Network


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

Hotspot


Dive into the research topics where Isabella M. Grumbach is active.

Publication


Featured researches published by Isabella M. Grumbach.


Nature Medicine | 2011

Oxidation of CaMKII determines the cardiotoxic effects of aldosterone

B. Julie He; Mei Ling A Joiner; Madhu V. Singh; Elizabeth D. Luczak; Paari Dominic Swaminathan; Olha M. Koval; William Kutschke; Chantal Allamargot; Jinying Yang; Xiaoqun Guan; Kathy Zimmerman; Isabella M. Grumbach; Robert M. Weiss; Douglas R. Spitz; Curt D. Sigmund; W. Matthijs Blankesteijn; Stephane Heymans; Peter J. Mohler; Mark E. Anderson

Excessive activation of the β-adrenergic, angiotensin II (Ang II) and aldosterone signaling pathways promotes mortality after myocardial infarction, and antagonists targeting these pathways are core therapies for treating this condition. Catecholamines and Ang II activate the multifunctional Ca2+/calmodulin-dependent protein kinase II (CaMKII), the inhibition of which prevents isoproterenol-mediated and Ang II–mediated cardiomyopathy. Here we show that aldosterone exerts direct toxic actions on myocardium by oxidative activation of CaMKII, causing cardiac rupture and increased mortality in mice after myocardial infarction. Aldosterone induces CaMKII oxidation by recruiting NADPH oxidase, and this oxidized and activated CaMKII promotes matrix metalloproteinase 9 (MMP9) expression in cardiomyocytes. Myocardial CaMKII inhibition, overexpression of methionine sulfoxide reductase A (an enzyme that reduces oxidized CaMKII) or NADPH oxidase deficiency prevented aldosterone-enhanced cardiac rupture after myocardial infarction. These findings show that oxidized myocardial CaMKII mediates the cardiotoxic effects of aldosterone on the cardiac matrix and establish CaMKII as a nodal signal for the neurohumoral pathways associated with poor outcomes after myocardial infarction.


Journal of Clinical Investigation | 2011

Oxidized CaMKII causes cardiac sinus node dysfunction in mice

Paari Dominic Swaminathan; Anil Purohit; Siddarth Soni; Niels Voigt; Madhu V. Singh; Alexey V. Glukhov; Zhan Gao; B. Julie He; Elizabeth D. Luczak; Mei Ling A Joiner; William Kutschke; Jinying Yang; J. Kevin Donahue; Robert M. Weiss; Isabella M. Grumbach; Masahiro Ogawa; Peng Sheng Chen; Igor R. Efimov; Dobromir Dobrev; Peter J. Mohler; Thomas J. Hund; Mark E. Anderson

Sinus node dysfunction (SND) is a major public health problem that is associated with sudden cardiac death and requires surgical implantation of artificial pacemakers. However, little is known about the molecular and cellular mechanisms that cause SND. Most SND occurs in the setting of heart failure and hypertension, conditions that are marked by elevated circulating angiotensin II (Ang II) and increased oxidant stress. Here, we show that oxidized calmodulin kinase II (ox-CaMKII) is a biomarker for SND in patients and dogs and a disease determinant in mice. In wild-type mice, Ang II infusion caused sinoatrial nodal (SAN) cell oxidation by activating NADPH oxidase, leading to increased ox-CaMKII, SAN cell apoptosis, and SND. p47-/- mice lacking functional NADPH oxidase and mice with myocardial or SAN-targeted CaMKII inhibition were highly resistant to SAN apoptosis and SND, suggesting that ox-CaMKII-triggered SAN cell death contributed to SND. We developed a computational model of the sinoatrial node that showed that a loss of SAN cells below a critical threshold caused SND by preventing normal impulse formation and propagation. These data provide novel molecular and mechanistic information to understand SND and suggest that targeted CaMKII inhibition may be useful for preventing SND in high-risk patients.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Calmodulin kinase II is required for fight or flight sinoatrial node physiology

Yuejin Wu; Zhan Gao; Biyi Chen; Olha M. Koval; Madhu V. Singh; Xiaoqun Guan; Thomas J. Hund; William Kutschke; Satyam Sarma; Isabella M. Grumbach; Xander H.T. Wehrens; Peter J. Mohler; Long-Sheng Song; Mark E. Anderson

The best understood “fight or flight” mechanism for increasing heart rate (HR) involves activation of a cyclic nucleotide-gated ion channel (HCN4) by β-adrenergic receptor (βAR) agonist stimulation. HCN4 conducts an inward “pacemaker” current (If) that increases the sinoatrial nodal (SAN) cell membrane diastolic depolarization rate (DDR), leading to faster SAN action potential generation. Surprisingly, HCN4 knockout mice were recently shown to retain physiological HR increases with isoproterenol (ISO), suggesting that other If-independent pathways are critical to SAN fight or flight responses. The multifunctional Ca2+ and calmodulin-dependent protein kinase II (CaMKII) is a downstream signal in the βAR pathway that activates Ca2+ homeostatic proteins in ventricular myocardium. Mice with genetic, myocardial and SAN cell CaMKII inhibition have significantly slower HRs than controls during stress, leading us to hypothesize that CaMKII actions on SAN Ca2+ homeostasis are critical for βAR agonist responses in SAN. Here we show that CaMKII mediates ISO HR increases by targeting SAN cell Ca2+ homeostasis. CaMKII inhibition prevents ISO effects on SAN Ca2+ uptake and release from intracellular sarcoplasmic reticulum (SR) stores that are necessary for increasing DDR. CaMKII inhibition has no effect on the ISO response in SAN cells when SR Ca2+ release is disabled and CaMKII inhibition is only effective at slowing HRs during βAR stimulation. These studies show the tightly coupled, but previously unanticipated, relationship of CaMKII to the βAR pathway in fight or flight physiology and establish CaMKII as a critical signaling molecule for physiological HR responses to catecholamines.


Proceedings of the National Academy of Sciences of the United States of America | 2010

CaV1.2 β-subunit coordinates CaMKII-triggered cardiomyocyte death and afterdepolarizations

Olha M. Koval; Xiaoquan Guan; Yuejin Wu; Mei Ling A Joiner; Zhan Gao; Biyi Chen; Isabella M. Grumbach; Elizabeth D. Luczak; Roger J. Colbran; Long-Sheng Song; Thomas J. Hund; Peter J. Mohler; Mark E. Anderson

Excessive activation of calmodulin kinase II (CaMKII) causes arrhythmias and heart failure, but the cellular mechanisms for CaMKII-targeted proteins causing disordered cell membrane excitability and myocardial dysfunction remain uncertain. Failing human cardiomyocytes exhibit increased CaMKII and voltage-gated Ca2+ channel (CaV1.2) activity, and enhanced expression of a specific CaV1.2 β-subunit protein isoform (β2a). We recently identified CaV1.2 β2a residues critical for CaMKII phosphorylation (Thr 498) and binding (Leu 493), suggesting the hypothesis that these amino acids are crucial for cardiomyopathic consequences of CaMKII signaling. Here we show WT β2a expression causes cellular Ca2+ overload, arrhythmia-triggering cell membrane potential oscillations called early afterdepolarizations (EADs), and premature death in paced adult rabbit ventricular myocytes. Prevention of intracellular Ca2+ release by ryanodine or global cellular CaMKII inhibition reduced EADs and improved cell survival to control levels in WT β2a-expressing ventricular myocytes. In contrast, expression of β2a T498A or L493A mutants mimicked the protective effects of ryanodine or global cellular CaMKII inhibition by reducing Ca2+ entry through CaV1.2 and inhibiting EADs. Furthermore, CaV1.2 currents recorded from cells overexpressing CaMKII phosphorylation- or binding-incompetent β2a subunits were incapable of entering a CaMKII-dependent high-activity gating mode (mode 2), indicating that β2a Thr 498 and Leu 493 are required for CaV1.2 activation by CaMKII in native cells. These data show that CaMKII binding and phosphorylation sites on β2a are concise but pivotal components of a molecular and biophysical and mechanism for EADs and impaired survival in adult cardiomyocytes.


Hypertension | 2012

Sildenafil prevents and reverses transverse-tubule remodeling and Ca(2+) handling dysfunction in right ventricle failure induced by pulmonary artery hypertension.

Yu Ping Xie; Biyi Chen; Philip N. Sanders; Ang Guo; Yue Li; Kathy Zimmerman; Lie Cheng Wang; Robert M. Weiss; Isabella M. Grumbach; Mark E. Anderson; Long-Sheng Song

Right ventricular (RV) failure (RVF) is the main cause of death in patients with pulmonary artery hypertension (PAH). Sildenafil, a phosphodiesterase type 5 inhibitor, was approved recently for treatment of PAH patients. However, the mechanisms underlying RV contractile malfunction and the benefits of sildenafil on RV function are not well understood. We aimed to investigate the following: (1) the ultrastructural and excitation-contraction coupling alterations underlying PAH-induced RVF; (2) whether the ultrastructural changes are reversible; and (3) the mechanisms underlying the therapeutic benefits of sildenafil in PAH-RVF. We used a single injection of monocrotaline in Wistar rats to induce pulmonary vascular proliferation, which led to PAH and RVF. RV myocytes displayed severe transverse (T)-tubule loss and disorganization, as well as blunted and dys-synchronous sarcoplasmic reticulum Ca2+ release. Sildenafil prevented and reversed the monocrotaline-induced PAH and LV filling impairment. Early intervention with sildenafil prevented RV hypertrophy and the development of RVF, T-tubule remodeling, and Ca2+ handling dysfunction. Although late treatment with sildenafil did not reverse RV hypertrophy in animals with established RVF, RV systolic function was improved. Furthermore, late intervention partially reversed both the impairment of myocyte T-tubule integrity and Ca2+ handling protein and sarcoplasmic reticulum Ca2+ release function in monocrotaline-treated rats. In conclusion, PAH-induced increase in RV afterload causes severe T-tubule remodeling and Ca2+ handling dysfunction in RV myocytes, leading to RV contractile failure. Sildenafil prevents and partially reverses ultrastructural, molecular, and functional remodeling of failing RV myocytes. Reversal of pathological T-tubule remodeling, although incomplete, is achievable without the regression of RV hypertrophy.


American Journal of Physiology-heart and Circulatory Physiology | 2010

Calmodulin kinase II is required for angiotensin II-mediated vascular smooth muscle hypertrophy

Hui Li; Weiwei Li; Arun K. Gupta; Peter J. Mohler; Mark E. Anderson; Isabella M. Grumbach

Despite our understanding that medial smooth muscle hypertrophy is a central feature of vascular remodeling, the molecular pathways underlying this pathology are still not well understood. Work over the past decade has illustrated a potential role for the multifunctional calmodulin-dependent kinase CaMKII in smooth muscle cell contraction, growth, and migration. Here we demonstrate that CaMKII is enriched in vascular smooth muscle (VSM) and that CaMKII inhibition blocks ANG II-dependent VSM cell hypertrophy in vitro and in vivo. Specifically, systemic CaMKII inhibition with KN-93 prevented ANG II-mediated hypertension and medial hypertrophy in vivo. Adenoviral transduction with the CaMKII peptide inhibitor CaMKIIN abrogated ANG II-induced VSM hypertrophy in vitro, which was augmented by overexpression of CaMKII-delta2. Finally, we identify the downstream signaling components critical for ANG II- and CaMKII-mediated VSM hypertrophy. Specifically, we demonstrate that CaMKII induces VSM hypertrophy by regulating histone deacetylase 4 (HDAC4) activity, thereby stimulating activity of the hypertrophic transcription factor MEF2. MEF2 transcription is activated by ANG II in vivo and abrogated by the CaMKII inhibitor KN-93. Together, our studies identify a complete pathway for ANG II-triggered arterial VSM hypertrophy and identify new potential therapeutic targets for chronic human hypertension.


American Journal of Respiratory Cell and Molecular Biology | 2015

Mitochondrial-Targeted Antioxidant Therapy Decreases Transforming Growth Factor-β–Mediated Collagen Production in a Murine Asthma Model

Omar A. Jaffer; A. Brent Carter; Philip N. Sanders; Megan E. Dibbern; Christopher J. Winters; Shubha Murthy; Alan J. Ryan; Adam G. Rokita; Anand M. Prasad; Joseph Zabner; Joel N. Kline; Isabella M. Grumbach; Mark E. Anderson

Asthma is a disease of acute and chronic inflammation in which cytokines play a critical role in orchestrating the allergic inflammatory response. IL-13 and transforming growth factor (TGF)-β promote fibrotic airway remodeling, a major contributor to disease severity. Improved understanding is needed, because current therapies are inadequate for suppressing development of airway fibrosis. IL-13 is known to stimulate respiratory epithelial cells to produce TGF-β, but the mechanism through which this occurs is unknown. Here, we tested the hypothesis that reactive oxygen species (ROS) are a critical signaling intermediary between IL-13 or allergen stimulation and TGF-β-dependent airway remodeling. We used cultured human bronchial epithelial cells and an in vivo mouse model of allergic asthma to map a pathway where allergens enhanced mitochondrial ROS, which is an essential upstream signal for TGF-β activation and enhanced collagen production and deposition in airway fibroblasts. We show that mitochondria in airway epithelium are an essential source of ROS that activate TGF-β expression and activity. TGF-β from airway epithelium stimulates collagen expression in fibroblasts, contributing to an early fibrotic response to allergen exposure in cultured human airway cells and in ovalbumin-challenged mice. Treatment with the mitochondrial-targeted antioxidant, (2-(2,2,6,6-Tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mitoTEMPO), significantly attenuated mitochondrial ROS, TGF-β, and collagen deposition in OVA-challenged mice and in cultured human epithelial cells. Our findings suggest that mitochondria are a critical source of ROS for promoting TGF-β activity that contributes to airway remodeling in allergic asthma. Mitochondrial-targeted antioxidants may be a novel approach for future asthma therapies.


Science Translational Medicine | 2013

CaMKII Is Essential for the Proasthmatic Effects of Oxidation

Philip N. Sanders; Olha M. Koval; Omar A. Jaffer; Anand M. Prasad; Thomas R. Businga; Jason A. Scott; Patrick J. Hayden; Elizabeth D. Luczak; David D. Dickey; Chantal Allamargot; Alicia K. Olivier; David K. Meyerholz; Alfred J. Robison; Danny G. Winder; Timothy S. Blackwell; Ryszard Dworski; David Sammut; Brett A. Wagner; Garry R. Buettner; Robert M. Pope; Francis J. Miller; Megan E. Dibbern; Hans Michael Haitchi; Peter J. Mohler; Peter H. Howarth; Joseph Zabner; Joel N. Kline; Isabella M. Grumbach; Mark E. Anderson

Ca2+/calmodulin-dependent protein kinase (CaMKII) transduces oxidative stress into asthma-related diseases. A Breath of Fresh Air for Asthma Patients Reactive oxygen species (ROS) have a bad reputation, and rightly so. They’ve been implicated in contributing to a wide swath of diseases, including coronary heart disease, cancer, and asthma. Indeed, asthma is an increasing public health burden—affecting 8.5% of the population in the United States alone. Now Sanders et al. find that oxidative activation of the Ca2+/calmodulin-dependent protein kinase (ox-CaMKII) may respond to ROS in lung epithelium and contribute to asthma pathogenesis. The authors observed that asthma patients have enhanced activation of ox-CaMKII in bronchial epithelium, which increases in response to inhaled antigen. Then they looked in two different mouse models of allergic asthma to examine the mechanistic connection. They found that blocking CaMKII either genetically or with a small-molecule inhibitor could alleviate ROS-mediated asthma progression. These data suggest that blocking CaMKII could be a new therapeutic strategy for asthma patients. Increased reactive oxygen species (ROS) contribute to asthma, but little is known about the molecular mechanisms connecting increased ROS with characteristic features of asthma. We show that enhanced oxidative activation of the Ca2+/calmodulin-dependent protein kinase (ox-CaMKII) in bronchial epithelium positively correlates with asthma severity and that epithelial ox-CaMKII increases in response to inhaled allergens in patients. We used mouse models of allergic airway disease induced by ovalbumin (OVA) or Aspergillus fumigatus (Asp) and found that bronchial epithelial ox-CaMKII was required to increase a ROS- and picrotoxin-sensitive Cl− current (ICl) and MUC5AC expression, upstream events in asthma progression. Allergen challenge increased epithelial ROS by activating NADPH oxidases. Mice lacking functional NADPH oxidases due to knockout of p47 and mice with epithelial-targeted transgenic expression of a CaMKII inhibitory peptide or wild-type mice treated with inhaled KN-93, an experimental small-molecule CaMKII antagonist, were protected against increases in ICl, MUC5AC expression, and airway hyperreactivity to inhaled methacholine. Our findings support the view that CaMKII is a ROS-responsive, pluripotent proasthmatic signal and provide proof-of-concept evidence that CaMKII is a therapeutic target in asthma.


American Journal of Physiology-heart and Circulatory Physiology | 2012

The multifunctional Ca2+/calmodulin-dependent kinase II regulates vascular smooth muscle migration through matrix metalloproteinase 9

Jason A. Scott; Litao Xie; Hui Li; Weiwei Li; Julie B. He; Philip N. Sanders; A. Brent Carter; Johannes Backs; Mark E. Anderson; Isabella M. Grumbach

The multifunctional CaMKII has been implicated in vascular smooth muscle cell (VSMC) migration, but little is known regarding its downstream targets that mediate migration. Here, we examined whether CaMKII regulates migration through modulation of matrix metalloproteinase 9 (MMP9). Using CaMKIIδ(-/-) mice as a model system, we evaluated migration and MMP9 regulation in vitro and in vivo. After ligation of the common carotid artery, CaMKII was activated in the neointima as determined by oxidation and autophosphorylation. We found that MMP9 was robustly expressed in the neointima and adventitia of carotid-ligated wild-type (WT) mice but was barely detectable in CaMKIIδ(-/-) mice. The perimeter of the external elastic lamina, a correlate of migration-related outward remodeling, was increased in WT but not in CaMKIIδ(-/-) mice. Migration induced by serum, platelet-derived growth factor, and tumor necrosis factor-α (TNF-α) was significantly decreased in CaMKIIδ(-/-) as compared with WT VSMCs, but migration was rescued with adenoviral overexpression of MMP9 in CaMKIIδ(-/-) VSMCs. Likewise, overexpression of CaMKIIδ in CaMKIIδ(-/-) VSMCs increased migration, whereas an oxidation-resistant mutant of CaMKIIδ did not. TNF-α strongly induced CaMKII oxidation and autophosphorylation as well as MMP9 activity, mRNA, and protein levels in WT, but not in CaMKIIδ(-/-) VSMC. Surprisingly, TNF-α strongly induced MMP9 promoter activity in WT and CaMKIIδ(-/-) VSMC. However, the MMP9 mRNA stability was significantly decreased in CaMKIIδ(-/-) VSMC. Our data demonstrate that CaMKII promotes VSMC migration through posttranscriptional regulation of MMP9 and suggest that CaMKII effects on MMP9 expression may be a therapeutic pathway in vascular injury.


Vascular Pharmacology | 2014

Oxidative activation of the Ca2+/calmodulin-dependent protein kinase II (CaMKII) regulates vascular smooth muscle migration and apoptosis

Linda J. Zhu; Paula J. Klutho; Jason Scott; Litao Xie; Elizabeth D. Luczak; Megan E. Dibbern; Anand Prasad; Omar A. Jaffer; Ashlee N. Venema; Emily K. Nguyen; Xiaoqun Guan; Mark E. Anderson; Isabella M. Grumbach

Activation of the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and reactive oxygen species (ROS) promote neointimal hyperplasia after vascular injury. CaMKII can be directly activated by ROS through oxidation. In this study, we determined whether abolishing the oxidative activation site of CaMKII alters vascular smooth muscle cell (VCMC) proliferation, migration and apoptosis in vitro and neointimal formation in vivo. VSMC isolated from a knock-in mouse with oxidation-resistant CaMKIIδ (CaMKII M2V) displayed similar proliferation but decreased migration and apoptosis. Surprisingly, ROS production and expression of the NADPH oxidase subunits p47 and p22 were decreased in M2V VSMC, whereas superoxide dismutase 2 protein expression was upregulated. In vivo, after carotid artery ligation, no differences in neointimal size or remodeling were observed. In contrast to VSMC, CaMKII expression and autonomous activity were significantly higher in M2V compared to WT carotid arteries, suggesting that an autoregulatory mechanism determines CaMKII activity in vivo. Our findings demonstrate that preventing oxidative activation of CaMKII decreases migration and apoptosis in vitro and suggest that CaMKII regulates ROS production. Our study presents novel evidence that CaMKII expression in vivo is regulated by a negative feedback loop following oxidative activation.

Collaboration


Dive into the Isabella M. Grumbach's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge