Network


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

Hotspot


Dive into the research topics where Arineh Khechaduri is active.

Publication


Featured researches published by Arineh Khechaduri.


Journal of Clinical Investigation | 2014

Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation

Yoshihiko Ichikawa; Mohsen Ghanefar; Marina Bayeva; Rongxue Wu; Arineh Khechaduri; Sathyamangla V. Naga Prasad; R. Kannan Mutharasan; Tejaswitha J Naik; Hossein Ardehali

Doxorubicin is an effective anticancer drug with known cardiotoxic side effects. It has been hypothesized that doxorubicin-dependent cardiotoxicity occurs through ROS production and possibly cellular iron accumulation. Here, we found that cardiotoxicity develops through the preferential accumulation of iron inside the mitochondria following doxorubicin treatment. In isolated cardiomyocytes, doxorubicin became concentrated in the mitochondria and increased both mitochondrial iron and cellular ROS levels. Overexpression of ABCB8, a mitochondrial protein that facilitates iron export, in vitro and in the hearts of transgenic mice decreased mitochondrial iron and cellular ROS and protected against doxorubicin-induced cardiomyopathy. Dexrazoxane, a drug that attenuates doxorubicin-induced cardiotoxicity, decreased mitochondrial iron levels and reversed doxorubicin-induced cardiac damage. Finally, hearts from patients with doxorubicin-induced cardiomyopathy had markedly higher mitochondrial iron levels than hearts from patients with other types of cardiomyopathies or normal cardiac function. These results suggest that the cardiotoxic effects of doxorubicin develop from mitochondrial iron accumulation and that reducing mitochondrial iron levels protects against doxorubicin-induced cardiomyopathy.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Disruption of ATP-binding cassette B8 in mice leads to cardiomyopathy through a decrease in mitochondrial iron export

Yoshihiko Ichikawa; Marina Bayeva; Mohsen Ghanefar; Vishnu Potini; Lin Sun; R. Kannan Mutharasan; Rongxue Wu; Arineh Khechaduri; Tejaswitha J Naik; Hossein Ardehali

Mitochondrial iron levels are tightly regulated, as iron is essential for the synthesis of Fe/S clusters and heme in the mitochondria, but high levels can cause oxidative stress. The ATP-binding cassette (ABC) transporter ABCB8 is a mitochondrial inner membrane protein with an unknown function. Here, we show that ABCB8 is involved in mitochondrial iron export and is essential for baseline cardiac function. Induced genetic deletion of ABCB8 in mouse hearts resulted in mitochondrial iron accumulation and cardiomyopathy, as assessed by echocardiography and invasive hemodynamics. Mice with ABCB8 deletion in the heart also displayed mitochondrial damage, and higher levels of reactive oxygen species and cell death. Down-regulation of ABCB8 in vitro resulted in decreased iron export from isolated mitochondria, whereas its overexpression had the opposite effect. Furthermore, ABCB8 is needed for the maturation of the cytosolic Fe/S proteins, as its deletion in vitro and in vivo led to decreased activity of cytosolic, but not mitochondrial, iron–sulfur-containing enzymes. These results indicate that ABCB8 is essential for normal cardiac function, maintenance of mitochondrial iron homeostasis and maturation of cytosolic Fe/S proteins. In summary, this report provides characterization of a protein involved in mitochondrial iron export.


Cell Metabolism | 2012

mTOR regulates cellular iron homeostasis through tristetraprolin.

Marina Bayeva; Arineh Khechaduri; Sergi Puig; Hsiang Chun Chang; Sonika Patial; Perry J. Blackshear; Hossein Ardehali

Iron is an essential cofactor with unique redox properties. Iron-regulatory proteins 1 and 2 (IRP1/2) have been established as important regulators of cellular iron homeostasis, but little is known about the role of other pathways in this process. Here we report that the mammalian target of rapamycin (mTOR) regulates iron homeostasis by modulating transferrin receptor 1 (TfR1) stability and altering cellular iron flux. Mechanistic studies identify tristetraprolin (TTP), a protein involved in anti-inflammatory response, as the downstream target of mTOR that binds to and enhances degradation of TfR1 mRNA. We also show that TTP is strongly induced by iron chelation, promotes downregulation of iron-requiring genes in both mammalian and yeast cells, and modulates survival in low-iron states. Taken together, our data uncover a link between metabolic, inflammatory, and iron-regulatory pathways, and point toward the existence of a yeast-like TTP-mediated iron conservation program in mammals.


Circulation Research | 2013

ATP-binding cassette B10 regulates early steps of heme synthesis

Marina Bayeva; Arineh Khechaduri; Rongxue Wu; Michael A. Burke; J. Andrew Wasserstrom; Neha Singh; Marc Liesa; Orian S. Shirihai; Nathaniel B. Langer; Barry H. Paw; Hossein Ardehali

Rationale: Heme plays a critical role in gas exchange, mitochondrial energy production, and antioxidant defense in cardiovascular system. The mitochondrial transporter ATP-binding cassette (ABC) B10 has been suggested to export heme out of the mitochondria and is required for normal hemoglobinization of erythropoietic cells and protection against ischemia–reperfusion injury in the heart; however, its primary function has not been established. Objective: The aim of this study was to identify the function of ABCB10 in heme synthesis in cardiac cells. Methods and Results: Knockdown of ABCB10 in cardiac myoblasts significantly reduced heme levels and the activities of heme-containing proteins, whereas supplementation with &dgr;-aminolevulinic acid reversed these defects. Overexpression of mitochondrial &dgr;-aminolevulinic acid synthase 2, the rate-limiting enzyme upstream of &dgr;-aminolevulinic acid export, failed to restore heme levels in cells with ABCB10 downregulation. ABCB10 and heme levels were increased by hypoxia, and reversal of ABCB10 upregulation caused oxidative stress and cell death. Furthermore, ABCB10 knockdown in neonatal rat cardiomyocytes resulted in a significant delay of calcium removal from the cytoplasm, suggesting a relaxation defect. Finally, ABCB10 expression and heme levels were altered in failing human hearts and mice with ischemic cardiomyopathy. Conclusions: ABCB10 plays a critical role in heme synthesis pathway by facilitating &dgr;-aminolevulinic acid production or export from the mitochondria. In contrast to previous reports, we show that ABCB10 is not a heme exporter and instead is required for the early mitochondrial steps of heme biosynthesis.


Journal of the American College of Cardiology | 2013

Heme Levels are Increased in Human Failing Hearts

Arineh Khechaduri; Marina Bayeva; Hsiang Chun Chang; Hossein Ardehali

OBJECTIVES The goal of this study was to characterize the regulation of heme and non-heme iron in human failing hearts. BACKGROUND Iron is an essential molecule for cellular physiology, but in excess it facilitates oxidative stress. Mitochondria are the key regulators of iron homeostasis through heme and iron-sulfur cluster synthesis. Because mitochondrial function is depressed in failing hearts and iron accumulation can lead to oxidative stress, we hypothesized that iron regulation may also be impaired in heart failure (HF). METHODS We measured mitochondrial and cytosolic heme and non-heme iron levels in failing human hearts retrieved during cardiac transplantation surgery. In addition, we examined the expression of genes regulating cellular iron homeostasis, the heme biosynthetic pathway, and micro-RNAs that may potentially target iron regulatory networks. RESULTS Although cytosolic non-heme iron levels were reduced in HF, mitochondrial iron content was maintained. Moreover, we observed a significant increase in heme levels in failing hearts, with corresponding feedback inhibition of the heme synthetic enzymes and no change in heme degradation. The rate-limiting enzyme in heme synthesis, delta-aminolevulinic acid synthase 2 (ALAS2), was significantly upregulated in HF. Overexpression of ALAS2 in H9c2 cardiac myoblasts resulted in increased heme levels, and hypoxia and erythropoietin treatment increased heme production through upregulation of ALAS2. Finally, increased heme levels in cardiac myoblasts were associated with excess production of reactive oxygen species and cell death, suggesting a maladaptive role for increased heme in HF. CONCLUSIONS Despite global mitochondrial dysfunction, heme levels are maintained above baseline in human failing hearts.


Circulation Research | 2013

ABCB10 Regulates Early Steps of Heme Synthesis

Marina Bayeva; Arineh Khechaduri; Rongxue Wu; Michael A. Burke; J. Andrew Wasserstrom; Neha Singh; Marc Liesa; Orian S. Shirihai; Nathaniel B. Langer; Barry H. Paw; Hossein Ardehali

Rationale: Heme plays a critical role in gas exchange, mitochondrial energy production, and antioxidant defense in cardiovascular system. The mitochondrial transporter ATP-binding cassette (ABC) B10 has been suggested to export heme out of the mitochondria and is required for normal hemoglobinization of erythropoietic cells and protection against ischemia–reperfusion injury in the heart; however, its primary function has not been established. Objective: The aim of this study was to identify the function of ABCB10 in heme synthesis in cardiac cells. Methods and Results: Knockdown of ABCB10 in cardiac myoblasts significantly reduced heme levels and the activities of heme-containing proteins, whereas supplementation with &dgr;-aminolevulinic acid reversed these defects. Overexpression of mitochondrial &dgr;-aminolevulinic acid synthase 2, the rate-limiting enzyme upstream of &dgr;-aminolevulinic acid export, failed to restore heme levels in cells with ABCB10 downregulation. ABCB10 and heme levels were increased by hypoxia, and reversal of ABCB10 upregulation caused oxidative stress and cell death. Furthermore, ABCB10 knockdown in neonatal rat cardiomyocytes resulted in a significant delay of calcium removal from the cytoplasm, suggesting a relaxation defect. Finally, ABCB10 expression and heme levels were altered in failing human hearts and mice with ischemic cardiomyopathy. Conclusions: ABCB10 plays a critical role in heme synthesis pathway by facilitating &dgr;-aminolevulinic acid production or export from the mitochondria. In contrast to previous reports, we show that ABCB10 is not a heme exporter and instead is required for the early mitochondrial steps of heme biosynthesis.


Journal of Clinical Investigation | 2014

Cardiac-specific ablation of ARNT leads to lipotoxicity and cardiomyopathy.

Rongxue Wu; Hsiang Chun Chang; Arineh Khechaduri; Kusum Chawla; Minh Tran; Xiaomeng Chai; Cory S. Wagg; Mohsen Ghanefar; Xinghang Jiang; Marina Bayeva; Frank J. Gonzalez; Gary D. Lopaschuk; Hossein Ardehali

Patients with type 2 diabetes often present with cardiovascular complications; however, it is not clear how diabetes promotes cardiac dysfunction. In murine models, deletion of the gene encoding aryl hydrocarbon nuclear translocator (ARNT, also known as HIF1β) in the liver or pancreas leads to a diabetic phenotype; however, the role of ARNT in cardiac metabolism is unknown. Here, we determined that cardiac-specific deletion of Arnt in adult mice results in rapid development of cardiomyopathy (CM) that is characterized by accumulation of lipid droplets. Compared with hearts from ARNT-expressing mice, ex vivo analysis of ARNT-deficient hearts revealed a 2-fold increase in fatty acid (FA) oxidation as well as a substantial increase in the expression of PPARα and its target genes. Furthermore, deletion of both Arnt and Ppara preserved cardiac function, improved survival, and completely reversed the FA accumulation phenotype, indicating that PPARα mediates the detrimental effects of Arnt deletion in the heart. Finally, we determined that ARNT directly regulates Ppara expression by binding to its promoter and forming a complex with HIF2α. Together, these findings suggest that ARNT is a critical regulator of myocardial FA metabolism and that its deletion leads to CM and an increase in triglyceride accumulation through PPARα.


Journal of the American Heart Association | 2013

MicroRNA‐210 Decreases heme Levels by Targeting Ferrochelatase in Cardiomyocytes

Aijun Qiao; Arineh Khechaduri; R. Kannan Mutharasan; Rongxue Wu; Varun Nagpal; Hossein Ardehali

Background MicroRNA‐210 (miR‐210) increases in hypoxia and regulates mitochondrial respiration through modulation of iron‐sulfur cluster assembly proteins (ISCU1/2), a protein that is involved in Fe/S cluster synthesis. However, it is not known how miR‐210 affects cellular iron levels or production of heme, another iron containing molecule that is also needed for cellular and mitochondrial function. Methods and Results To screen for micro‐ribonucleic acids (miRNAs) regulated by iron, we performed a miRNA gene array in neonatal rat cardiomyocytes treated with iron chelators. Levels of miR‐210 are significantly increased with iron chelation, however, this response was mediated entirely through the hypoxia‐inducible factor (HIF) pathway. Furthermore, miR‐210 reduced cellular heme levels and the activity of mitochondrial and cytosolic heme‐containing proteins by modulating ferrochelatase (FECH), the last enzyme in heme biosynthesis. Mutation of the 2 miR‐210 binding sites in the 3′ untranslated region (UTR) of FECH reversed the miR‐210 response, while mutation of either binding site in isolation did not exert any effects. Changes mediated by miR‐210 in heme and FECH were independent of ISCU, as overexpression of an ISCU construct lacking the 3′ UTR does not alter miR‐210 regulation of heme and FECH. Finally, FECH levels increased in hypoxia, and this effect was not reversed by miR‐210 knockdown, suggesting that the effects of miR‐210 on heme are restricted to normoxic conditions, and that the pathway is overriden in hypoxia. Conclusions Our results identify a role for miR‐210 in the regulation of heme production by targeting and inhibiting FECH under normoxic conditions.


Journal of the American Heart Association | 2015

Increased Heme Levels in the Heart Lead to Exacerbated Ischemic Injury

Konrad T. Sawicki; Meng Shang; Rongxue Wu; Hsiang Chun Chang; Arineh Khechaduri; Tatsuya Sato; Christine Kamide; Ting Liu; Sathyamangla V. Naga Prasad; Hossein Ardehali

Background Heme is an essential iron-containing molecule for cardiovascular physiology, but in excess it may increase oxidative stress. Failing human hearts have increased heme levels, with upregulation of the rate-limiting enzyme in heme synthesis, δ-aminolevulinic acid synthase 2 (ALAS2), which is normally not expressed in cardiomyocytes. We hypothesized that increased heme accumulation (through cardiac overexpression of ALAS2) leads to increased oxidative stress and cell death in the heart. Methods and Results We first showed that ALAS2 and heme levels are increased in the hearts of mice subjected to coronary ligation. To determine the causative role of increased heme in the development of heart failure, we generated transgenic mice with cardiac-specific overexpression of ALAS2. While ALAS2 transgenic mice have normal cardiac function at baseline, their hearts display increased heme content, higher oxidative stress, exacerbated cell death, and worsened cardiac function after coronary ligation compared to nontransgenic littermates. We confirmed in cultured cardiomyoblasts that the increased oxidative stress and cell death observed with ALAS2 overexpression is mediated by increased heme accumulation. Furthermore, knockdown of ALAS2 in cultured cardiomyoblasts exposed to hypoxia reversed the increases in heme content and cell death. Administration of the mitochondrial antioxidant MitoTempo to ALAS2-overexpressing cardiomyoblasts normalized the elevated oxidative stress and cell death levels to baseline, indicating that the effects of increased ALAS2 and heme are through elevated mitochondrial oxidative stress. The clinical relevance of these findings was supported by the finding of increased ALAS2 induction and heme accumulation in failing human hearts from patients with ischemic cardiomyopathy compared to nonischemic cardiomyopathy. Conclusions Heme accumulation is detrimental to cardiac function under ischemic conditions, and reducing heme in the heart may be a novel approach for protection against the development of heart failure.


Nature Communications | 2017

Corrigendum: Snf1-related kinase improves cardiac mitochondrial efficiency and decreases mitochondrial uncoupling

Amy K. Rines; Hsiang-Chun Chang; Rongxue Wu; Tatsuya Sato; Arineh Khechaduri; Hidemichi Kouzu; Jason S. Shapiro; Meng Shang; Michael A. Burke; Eltyeb Abdelwahid; Xinghang Jiang; Chunlei Chen; Tenley Rawlings; Gary D. Lopaschuk; Paul T. Schumacker; E. Dale Abel; Hossein Ardehali

This corrects the article DOI: 10.1038/ncomms14095.

Collaboration


Dive into the Arineh Khechaduri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rongxue Wu

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tatsuya Sato

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy K. Rines

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

E. Dale Abel

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge