation In the Iron-Loaded Gerbil

Maya Otto-Duessel; Casey Brewer; Aleya Hyderi; Jens Lykkesfeldt; Ignacio Gonzalez-Gomez; Hanspeter Nick; John C. Wood

Collaboration


Dive into the Casey Brewer's collaboration.

Top Co-Authors

Avatar

John C. Wood

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Maya Otto-Duessel

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Aleya Hyderi

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Laura Perin

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Paul J. Kokorowski

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Roger DeFilippo

Children's Hospital Los Angeles

View shared research outputs
Top Co-Authors

Avatar

Ruth I. Wood

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Sargis Sedrakyan

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge