Network


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

Hotspot


Dive into the research topics where Hesham A. Sadek is active.

Publication


Featured researches published by Hesham A. Sadek.


Science | 2011

Transient Regenerative Potential of the Neonatal Mouse Heart

Enzo R. Porrello; Ahmed I. Mahmoud; E R Simpson; Joseph A. Hill; James A. Richardson; Eric N. Olson; Hesham A. Sadek

The heart in a newborn mouse can rebuild itself after injury, but this regenerative capacity is lost within a few days. Certain fish and amphibians retain a robust capacity for cardiac regeneration throughout life, but the same is not true of the adult mammalian heart. Whether the capacity for cardiac regeneration is absent in mammals or whether it exists and is switched off early after birth has been unclear. We found that the hearts of 1-day-old neonatal mice can regenerate after partial surgical resection, but this capacity is lost by 7 days of age. This regenerative response in 1-day-old mice was characterized by cardiomyocyte proliferation with minimal hypertrophy or fibrosis, thereby distinguishing it from repair processes. Genetic fate mapping indicated that the majority of cardiomyocytes within the regenerated tissue originated from preexisting cardiomyocytes. Echocardiography performed 2 months after surgery revealed that the regenerated ventricular apex had normal systolic function. Thus, for a brief period after birth, the mammalian heart appears to have the capacity to regenerate.


Cell Stem Cell | 2010

The Distinct Metabolic Profile of Hematopoietic Stem Cells Reflects Their Location in a Hypoxic Niche

Tugba Simsek; Fatih Kocabas; Junke Zheng; Ralph J. DeBerardinis; Ahmed I. Mahmoud; Eric N. Olson; Jay W. Schneider; Cheng Cheng Zhang; Hesham A. Sadek

Bone marrow transplantation is the primary therapy for numerous hematopoietic disorders. The efficiency of bone marrow transplantation depends on the function of long-term hematopoietic stem cells (LT-HSCs), which is markedly influenced by their hypoxic niche. Survival in this low-oxygen microenvironment requires significant metabolic adaptation. Here, we show that LT-HSCs utilize glycolysis instead of mitochondrial oxidative phosphorylation to meet their energy demands. We used flow cytometry to identify a unique low mitochondrial activity/glycolysis-dependent subpopulation that houses the majority of hematopoietic progenitors and LT-HSCs. Finally, we demonstrate that Meis1 and Hif-1alpha are markedly enriched in LT-HSCs and that Meis1 regulates HSC metabolism through transcriptional activation of Hif-1alpha. These findings reveal an important transcriptional network that regulates HSC metabolism.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Regulation of neonatal and adult mammalian heart regeneration by the miR-15 family

Enzo R. Porrello; Ahmed I. Mahmoud; E R Simpson; Brett A. Johnson; David Grinsfelder; Diana C. Canseco; Pradeep P.A. Mammen; Beverly A. Rothermel; Eric N. Olson; Hesham A. Sadek

We recently identified a brief time period during postnatal development when the mammalian heart retains significant regenerative potential after amputation of the ventricular apex. However, one major unresolved question is whether the neonatal mouse heart can also regenerate in response to myocardial ischemia, the most common antecedent of heart failure in humans. Here, we induced ischemic myocardial infarction (MI) in 1-d-old mice and found that this results in extensive myocardial necrosis and systolic dysfunction. Remarkably, the neonatal heart mounted a robust regenerative response, through proliferation of preexisting cardiomyocytes, resulting in full functional recovery within 21 d. Moreover, we show that the miR-15 family of microRNAs modulates neonatal heart regeneration through inhibition of postnatal cardiomyocyte proliferation. Finally, we demonstrate that inhibition of the miR-15 family from an early postnatal age until adulthood increases myocyte proliferation in the adult heart and improves left ventricular systolic function after adult MI. We conclude that the neonatal mammalian heart can regenerate after myocardial infarction through proliferation of preexisting cardiomyocytes and that the miR-15 family contributes to postnatal loss of cardiac regenerative capacity.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Hippo pathway effector Yap promotes cardiac regeneration

Mei Xin; Yuri Kim; Lillian B. Sutherland; Masao Murakami; Xiaoxia Qi; John McAnally; Enzo R. Porrello; Ahmed I. Mahmoud; Wei Tan; John M. Shelton; James A. Richardson; Hesham A. Sadek; Rhonda Bassel-Duby; Eric N. Olson

The adult mammalian heart has limited potential for regeneration. Thus, after injury, cardiomyocytes are permanently lost, and contractility is diminished. In contrast, the neonatal heart can regenerate owing to sustained cardiomyocyte proliferation. Identification of critical regulators of cardiomyocyte proliferation and quiescence represents an important step toward potential regenerative therapies. Yes-associated protein (Yap), a transcriptional cofactor in the Hippo signaling pathway, promotes proliferation of embryonic cardiomyocytes by activating the insulin-like growth factor and Wnt signaling pathways. Here we report that mice bearing mutant alleles of Yap and its paralog WW domain containing transcription regulator 1 (Taz) exhibit gene dosage-dependent cardiac phenotypes, suggesting redundant roles of these Hippo pathway effectors in establishing proper myocyte number and maintaining cardiac function. Cardiac-specific deletion of Yap impedes neonatal heart regeneration, resulting in a default fibrotic response. Conversely, forced expression of a constitutively active form of Yap in the adult heart stimulates cardiac regeneration and improves contractility after myocardial infarction. The regenerative activity of Yap is correlated with its activation of embryonic and proliferative gene programs in cardiomyocytes. These findings identify Yap as an important regulator of cardiac regeneration and provide an experimental entry point to enhance this process.


Journal of Clinical Investigation | 2014

Macrophages are required for neonatal heart regeneration

Arin B. Aurora; Enzo R. Porrello; Wei Tan; Ahmed I. Mahmoud; Joseph A. Hill; Rhonda Bassel-Duby; Hesham A. Sadek; Eric N. Olson

Myocardial infarction (MI) leads to cardiomyocyte death, which triggers an immune response that clears debris and restores tissue integrity. In the adult heart, the immune system facilitates scar formation, which repairs the damaged myocardium but compromises cardiac function. In neonatal mice, the heart can regenerate fully without scarring following MI; however, this regenerative capacity is lost by P7. The signals that govern neonatal heart regeneration are unknown. By comparing the immune response to MI in mice at P1 and P14, we identified differences in the magnitude and kinetics of monocyte and macrophage responses to injury. Using a cell-depletion model, we determined that heart regeneration and neoangiogenesis following MI depends on neonatal macrophages. Neonates depleted of macrophages were unable to regenerate myocardia and formed fibrotic scars, resulting in reduced cardiac function and angiogenesis. Immunophenotyping and gene expression profiling of cardiac macrophages from regenerating and nonregenerating hearts indicated that regenerative macrophages have a unique polarization phenotype and secrete numerous soluble factors that may facilitate the formation of new myocardium. Our findings suggest that macrophages provide necessary signals to drive angiogenesis and regeneration of the neonatal mouse heart. Modulating inflammation may provide a key therapeutic strategy to support heart regeneration.


Journal of Clinical Investigation | 2012

MicroRNA-214 protects the mouse heart from ischemic injury by controlling Ca2+ overload and cell death

Arin B. Aurora; Ahmed I. Mahmoud; Xiang Luo; Brett Johnson; Eva van Rooij; Satoshi Matsuzaki; Kenneth M. Humphries; Joseph A. Hill; Rhonda Bassel-Duby; Hesham A. Sadek; Eric N. Olson

Early reperfusion of ischemic cardiac tissue remains the most effective intervention for improving clinical outcome following myocardial infarction. However, abnormal increases in intracellular Ca²⁺ during myocardial reperfusion can cause cardiomyocyte death and consequent loss of cardiac function, referred to as ischemia/reperfusion (IR) injury. Therapeutic modulation of Ca²⁺ handling provides some cardioprotection against the paradoxical effects of restoring blood flow to the heart, highlighting the significance of Ca²⁺ overload to IR injury. Cardiac IR is also accompanied by dynamic changes in the expression of microRNAs (miRNAs); for example, miR-214 is upregulated during ischemic injury and heart failure, but its potential role in these processes is unknown. Here, we show that genetic deletion of miR-214 in mice causes loss of cardiac contractility, increased apoptosis, and excessive fibrosis in response to IR injury. The cardioprotective roles of miR-214 during IR injury were attributed to repression of the mRNA encoding sodium/calcium exchanger 1 (Ncx1), a key regulator of Ca²⁺ influx; and to repression of several downstream effectors of Ca²⁺ signaling that mediate cell death. These findings reveal a pivotal role for miR-214 as a regulator of cardiomyocyte Ca²⁺ homeostasis and survival during cardiac injury.


Nature | 2013

Meis1 regulates postnatal cardiomyocyte cell cycle arrest

Ahmed I. Mahmoud; Fatih Kocabas; Shalini Muralidhar; Wataru Kimura; Ahmed S. Koura; Suwannee Thet; Enzo R. Porrello; Hesham A. Sadek

The neonatal mammalian heart is capable of substantial regeneration following injury through cardiomyocyte proliferation. However, this regenerative capacity is lost by postnatal day 7 and the mechanisms of cardiomyocyte cell cycle arrest remain unclear. The homeodomain transcription factor Meis1 is required for normal cardiac development but its role in cardiomyocytes is unknown. Here we identify Meis1 as a critical regulator of the cardiomyocyte cell cycle. Meis1 deletion in mouse cardiomyocytes was sufficient for extension of the postnatal proliferative window of cardiomyocytes, and for re-activation of cardiomyocyte mitosis in the adult heart with no deleterious effect on cardiac function. In contrast, overexpression of Meis1 in cardiomyocytes decreased neonatal myocyte proliferation and inhibited neonatal heart regeneration. Finally, we show that Meis1 is required for transcriptional activation of the synergistic CDK inhibitors p15, p16 and p21. These results identify Meis1 as a critical transcriptional regulator of cardiomyocyte proliferation and a potential therapeutic target for heart regeneration.


Cell | 2014

The Oxygen-Rich Postnatal Environment Induces Cardiomyocyte Cell-Cycle Arrest through DNA Damage Response

Bao N. Puente; Wataru Kimura; Shalini Muralidhar; Jesung Moon; James F. Amatruda; Katherine J Phelps; David Grinsfelder; Beverly A. Rothermel; Rui Chen; Joseph A. Garcia; Celio X.C. Santos; Suwannee Thet; Eiichiro Mori; Michael Kinter; Paul M. Rindler; Serena Zacchigna; Shibani Mukherjee; David J. Chen; Ahmed I. Mahmoud; Mauro Giacca; Peter S. Rabinovitch; Asaithamby Aroumougame; Ajay M. Shah; Luke I. Szweda; Hesham A. Sadek

The mammalian heart has a remarkable regenerative capacity for a short period of time after birth, after which the majority of cardiomyocytes permanently exit cell cycle. We sought to determine the primary postnatal event that results in cardiomyocyte cell-cycle arrest. We hypothesized that transition to the oxygen-rich postnatal environment is the upstream signal that results in cell-cycle arrest of cardiomyocytes. Here, we show that reactive oxygen species (ROS), oxidative DNA damage, and DNA damage response (DDR) markers significantly increase in the heart during the first postnatal week. Intriguingly, postnatal hypoxemia, ROS scavenging, or inhibition of DDR all prolong the postnatal proliferative window of cardiomyocytes, whereas hyperoxemia and ROS generators shorten it. These findings uncover a protective mechanism that mediates cardiomyocyte cell-cycle arrest in exchange for utilization of oxygen-dependent aerobic metabolism. Reduction of mitochondrial-dependent oxidative stress should be an important component of cardiomyocyte proliferation-based therapeutic approaches.


Circulation Research | 2010

Myocardin-Related Transcription Factor-A Controls Myofibroblast Activation and Fibrosis in Response to Myocardial Infarction

Eric M. Small; Jeffrey E. Thatcher; Lillian B. Sutherland; Hideyuki Kinoshita; Robert D. Gerard; James A. Richardson; J. Michael DiMaio; Hesham A. Sadek; Koichiro Kuwahara; Eric N. Olson

Rationale: Myocardial infarction (MI) results in loss of cardiac myocytes in the ischemic zone of the heart, followed by fibrosis and scar formation, which diminish cardiac contractility and impede angiogenesis and repair. Myofibroblasts, a specialized cell type that switches from a fibroblast-like state to a contractile, smooth muscle-like state, are believed to be primarily responsible for fibrosis of the injured heart and other tissues, although the transcriptional mediators of fibrosis and myofibroblast activation remain poorly defined. Myocardin-related transcription factors (MRTFs) are serum response factor (SRF) cofactors that promote a smooth muscle phenotype and are emerging as components of stress-responsive signaling. Objective: We aimed to examine the effect of MRTF-A on cardiac remodeling and fibrosis. Methods and Results: Here, we show that MRTF-A controls the expression of a fibrotic gene program that includes genes involved in extracellular matrix production and smooth muscle cell differentiation in the heart. In MRTF-A–null mice, fibrosis and scar formation following MI or angiotensin II treatment are dramatically diminished compared with wild-type littermates. This protective effect of MRTF-A deletion is associated with a reduction in expression of fibrosis-associated genes, including collagen 1a2, a direct transcriptional target of SRF/MRTF-A. Conclusions: We conclude that MRTF-A regulates myofibroblast activation and fibrosis in response to the renin–angiotensin system and post-MI remodeling.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Cardiogenic small molecules that enhance myocardial repair by stem cells

Hesham A. Sadek; Britta Hannack; Elizabeth Choe; Jessica Wang; Shuaib Latif; Mary G. Garry; Daniel J. Garry; Jamie Longgood; Doug E. Frantz; Eric N. Olson; Jenny Hsieh; Jay W. Schneider

The clinical success of stem cell therapy for myocardial repair hinges on a better understanding of cardiac fate mechanisms. We have identified small molecules involved in cardiac fate by screening a chemical library for activators of the signature gene Nkx2.5, using a luciferase knockin bacterial artificial chromosome (BAC) in mouse P19CL6 pluripotent stem cells. We describe a family of sulfonyl-hydrazone (Shz) small molecules that can trigger cardiac mRNA and protein expression in a variety of embryonic and adult stem/progenitor cells, including human mobilized peripheral blood mononuclear cells (M-PBMCs). Small-molecule-enhanced M-PBMCs engrafted into the rat heart in proximity to an experimental injury improved cardiac function better than control cells. Recovery of cardiac function correlated with persistence of viable human cells, expressing human-specific cardiac mRNAs and proteins. Shz small molecules are promising starting points for drugs to promote myocardial repair/regeneration by activating cardiac differentiation in M-PBMCs.

Collaboration


Dive into the Hesham A. Sadek's collaboration.

Top Co-Authors

Avatar

Ahmed I. Mahmoud

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Wataru Kimura

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eric N. Olson

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shalini Muralidhar

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Cheng Cheng Zhang

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Suwannee Thet

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Diana C. Canseco

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Fatih Kocabas

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar

Joseph A. Garcia

University of Texas Southwestern Medical Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge