Network


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

Hotspot


Dive into the research topics where Leonid V. Zingman is active.

Publication


Featured researches published by Leonid V. Zingman.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Inhibition of MCU forces extramitochondrial adaptations governing physiological and pathological stress responses in heart

Tyler P. Rasmussen; Yuejin Wu; Mei-ling A. Joiner; Olha M. Koval; Nicholas R. Wilson; Elizabeth D. Luczak; Qinchuan Wang; Biyi Chen; Zhan Gao; Zhiyong Zhu; Brett A. Wagner; Jamie Soto; Michael L. McCormick; William Kutschke; Robert M. Weiss; Liping Yu; Ryan L. Boudreau; E. Dale Abel; Fenghuang Zhan; Douglas R. Spitz; Garry R. Buettner; Long-Sheng Song; Leonid V. Zingman; Mark E. Anderson

Significance Mitochondrial Ca2+ is a fundamental signal that allows for adaptation to physiological stress but a liability during ischemia-reperfusion injury in heart. On one hand, mitochondrial Ca2+ entry coordinates energy supply and demand in myocardium by increasing the activity of matrix dehydrogenases to augment ATP production by oxidative phosphorylation. On the other hand, inhibiting mitochondrial Ca2+ overload is promulgated as a therapeutic approach to preserve myocardial tissue following ischemia-reperfusion injury. We developed a new mouse model of myocardial-targeted transgenic dominant-negative mitochondrial Ca2+ uniporter (MCU) expression to test consequences of chronic loss of MCU-mediated Ca2+ entry in heart. Here we show that MCU inhibition has unanticipated consequences on extramitochondrial pathways affecting oxygen utilization, cytoplasmic Ca2+ homeostasis, physiologic responses to stress, and pathologic responses to ischemia-reperfusion injury. Myocardial mitochondrial Ca2+ entry enables physiological stress responses but in excess promotes injury and death. However, tissue-specific in vivo systems for testing the role of mitochondrial Ca2+ are lacking. We developed a mouse model with myocardial delimited transgenic expression of a dominant negative (DN) form of the mitochondrial Ca2+ uniporter (MCU). DN-MCU mice lack MCU-mediated mitochondrial Ca2+ entry in myocardium, but, surprisingly, isolated perfused hearts exhibited higher O2 consumption rates (OCR) and impaired pacing induced mechanical performance compared with wild-type (WT) littermate controls. In contrast, OCR in DN-MCU–permeabilized myocardial fibers or isolated mitochondria in low Ca2+ were not increased compared with WT, suggesting that DN-MCU expression increased OCR by enhanced energetic demands related to extramitochondrial Ca2+ homeostasis. Consistent with this, we found that DN-MCU ventricular cardiomyocytes exhibited elevated cytoplasmic [Ca2+] that was partially reversed by ATP dialysis, suggesting that metabolic defects arising from loss of MCU function impaired physiological intracellular Ca2+ homeostasis. Mitochondrial Ca2+ overload is thought to dissipate the inner mitochondrial membrane potential (ΔΨm) and enhance formation of reactive oxygen species (ROS) as a consequence of ischemia-reperfusion injury. Our data show that DN-MCU hearts had preserved ΔΨm and reduced ROS during ischemia reperfusion but were not protected from myocardial death compared with WT. Taken together, our findings show that chronic myocardial MCU inhibition leads to previously unanticipated compensatory changes that affect cytoplasmic Ca2+ homeostasis, reprogram transcription, increase OCR, reduce performance, and prevent anticipated therapeutic responses to ischemia-reperfusion injury.


Annals of the New York Academy of Sciences | 2005

Administration of Allogenic Stem Cells Dosed to Secure Cardiogenesis and Sustained Infarct Repair

Atta Behfar; Denice M. Hodgson; Leonid V. Zingman; Satsuki Yamada; Garvan C. Kane; Alexey E. Alekseev; Michel Pucéat; Andre Terzic

Abstract: The mitotic capacity of heart muscle is too limited to fully substitute for cells lost following myocardial infarction. Emerging stem cell‐based strategies have been proposed to overcome the self‐renewal shortfall of native cardiomyocytes, yet there is limited evidence for their capability to achieve safe de novo cardiogenesis and repair. We present our recent experience in treating long‐term, infarcted hearts with embryonic stem cells, a prototype source for allogenic cell therapy. The cardiogenic potential of the engrafted murine embryonic stem cell colony was pre‐tested by in vitro differentiation, with derived cells positive for nuclear cardiac transcription factors, sarcomeric proteins and functional excitation‐contraction coupling. Eight weeks after infarct, rats were randomized into sham‐ or embryonic stem cell‐treated groups. Acellular sham controls or embryonic stem cells, engineered to express enhanced cyan fluorescent protein (ECFP) under control of the cardiac actin promoter, were injected through a 28‐gauge needle at three sites into the peri‐infarct zone for serial assessment of functional and structural impact. In contrast to results with sham‐treated animals, stem cell therapy yielded, over the 5‐month follow‐up period, new ECFP‐positive cardiomyocytes that integrated with the infarcted myocardium. The stem cell‐treated group showed a stable contractile performance benefit with normalization of myocardial architecture post infarction. Transition of embryonic stem cells into cardiomyocytes required host signaling to support cardiac‐specific differentiation and could result in tumorigenesis if the stem cell dose exceeded the hearts cardioinductive capacity. Supported by the host environment, proper dosing and administration of embryonic stem cells is thus here shown useful in the chronic management of cardiac injury promoting sustained repair.


Cardiovascular Research | 2013

Critical roles of junctophilin-2 in T-tubule and excitation–contraction coupling maturation during postnatal development

Biyi Chen; Ang Guo; Caimei Zhang; Rong Chen; Yanqi Zhu; Jiang Hong; William Kutschke; Kathy Zimmerman; Robert M. Weiss; Leonid V. Zingman; Mark E. Anderson; Xander H.T. Wehrens; Long-Sheng Song

AIMS Emerging evidence indicates a critical role for junctophilin-2 (JP2) in T-tubule integrity and assembly of cardiac dyads in adult ventricular myocytes. In the postnatal stage, one of the critical features of myocyte maturation is development of the T-tubule system, though the mechanisms remain poorly understood. In this study, we aim to determine whether JP2 is required for normal cardiac T-tubule maturation. METHODS AND RESULTS Using in situ confocal imaging of intact murine hearts, we found T-tubules were absent in both left- and right-ventricular myocytes at postnatal Day 8 and did not appear until Day 10. Quantification of T-tubule structural integrity using the T-tubule power (TT(power)) index revealed a progressive increase in TT(power) between postnatal Days 10 and 19. By postnatal Day 19, TT(power) was similar to that in adult murine cardiomyocytes, indicative of a nearly matured T-tubule network. JP2 levels increased dramatically during development, reaching levels observed in adult hearts by postnatal Day 14. Deficiency of JP2, using a mouse model in which a JP2-specific shRNA is expressed during embryonic development, severely impaired T-tubule maturation, with equivalent decreases in the left- and right-ventricular TT(power). We also detected a gradual increase in the density of transverse but not longitudinal tubules during development, and JP2 deficiency abolished the increase in the density of transverse elements. Alterations in T-tubules caused significant reduction in Ca(2+) transient amplitude and marked increase in Ca(2+) release dyssynchrony, Ca(2+) alternans, and spontaneous Ca(2+) waves, leading to contractile failure. CONCLUSION Our data identify a critical role for JP2 in T-tubule and excitation-contraction coupling maturation during development.


Journal of Molecular and Cellular Cardiology | 2011

Exercise-induced expression of cardiac ATP-sensitive potassium channels promotes action potential shortening and energy conservation.

Leonid V. Zingman; Zhiyong Zhu; Ana Sierra; Elizabeth Stepniak; Colin M.L. Burnett; Gennadiy Maksymov; Mark E. Anderson; William A. Coetzee; Denice M. Hodgson-Zingman

Physical activity is one of the most important determinants of cardiac function. The ability of the heart to increase delivery of oxygen and metabolic fuels relies on an array of adaptive responses necessary to match bodily demand while avoiding exhaustion of cardiac resources. The ATP-sensitive potassium (K(ATP)) channel has the unique ability to adjust cardiac membrane excitability in accordance with ATP and ADP levels, and up-regulation of its expression that occurs in response to exercise could represent a critical element of this adaption. However, the mechanism by which K(ATP) channel expression changes result in a beneficial effect on cardiac excitability and function remains to be established. Here, we demonstrate that an exercise-induced rise in K(ATP) channel expression enhanced the rate and magnitude of action potential shortening in response to heart rate acceleration. This adaptation in membrane excitability promoted significant reduction in cardiac energy consumption under escalating workloads. Genetic disruption of normal K(ATP) channel pore function abolished the exercise-related changes in action potential duration adjustment and caused increased cardiac energy consumption. Thus, an expression-driven enhancement in the K(ATP) channel-dependent membrane response to alterations in cardiac workload represents a previously unrecognized mechanism for adaptation to physical activity and a potential target for cardioprotection.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Musclin is an activity-stimulated myokine that enhances physical endurance.

Ekaterina Subbotina; Ana Sierra; Zhiyong Zhu; Zhan Gao; Siva Rama Krishna Koganti; Santiago Reyes; Elizabeth Stepniak; Susan A. Walsh; Michael R. Acevedo; Denice M. Hodgson-Zingman; Leonid V. Zingman

Significance Skeletal muscle is increasingly recognized as a secretory organ. Revealing the identity and function of myokines can improve our understanding of skeletal muscle function under sedentary or exercise conditions, as well as its coordination with other organs, tissues, and overall body metabolism. This study identifies musclin as an exercise-responsive myokine critical for skeletal muscle adaptation to physical activity. We develop a musclin-encoding gene (Ostn) knockout mouse, which allows us to determine a previously unrecognized physiologic function of musclin in regulation of skeletal muscle mitochondrial biogenesis and physical endurance. The demonstrated molecular mechanism for musclin-dependent skeletal muscle adaptation to exercise also transforms the perspective on natriuretic peptide signaling, particularly as it relates to physical activity and exercise-induced remodeling in different tissues. Exercise remains the most effective way to promote physical and metabolic wellbeing, but molecular mechanisms underlying exercise tolerance and its plasticity are only partially understood. In this study we identify musclin—a peptide with high homology to natriuretic peptides (NP)—as an exercise-responsive myokine that acts to enhance exercise capacity in mice. We use human primary myoblast culture and in vivo murine models to establish that the activity-related production of musclin is driven by Ca2+-dependent activation of Akt1 and the release of musclin-encoding gene (Ostn) transcription from forkhead box O1 transcription factor inhibition. Disruption of Ostn and elimination of musclin secretion in mice results in reduced exercise tolerance that can be rescued by treatment with recombinant musclin. Reduced exercise capacity in mice with disrupted musclin signaling is associated with a trend toward lower levels of plasma atrial NP (ANP) and significantly smaller levels of cyclic guanosine monophosphate (cGMP) and peroxisome proliferator-activated receptor gamma coactivator 1-α in skeletal muscles after exposure to exercise. Furthermore, in agreement with the established musclin ability to interact with NP clearance receptors, but not with NP guanyl cyclase-coupled signaling receptors, we demonstrate that musclin enhances cGMP production in cultured myoblasts only when applied together with ANP. Elimination of the activity-related musclin-dependent boost of ANP/cGMP signaling results in significantly lower maximum aerobic capacity, mitochondrial protein content, respiratory complex protein expression, and succinate dehydrogenase activity in skeletal muscles. Together, these data indicate that musclin enhances physical endurance by promoting mitochondrial biogenesis.


Channels | 2007

Calmodulin kinase ii inhibition enhances ischemic preconditioning by augmenting ATP-sensitive K+ current

Jingdong Li; Céline Marionneau; Olha M. Koval; Leonid V. Zingman; Peter J. Mohler; Jeanne M. Nerbonne; Mark E. Anderson

Mice with genetic inhibition (AC3-I) of the multifunctional Ca2+/calmodulin dependent protein kinase II (CaMKII) have improved cardiomyocyte survival after ischemia. Some K+ currents are up-regulated in AC3-I hearts, but it is unknown if CaMKII inhibition increases the ATP sensitive K+ current (IKATP) that underlies ischemic preconditioning (IP) and confers resistance to ischemia. We hypothesized increased IKATP was part of the mechanism for improved ventricular myocyte survival during ischemia in AC3-I mice. AC3-I hearts were protected against global ischemia due to enhanced IP compared to wild type (WT) and transgenic control (AC3-C) hearts. IKATP was significantly increased, while the negative regulatory dose-dependence of ATP was unchanged in AC3-I compared to WT and AC3-C ventricular myocytes, suggesting that CaMKII inhibition increased the number of functional IKATP channels available for IP. We measured increased sarcolemmal Kir6.2, a pore-forming IKATP subunit, but not a change in total Kir6.2 in cell lysates or single channel IKATP opening probability from AC3-I compared to WT and AC3-C ventricles, showing CaMKII inhibition increased sarcolemmal IKATP channel expression. There were no differences in mRNA for genes encoding IKATP channel subunits in AC3-I, WT and AC3-C ventricles. The IKATP opener pinacidil (100 M) reduced MI area in WT to match AC3-I hearts, while the IKATP antagonist HMR1098 (30 M) increased MI area to an equivalent level in all groups, indicating that increased IKATP and augmented IP are important for reduced ischemic cell death in AC3-I hearts. Our study results show CaMKII inhibition enhances beneficial effects of IP by increasing IKATP.


Biochemical and Biophysical Research Communications | 2011

Reduction in number of sarcolemmal KATP channels slows cardiac action potential duration shortening under hypoxia

Zhiyong Zhu; Colin M.L. Burnett; Gennadiy Maksymov; Elizabeth Stepniak; Ana Sierra; Ekaterina Subbotina; Mark E. Anderson; William A. Coetzee; Denice M. Hodgson-Zingman; Leonid V. Zingman

The cardiovascular system operates under demands ranging from conditions of rest to extreme stress. One mechanism of cardiac stress tolerance is action potential duration shortening driven by ATP-sensitive potassium (K(ATP)) channels. K(ATP) channel expression has a significant physiologic impact on action potential duration shortening and myocardial energy consumption in response to physiologic heart rate acceleration. However, the effect of reduced channel expression on action potential duration shortening in response to severe metabolic stress is yet to be established. Here, transgenic mice with myocardium-specific expression of a dominant negative K(ATP) channel subunit were compared with littermate controls. Evaluation of K(ATP) channel whole cell current and channel number/patch was assessed by patch clamp in isolated ventricular cardiomyocytes. Monophasic action potentials were monitored in retrogradely perfused, isolated hearts during the transition to hypoxic perfusate. An 80-85% reduction in cardiac K(ATP) channel current density results in a similar magnitude, but significantly slower rate, of shortening of the ventricular action potential duration in response to severe hypoxia, despite no significant difference in coronary flow. Therefore, the number of functional cardiac sarcolemmal K(ATP) channels is a critical determinant of the rate of adaptation of myocardial membrane excitability, with implications for optimization of cardiac energy consumption and consequent cardioprotection under conditions of severe metabolic stress.


Journal of Biological Chemistry | 2013

Regulation of Cardiac ATP-sensitive Potassium Channel Surface Expression by Calcium/Calmodulin-dependent Protein Kinase II

Ana Sierra; Zhiyong Zhu; Nicolas Sapay; Vikas Sharotri; Crystal F. Kline; Elizabeth D. Luczak; Ekaterina Subbotina; Asipu Sivaprasadarao; Peter M. Snyder; Peter J. Mohler; Mark E. Anderson; Michel Vivaudou; Leonid V. Zingman; Denice M. Hodgson-Zingman

Background: Surface expression of cardiac ATP-sensitive potassium (KATP) channels impacts cellular energy homeostasis. Results: Activation of calcium/calmodulin-dependent protein kinase II (CaMKII) results in KATP channel internalization, requiring specific motifs on the Kir6.2 channel subunit. Conclusion: CaMKII phosphorylation of Kir6.2 promotes endocytosis of cardiac KATP channels. Significance: This mechanism reveals new targets to improve cardiac energy efficiency and stress resistance. Cardiac ATP-sensitive potassium (KATP) channels are key sensors and effectors of the metabolic status of cardiomyocytes. Alteration in their expression impacts their effectiveness in maintaining cellular energy homeostasis and resistance to injury. We sought to determine how activation of calcium/calmodulin-dependent protein kinase II (CaMKII), a central regulator of calcium signaling, translates into reduced membrane expression and current capacity of cardiac KATP channels. We used real-time monitoring of KATP channel current density, immunohistochemistry, and biotinylation studies in isolated hearts and cardiomyocytes from wild-type and transgenic mice as well as HEK cells expressing wild-type and mutant KATP channel subunits to track the dynamics of KATP channel surface expression. Results showed that activation of CaMKII triggered dynamin-dependent internalization of KATP channels. This process required phosphorylation of threonine at 180 and 224 and an intact 330YSKF333 endocytosis motif of the KATP channel Kir6.2 pore-forming subunit. A molecular model of the μ2 subunit of the endocytosis adaptor protein, AP2, complexed with Kir6.2 predicted that μ2 docks by interaction with 330YSKF333 and Thr-180 on one and Thr-224 on the adjacent Kir6.2 subunit. Phosphorylation of Thr-180 and Thr-224 would favor interactions with the corresponding arginine- and lysine-rich loops on μ2. We concluded that calcium-dependent activation of CaMKII results in phosphorylation of Kir6.2, which promotes endocytosis of cardiac KATP channel subunits. This mechanism couples the surface expression of cardiac KATP channels with calcium signaling and reveals new targets to improve cardiac energy efficiency and stress resistance.


The Journal of General Physiology | 2014

Sarcolemmal ATP-sensitive potassium channels modulate skeletal muscle function under low-intensity workloads

Zhiyong Zhu; Ana Sierra; Colin M.L. Burnett; Biyi Chen; Ekaterina Subbotina; Siva Rama Krishna Koganti; Zhan Gao; Yuejin Wu; Mark E. Anderson; Long-Sheng Song; David J. Goldhamer; William A. Coetzee; Denice M. Hodgson-Zingman; Leonid V. Zingman

ATP-sensitive potassium (KATP) channels have the unique ability to adjust membrane excitability and functions in accordance with the metabolic status of the cell. Skeletal muscles are primary sites of activity-related energy consumption and have KATP channels expressed in very high density. Previously, we demonstrated that transgenic mice with skeletal muscle–specific disruption of KATP channel function consume more energy than wild-type littermates. However, how KATP channel activation modulates skeletal muscle resting and action potentials under physiological conditions, particularly low-intensity workloads, and how this can be translated to muscle energy expenditure are yet to be determined. Here, we developed a technique that allows evaluation of skeletal muscle excitability in situ, with minimal disruption of the physiological environment. Isometric twitching of the tibialis anterior muscle at 1 Hz was used as a model of low-intensity physical activity in mice with normal and genetically disrupted KATP channel function. This workload was sufficient to induce KATP channel opening, resulting in membrane hyperpolarization as well as reduction in action potential overshoot and duration. Loss of KATP channel function resulted in increased calcium release and aggravated activity-induced heat production. Thus, this study identifies low-intensity workload as a trigger for opening skeletal muscle KATP channels and establishes that this coupling is important for regulation of myocyte function and thermogenesis. These mechanisms may provide a foundation for novel strategies to combat metabolic derangements when energy conservation or dissipation is required.


Clinical Pharmacology & Therapeutics | 2016

Morpholino‐driven gene editing: A new horizon for disease treatment and prevention

Ekaterina Subbotina; Srk Koganti; Denice M. Hodgson-Zingman; Leonid V. Zingman

The development of genetic and molecular biology tools permitting the connection of specific genes to their functions has accelerated our understanding of molecular pathways underlying health and disease. The resulting gains in knowledge have propelled gene targeting to the forefront of promising therapeutic strategies. Here we discuss the uniquely powerful and adaptable approach of morpholino-driven modification of normal and mutant gene expression as a pathway to health.

Collaboration


Dive into the Leonid V. Zingman's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zhiyong Zhu

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ana Sierra

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ekaterina Subbotina

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Mark E. Anderson

Johns Hopkins University School of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Siva Rama Krishna Koganti

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Biyi Chen

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Colin M.L. Burnett

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge