31 results on '"Ranek MJ"'
Search Results
2. Phosphodiesterases: Evolving Concepts and Implications for Human Therapeutics.
- Author
-
Kelly ED, Ranek MJ, Zhang M, Kass DA, and Muller GK
- Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides. While the 11 PDE subfamilies share common features, key differences confer signaling specificity. The differences include substrate selectivity, enzymatic activity regulation, tissue expression, and subcellular localization. Selective inhibitors of each subfamily have elucidated the protean role of PDEs on normal cell function. PDEs are also linked to diseases, some of which affect the immune, cardiac, and vascular systems. Selective PDE inhibitors are clinically used to treat these specific disorders. Ongoing preclinical studies and clinical trials are likely to lead to the approval of additional PDE-targeting drugs for therapy in human disease. In this review, we discuss the structure and function of PDEs and examine current and evolving therapeutic uses of PDE inhibitors, highlighting their mechanisms and innovative applications that could further leverage this crucial family of enzymes in clinical settings.
- Published
- 2024
- Full Text
- View/download PDF
3. Growth hormone-releasing hormone receptor antagonist MIA-602 attenuates cardiopulmonary injury induced by BSL-2 rVSV-SARS-CoV-2 in hACE2 mice.
- Author
-
Condor Capcha JM, Kamiar A, Robleto E, Saad AG, Cui T, Wong A, Villano J, Zhong W, Pekosz A, Medina E, Cai R, Sha W, Ranek MJ, Webster KA, Schally AV, Jackson RM, and Shehadeh LA
- Subjects
- Mice, Male, Female, Animals, SARS-CoV-2, Lung pathology, Inflammation pathology, Weight Loss, Mice, Transgenic, Disease Models, Animal, COVID-19 pathology, Respiratory Distress Syndrome pathology
- Abstract
COVID-19 pneumonia causes acute lung injury and acute respiratory distress syndrome (ALI/ARDS) characterized by early pulmonary endothelial and epithelial injuries with altered pulmonary diffusing capacity and obstructive or restrictive physiology. Growth hormone-releasing hormone receptor (GHRH-R) is expressed in the lung and heart. GHRH-R antagonist, MIA-602, has been reported to modulate immune responses to bleomycin lung injury and inflammation in granulomatous sarcoidosis. We hypothesized that MIA-602 would attenuate rVSV-SARS-CoV-2-induced pulmonary dysfunction and heart injury in a BSL-2 mouse model. Male and female K18-h ACE2 tg mice were inoculated with SARS-CoV-2/USA-WA1/2020, BSL-2-compliant recombinant VSV-eGFP-SARS-CoV-2-Spike (rVSV-SARS-CoV-2), or PBS, and lung viral load, weight loss, histopathology, and gene expression were compared. K18-h ACE2 tg mice infected with rVSV-SARS-CoV-2 were treated daily with subcutaneous MIA-602 or vehicle and conscious, unrestrained plethysmography performed on days 0, 3, and 5 (n = 7 to 8). Five days after infection mice were killed, and blood and tissues collected for histopathology and protein/gene expression. Both native SARS-CoV-2 and rVSV-SARS-CoV-2 presented similar patterns of weight loss, infectivity (~60%), and histopathologic changes. Daily treatment with MIA-602 conferred weight recovery, reduced lung perivascular inflammation/pneumonia, and decreased lung/heart ICAM-1 expression compared to vehicle. MIA-602 rescued altered respiratory rate, increased expiratory parameters (Te, PEF, EEP), and normalized airflow parameters (Penh and Rpef) compared to vehicle, consistent with decreased airway inflammation. RNASeq followed by protein analysis revealed heightened levels of inflammation and end-stage necroptosis markers, including ZBP1 and pMLKL induced by rVSV-SARS-CoV-2, that were normalized by MIA-602 treatment, consistent with an anti-inflammatory and pro-survival mechanism of action in this preclinical model of COVID-19 pneumonia., Competing Interests: Competing interests statement:A.V.S. and R.M.J. are listed as co-inventors on patents of GHRH analogs, which were assigned to the University of Miami and Veterans Affairs Department.
- Published
- 2023
- Full Text
- View/download PDF
4. TSC2 S1365A mutation potently regulates CD8+ T cell function and differentiation and improves adoptive cellular cancer therapy.
- Author
-
Patel CH, Dong Y, Koleini N, Wang X, Dunkerly-Eyring BL, Wen J, Ranek MJ, Bartle LM, Henderson DB, Sagert J, Kass DA, and Powell JD
- Subjects
- Mice, Humans, Animals, Mechanistic Target of Rapamycin Complex 1, CD8-Positive T-Lymphocytes, Mutation, Cell Differentiation, Tumor Microenvironment, Tuberous Sclerosis
- Abstract
MTORC1 integrates signaling from the immune microenvironment to regulate T cell activation, differentiation, and function. TSC2 in the tuberous sclerosis complex tightly regulates mTORC1 activation. CD8+ T cells lacking TSC2 have constitutively enhanced mTORC1 activity and generate robust effector T cells; however, sustained mTORC1 activation prevents generation of long-lived memory CD8+ T cells. Here we show that manipulating TSC2 at Ser1365 potently regulated activated but not basal mTORC1 signaling in CD8+ T cells. Unlike nonstimulated TSC2-KO cells, CD8+ T cells expressing a phosphosilencing mutant TSC2-S1365A (TSC2-SA) retained normal basal mTORC1 activity. PKC and T cell receptor (TCR) stimulation induced TSC2 S1365 phosphorylation, and preventing this with the SA mutation markedly increased mTORC1 activation and T cell effector function. Consequently, SA CD8+ T cells displayed greater effector responses while retaining their capacity to become long-lived memory T cells. SA CD8+ T cells also displayed enhanced effector function under hypoxic and acidic conditions. In murine and human solid-tumor models, SA CD8+ T cells used as adoptive cell therapy displayed greater antitumor immunity than WT CD8+ T cells. These findings reveal an upstream mechanism to regulate mTORC1 activity in T cells. The TSC2-SA mutation enhanced both T cell effector function and long-term persistence/memory formation, supporting an approach to engineer better CAR-T cells for treating cancer.
- Published
- 2023
- Full Text
- View/download PDF
5. Transient receptor potential canonical type 6 (TRPC6) O-GlcNAcylation at Threonine-221 plays potent role in channel regulation.
- Author
-
Mishra S, Ma J, McKoy D, Sasaki M, Farinelli F, Page RC, Ranek MJ, Zachara N, and Kass DA
- Abstract
Transient receptor potential canonical type 6 (TRPC6) is a non-voltage-gated channel that principally conducts calcium. Elevated channel activation contributes to fibrosis, hypertrophy, and proteinuria, often coupled to stimulation of nuclear factor of activated T-cells (NFAT). TRPC6 is post-translationally regulated, but a role for O-linked β-N-acetyl glucosamine (O-GlcNAcylation) as elevated by diabetes, is unknown. Here we show TRPC6 is constitutively O-GlcNAcylated at Ser14, Thr70, and Thr221 in the N-terminus ankryn-4 (AR4) and linker (LH1) domains. Mutagenesis to alanine reveals T221 as a critical controller of resting TRPC6 conductance, and associated NFAT activity and pro-hypertrophic signaling. T→A mutations at sites homologous in closely related TRPC3 and TRPC7 also increases their activity. Molecular modeling predicts interactions between Thr221- O -GlcNAc and Ser199, Glu200, and Glu246, and combined alanine substitutions of the latter similarly elevates resting NFAT activity. Thus, O-GlcNAcylated T221 and interactions with coordinating residues is required for normal TRPC6 channel conductance and NFAT activation., Competing Interests: D.A.K. receives grant support from Boehringer Ingelheim pursuing the use of a selective TRPC6 antagonist for treatment of Duchenne Muscular Dystrophy., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
6. Single serine on TSC2 exerts biased control over mTORC1 activation mediated by ERK1/2 but not Akt.
- Author
-
Dunkerly-Eyring BL, Pan S, Pinilla-Vera M, McKoy D, Mishra S, Grajeda Martinez MI, Oeing CU, Ranek MJ, and Kass DA
- Subjects
- Humans, Proto-Oncogene Proteins c-akt metabolism, Serine metabolism, Tumor Suppressor Proteins metabolism, MAP Kinase Signaling System, Mechanistic Target of Rapamycin Complex 1 metabolism, Tuberous Sclerosis Complex 2 Protein genetics, Tuberous Sclerosis Complex 2 Protein metabolism
- Abstract
Tuberous sclerosis complex-2 (TSC2) negatively regulates mammalian target of rapamycin complex 1 (mTORC1), and its activity is reduced by protein kinase B (Akt) and extracellular response kinase (ERK1/2) phosphorylation to activate mTORC1. Serine 1364 (human) on TSC2 bidirectionally modifies mTORC1 activation by pathological growth factors or hemodynamic stress but has no impact on resting activity. We now show this modification biases to ERK1/2 but not Akt-dependent TSC2-mTORC1 activation. Endothelin-1-stimulated mTORC1 requires ERK1/2 activation and is bidirectionally modified by phospho-mimetic (S1364E) or phospho-silenced (S1364A) mutations. However, mTORC1 activation by Akt-dependent stimuli (insulin or PDGF) is unaltered by S1364 modification. Thrombin stimulates both pathways, yet only the ERK1/2 component is modulated by S1364. S1364 also has negligible impact on mTORC1 regulation by energy or nutrient status. In vivo, diet-induced obesity, diabetes, and fatty liver couple to Akt activation and are also unaltered by TSC2 S1364 mutations. This contrasts to prior reports showing a marked impact of both on pathological pressure-stress. Thus, S1364 provides ERK1/2-selective mTORC1 control and a genetic means to modify pathological versus physiological mTOR stimuli., (© 2022 Dunkerly-Eyring et al.)
- Published
- 2022
- Full Text
- View/download PDF
7. Editorial: Post-translational Modifications and Compartmentalized Protein Quality Control in Cardiac Muscle and Disease.
- Author
-
Ranek MJ, Gomes AV, and Su H
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
8. Editorial: Targeting Cardiac Proteotoxicity.
- Author
-
Ranek MJ, Bhuiyan MS, and Wang X
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2021
- Full Text
- View/download PDF
9. MTORC1-Regulated Metabolism Controlled by TSC2 Limits Cardiac Reperfusion Injury.
- Author
-
Oeing CU, Jun S, Mishra S, Dunkerly-Eyring BL, Chen A, Grajeda MI, Tahir UA, Gerszten RE, Paolocci N, Ranek MJ, and Kass DA
- Subjects
- Animals, Carnitine analogs & derivatives, Carnitine metabolism, Cells, Cultured, Glucose metabolism, Ischemic Preconditioning, Lactic Acid metabolism, Male, Mice, Mice, Inbred C57BL, Mitochondria, Heart metabolism, Mutation, Myocardial Reperfusion Injury therapy, Oxygen metabolism, Phosphorylation, Rats, Tuberous Sclerosis Complex 2 Protein genetics, Tuberous Sclerosis Complex 2 Protein metabolism, Glycolysis, Mechanistic Target of Rapamycin Complex 1 metabolism, Myocardial Reperfusion Injury metabolism, Myocytes, Cardiac metabolism
- Abstract
Rationale: The mTORC1 (mechanistic target of rapamycin complex-1) controls metabolism and protein homeostasis and is activated following ischemia reperfusion (IR) injury and by ischemic preconditioning (IPC). However, studies vary as to whether this activation is beneficial or detrimental, and its influence on metabolism after IR is little reported. A limitation of prior investigations is their use of broad gain/loss of mTORC1 function, mostly applied before ischemic stress. This can be circumvented by regulating one serine (S1365) on TSC2 (tuberous sclerosis complex) to achieve bidirectional mTORC1 modulation but only with TCS2-regulated costimulation., Objective: We tested the hypothesis that reduced TSC2 S1365 phosphorylation protects the myocardium against IR and is required for IPC by amplifying mTORC1 activity to favor glycolytic metabolism., Methods and Results: Mice with either S1365A (TSC2
SA ; phospho-null) or S1365E (TSC2SE ; phosphomimetic) knockin mutations were studied ex vivo and in vivo. In response to IR, hearts from TSC2SA mice had amplified mTORC1 activation and improved heart function compared with wild-type and TSC2SE hearts. The magnitude of protection matched IPC. IPC requited less S1365 phosphorylation, as TSC2SE hearts gained no benefit and failed to activate mTORC1 with IPC. IR metabolism was altered in TSC2SA , with increased mitochondrial oxygen consumption rate and glycolytic capacity (stressed/maximal extracellular acidification) after myocyte hypoxia-reperfusion. In whole heart, lactate increased and long-chain acylcarnitine levels declined during ischemia. The relative IR protection in TSC2SA was lost by lowering glucose in the perfusate by 36%. Adding fatty acid (palmitate) compensated for reduced glucose in wild type and TSC2SE but not TSC2SA which had the worst post-IR function under these conditions., Conclusions: TSC2-S1365 phosphorylation status regulates myocardial substrate utilization, and its decline activates mTORC1 biasing metabolism away from fatty acid oxidation to glycolysis to confer protection against IR. This pathway is also engaged and reduced TSC2 S1365 phosphorylation required for effective IPC. Graphic Abstract: A graphic abstract is available for this article.- Published
- 2021
- Full Text
- View/download PDF
10. Pulmonary artery pulsatility index predicts right ventricular myofilament dysfunction in advanced human heart failure.
- Author
-
Aslam MI, Jani V, Lin BL, Dunkerly-Eyring B, Livingston CE, Ramachandran A, Ranek MJ, Bedi KC, Margulies KB, Kass DA, and Hsu S
- Subjects
- Humans, Myofibrils, Pulmonary Artery, Ventricular Function, Right, Heart Failure, Heart-Assist Devices, Ventricular Dysfunction, Right
- Published
- 2021
- Full Text
- View/download PDF
11. Phosphorylation Modifications Regulating Cardiac Protein Quality Control Mechanisms.
- Author
-
Mishra S, Dunkerly-Eyring BL, Keceli G, and Ranek MJ
- Abstract
Many forms of cardiac disease, including heart failure, present with inadequate protein quality control (PQC). Pathological conditions often involve impaired removal of terminally misfolded proteins. This results in the formation of large protein aggregates, which further reduce cellular viability and cardiac function. Cardiomyocytes have an intricately collaborative PQC system to minimize cellular proteotoxicity. Increased expression of chaperones or enhanced clearance of misfolded proteins either by the proteasome or lysosome has been demonstrated to attenuate disease pathogenesis, whereas reduced PQC exacerbates pathogenesis. Recent studies have revealed that phosphorylation of key proteins has a potent regulatory role, both promoting and hindering the PQC machinery. This review highlights the recent advances in phosphorylations regulating PQC, the impact in cardiac pathology, and the therapeutic opportunities presented by harnessing these modifications., (Copyright © 2020 Mishra, Dunkerly-Eyring, Keceli and Ranek.)
- Published
- 2020
- Full Text
- View/download PDF
12. CHIP phosphorylation by protein kinase G enhances protein quality control and attenuates cardiac ischemic injury.
- Author
-
Ranek MJ, Oeing C, Sanchez-Hodge R, Kokkonen-Simon KM, Dillard D, Aslam MI, Rainer PP, Mishra S, Dunkerly-Eyring B, Holewinski RJ, Virus C, Zhang H, Mannion MM, Agrawal V, Hahn V, Lee DI, Sasaki M, Van Eyk JE, Willis MS, Page RC, Schisler JC, and Kass DA
- Subjects
- Amino Acid Motifs, Animals, Cyclic GMP-Dependent Protein Kinases genetics, Female, Heart physiopathology, Humans, Ischemia enzymology, Ischemia genetics, Ischemia physiopathology, Male, Mice, Myocardium metabolism, Phosphorylation, Ubiquitin-Protein Ligases chemistry, Ubiquitin-Protein Ligases genetics, Cyclic GMP-Dependent Protein Kinases metabolism, Ischemia metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Proteotoxicity from insufficient clearance of misfolded/damaged proteins underlies many diseases. Carboxyl terminus of Hsc70-interacting protein (CHIP) is an important regulator of proteostasis in many cells, having E3-ligase and chaperone functions and often directing damaged proteins towards proteasome recycling. While enhancing CHIP functionality has broad therapeutic potential, prior efforts have all relied on genetic upregulation. Here we report that CHIP-mediated protein turnover is markedly post-translationally enhanced by direct protein kinase G (PKG) phosphorylation at S20 (mouse, S19 human). This increases CHIP binding affinity to Hsc70, CHIP protein half-life, and consequent clearance of stress-induced ubiquitinated-insoluble proteins. PKG-mediated CHIP-pS20 or expressing CHIP-S20E (phosphomimetic) reduces ischemic proteo- and cytotoxicity, whereas a phospho-silenced CHIP-S20A amplifies both. In vivo, depressing PKG activity lowers CHIP-S20 phosphorylation and protein, exacerbating proteotoxicity and heart dysfunction after ischemic injury. CHIP-S20E knock-in mice better clear ubiquitinated proteins and are cardio-protected. PKG activation provides post-translational enhancement of protein quality control via CHIP.
- Published
- 2020
- Full Text
- View/download PDF
13. PKG1α Cysteine-42 Redox State Controls mTORC1 Activation in Pathological Cardiac Hypertrophy.
- Author
-
Oeing CU, Nakamura T, Pan S, Mishra S, Dunkerly-Eyring BL, Kokkonen-Simon KM, Lin BL, Chen A, Zhu G, Bedja D, Lee DI, Kass DA, and Ranek MJ
- Subjects
- Animals, Aorta, Autophagy physiology, Benzoates metabolism, Biphenyl Compounds metabolism, Constriction, Pathologic, Cyclic GMP-Dependent Protein Kinase Type I genetics, Cysteine metabolism, Endothelin-1 pharmacology, Enzyme Activation, Everolimus pharmacology, Gene Knock-In Techniques, Hydrocarbons, Fluorinated metabolism, Mechanistic Target of Rapamycin Complex 1 antagonists & inhibitors, Mice, Mice, Inbred C57BL, Myocytes, Cardiac drug effects, Oxidation-Reduction, Oxidative Stress, Phosphorylation, Pressure, Proteostasis, Rats, Tuberous Sclerosis Complex 2 Protein genetics, Tuberous Sclerosis Complex 2 Protein metabolism, Cardiomegaly metabolism, Cyclic GMP-Dependent Protein Kinase Type I metabolism, Guanylate Cyclase metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Myocytes, Cardiac metabolism
- Abstract
Rationale: Stimulated PKG1α (protein kinase G-1α) phosphorylates TSC2 (tuberous sclerosis complex 2) at serine 1365, potently suppressing mTORC1 (mechanistic [mammalian] target of rapamycin complex 1) activation by neurohormonal and hemodynamic stress. This reduces pathological hypertrophy and dysfunction and increases autophagy. PKG1α oxidation at cysteine-42 is also induced by these stressors, which blunts its cardioprotective effects., Objective: We tested the dependence of mTORC1 activation on PKG1α C42 oxidation and its capacity to suppress such activation by soluble GC-1 (guanylyl cyclase 1) activation., Methods and Results: Cardiomyocytes expressing wild-type (WT) PKG1α (PKG1α
WT ) or cysteine-42 to serine mutation redox-dead (PKG1αCS/CS ) were exposed to ET-1 (endothelin 1). Cells expressing PKG1αWT exhibited substantial mTORC1 activation (p70 S6K [p70 S6 kinase], 4EBP1 [elF4E binding protein-1], and Ulk1 [Unc-51-like kinase 1] phosphorylation), reduced autophagy/autophagic flux, and abnormal protein aggregation; all were markedly reversed by PKG1αCS/CS expression. Mice with global knock-in of PKG1αCS/CS subjected to pressure overload (PO) also displayed markedly reduced mTORC1 activation, protein aggregation, hypertrophy, and ventricular dysfunction versus PO in PKG1αWT mice. Cardioprotection against PO was equalized between groups by co-treatment with the mTORC1 inhibitor everolimus. TSC2-S1365 phosphorylation increased in PKG1αCS/CS more than PKG1αWT myocardium following PO. TSC2S1365A/S1365A (TSC2 S1365 phospho-null, created by a serine to alanine mutation) knock-in mice lack TSC2 phosphorylation by PKG1α, and when genetically crossed with PKG1αCS/CS mice, protection against PO-induced mTORC1 activation, cardiodepression, and mortality in PKG1αCS/CS mice was lost. Direct stimulation of GC-1 (BAY-602770) offset disparate mTORC1 activation between PKG1αWT and PKG1αCS/CS after PO and blocked ET-1 stimulated mTORC1 in TSC2S1365A -expressing myocytes., Conclusions: Oxidation of PKG1α at C42 reduces its phosphorylation of TSC2, resulting in amplified PO-stimulated mTORC1 activity and associated hypertrophy, dysfunction, and depressed autophagy. This is ameliorated by direct GC-1 stimulation.- Published
- 2020
- Full Text
- View/download PDF
14. Targeting Protein Kinase G to Treat Cardiac Proteotoxicity.
- Author
-
Oeing CU, Mishra S, Dunkerly-Eyring BL, and Ranek MJ
- Abstract
Impaired or insufficient protein kinase G (PKG) signaling and protein quality control (PQC) are hallmarks of most forms of cardiac disease, including heart failure. Their dysregulation has been shown to contribute to and exacerbate cardiac hypertrophy and remodeling, reduced cell survival and disease pathogenesis. Enhancement of PKG signaling and PQC are associated with improved cardiac function and survival in many pre-clinical models of heart disease. While many clinically used pharmacological approaches exist to stimulate PKG, there are no FDA-approved therapies to safely enhance cardiomyocyte PQC. The latter is predominantly due to our lack of knowledge and identification of proteins regulating cardiomyocyte PQC. Recently, multiple studies have demonstrated that PKG regulates PQC in the heart, both during physiological and pathological states. These studies tested already FDA-approved pharmacological therapies to activate PKG, which enhanced cardiomyocyte PQC and alleviated cardiac disease. This review examines the roles of PKG and PQC during disease pathogenesis and summarizes the experimental and clinical data supporting the utility of stimulating PKG to target cardiac proteotoxicity., (Copyright © 2020 Oeing, Mishra, Dunkerly-Eyring and Ranek.)
- Published
- 2020
- Full Text
- View/download PDF
15. In vivo selective inhibition of TRPC6 by antagonist BI 749327 ameliorates fibrosis and dysfunction in cardiac and renal disease.
- Author
-
Lin BL, Matera D, Doerner JF, Zheng N, Del Camino D, Mishra S, Bian H, Zeveleva S, Zhen X, Blair NT, Chong JA, Hessler DP, Bedja D, Zhu G, Muller GK, Ranek MJ, Pantages L, McFarland M, Netherton MR, Berry A, Wong D, Rast G, Qian HS, Weldon SM, Kuo JJ, Sauer A, Sarko C, Moran MM, Kass DA, and Pullen SS
- Subjects
- Animals, Drug Evaluation, Preclinical, Fibrosis, HEK293 Cells, Heart drug effects, Humans, Kidney drug effects, Mice, Cardiomegaly drug therapy, Nephrosclerosis drug therapy, TRPC6 Cation Channel antagonists & inhibitors
- Abstract
Transient receptor potential canonical type 6 (TRPC6) is a nonselective receptor-operated cation channel that regulates reactive fibrosis and growth signaling. Increased TRPC6 activity from enhanced gene expression or gain-of-function mutations contribute to cardiac and/or renal disease. Despite evidence supporting a pathophysiological role, no orally bioavailable selective TRPC6 inhibitor has yet been developed and tested in vivo in disease models. Here, we report an orally bioavailable TRPC6 antagonist (BI 749327; IC
50 13 nM against mouse TRPC6, t1/2 8.5-13.5 hours) with 85- and 42-fold selectivity over the most closely related channels, TRPC3 and TRPC7. TRPC6 calcium conductance results in the stimulation of nuclear factor of activated T cells (NFAT) that triggers pathological cardiac and renal fibrosis and disease. BI 749327 suppresses NFAT activation in HEK293T cells expressing wild-type or gain-of-function TRPC6 mutants (P112Q, M132T, R175Q, R895C, and R895L) and blocks associated signaling and expression of prohypertrophic genes in isolated myocytes. In vivo, BI 749327 (30 mg/kg/day, yielding unbound trough plasma concentration ∼180 nM) improves left heart function, reduces volume/mass ratio, and blunts expression of profibrotic genes and interstitial fibrosis in mice subjected to sustained pressure overload. Additionally, BI 749327 dose dependently reduces renal fibrosis and associated gene expression in mice with unilateral ureteral obstruction. These results provide in vivo evidence of therapeutic efficacy for a selective pharmacological TRPC6 inhibitor with oral bioavailability and suitable pharmacokinetics to ameliorate cardiac and renal stress-induced disease with fibrosis., Competing Interests: Conflict of interest statement: D.M., J.F.D., L.P., D.W., G.R., S.M.W., S.Z., H.S.Q, J.J.K, A.S., and S.S.P. are full-time employees of Boehringer Ingelheim Pharmaceuticals, Inc. M.M, M.R.N, A.B., and C.S. were full-time employees of Boehringer Ingelheim Pharmaceuticals, Inc. A.B. and M.R.N. are listed as coinventors on a US provisional patent application filed by Boehringer Ingelheim Pharmaceuticals, Inc. relevant to this work. J.F.D., N.Z., D.d.C., X.Z., N.T.B., J.A.C., D.P.H., and M.M.M. were employees of Hydra Biosciences, and received options. This work was supported in part by Boehringer Ingelheim Pharmaceuticals, Inc.- Published
- 2019
- Full Text
- View/download PDF
16. PKG1-modified TSC2 regulates mTORC1 activity to counter adverse cardiac stress.
- Author
-
Ranek MJ, Kokkonen-Simon KM, Chen A, Dunkerly-Eyring BL, Vera MP, Oeing CU, Patel CH, Nakamura T, Zhu G, Bedja D, Sasaki M, Holewinski RJ, Van Eyk JE, Powell JD, Lee DI, and Kass DA
- Subjects
- Animals, Autophagy, Cells, Cultured, Disease Progression, Enzyme Activation, Everolimus pharmacology, Female, Gene Knock-In Techniques, HEK293 Cells, Heart Diseases genetics, Heart Diseases pathology, Humans, Hypertrophy drug therapy, Hypertrophy pathology, Male, Mechanistic Target of Rapamycin Complex 1 antagonists & inhibitors, Mice, Mutation, Myocytes, Cardiac pathology, Phosphorylation, Phosphoserine metabolism, Pressure, Rats, Rats, Wistar, Serine genetics, Serine metabolism, Tuberous Sclerosis Complex 2 Protein genetics, Cyclic GMP-Dependent Protein Kinases metabolism, Heart Diseases physiopathology, Heart Diseases prevention & control, Mechanistic Target of Rapamycin Complex 1 metabolism, Tuberous Sclerosis Complex 2 Protein chemistry, Tuberous Sclerosis Complex 2 Protein metabolism
- Abstract
The mechanistic target of rapamycin complex-1 (mTORC1) coordinates regulation of growth, metabolism, protein synthesis and autophagy
1 . Its hyperactivation contributes to disease in numerous organs, including the heart1,2 , although broad inhibition of mTORC1 risks interference with its homeostatic roles. Tuberin (TSC2) is a GTPase-activating protein and prominent intrinsic regulator of mTORC1 that acts through modulation of RHEB (Ras homologue enriched in brain). TSC2 constitutively inhibits mTORC1; however, this activity is modified by phosphorylation from multiple signalling kinases that in turn inhibits (AMPK and GSK-3β) or stimulates (AKT, ERK and RSK-1) mTORC1 activity3-9 . Each kinase requires engagement of multiple serines, impeding analysis of their role in vivo. Here we show that phosphorylation or gain- or loss-of-function mutations at either of two adjacent serine residues in TSC2 (S1365 and S1366 in mice; S1364 and S1365 in humans) can bidirectionally control mTORC1 activity stimulated by growth factors or haemodynamic stress, and consequently modulate cell growth and autophagy. However, basal mTORC1 activity remains unchanged. In the heart, or in isolated cardiomyocytes or fibroblasts, protein kinase G1 (PKG1) phosphorylates these TSC2 sites. PKG1 is a primary effector of nitric oxide and natriuretic peptide signalling, and protects against heart disease10-13 . Suppression of hypertrophy and stimulation of autophagy in cardiomyocytes by PKG1 requires TSC2 phosphorylation. Homozygous knock-in mice that express a phosphorylation-silencing mutation in TSC2 (TSC2(S1365A)) develop worse heart disease and have higher mortality after sustained pressure overload of the heart, owing to mTORC1 hyperactivity that cannot be rescued by PKG1 stimulation. However, cardiac disease is reduced and survival of heterozygote Tsc2S1365A knock-in mice subjected to the same stress is improved by PKG1 activation or expression of a phosphorylation-mimicking mutation (TSC2(S1365E)). Resting mTORC1 activity is not altered in either knock-in model. Therefore, TSC2 phosphorylation is both required and sufficient for PKG1-mediated cardiac protection against pressure overload. The serine residues identified here provide a genetic tool for bidirectional regulation of the amplitude of stress-stimulated mTORC1 activity.- Published
- 2019
- Full Text
- View/download PDF
17. Histone lysine dimethyl-demethylase KDM3A controls pathological cardiac hypertrophy and fibrosis.
- Author
-
Zhang QJ, Tran TAT, Wang M, Ranek MJ, Kokkonen-Simon KM, Gao J, Luo X, Tan W, Kyrychenko V, Liao L, Xu J, Hill JA, Olson EN, Kass DA, Martinez ED, and Liu ZP
- Subjects
- Aminopyridines pharmacology, Animals, Animals, Newborn, Cardiomegaly enzymology, Cells, Cultured, Fibrosis genetics, Gene Expression Profiling, Gene Expression Regulation drug effects, Histone Demethylases antagonists & inhibitors, Histone Demethylases metabolism, Humans, Hydrazones pharmacology, Mice, Knockout, Mice, Transgenic, Myocardium enzymology, Myocardium pathology, Myocytes, Cardiac metabolism, Rats, Sprague-Dawley, Cardiomegaly genetics, Gene Expression Regulation genetics, Histone Demethylases genetics, Myocardium metabolism
- Abstract
Left ventricular hypertrophy (LVH) is a major risk factor for cardiovascular morbidity and mortality. Pathological LVH engages transcriptional programs including reactivation of canonical fetal genes and those inducing fibrosis. Histone lysine demethylases (KDMs) are emerging regulators of transcriptional reprogramming in cancer, though their potential role in abnormal heart growth and fibrosis remains little understood. Here, we investigate gain and loss of function of an H3K9me2 specific demethylase, Kdm3a, and show it promotes LVH and fibrosis in response to pressure-overload. Cardiomyocyte KDM3A activates Timp1 transcription with pro-fibrotic activity. By contrast, a pan-KDM inhibitor, JIB-04, suppresses pressure overload-induced LVH and fibrosis. JIB-04 inhibits KDM3A and suppresses the transcription of fibrotic genes that overlap with genes downregulated in Kdm3a-KO mice versus WT controls. Our study provides genetic and biochemical evidence for a pro-hypertrophic function of KDM3A and proof-of principle for pharmacological targeting of KDMs as an effective strategy to counter LVH and pathological fibrosis.
- Published
- 2018
- Full Text
- View/download PDF
18. Marked disparity of microRNA modulation by cGMP-selective PDE5 versus PDE9 inhibitors in heart disease.
- Author
-
Kokkonen-Simon KM, Saberi A, Nakamura T, Ranek MJ, Zhu G, Bedja D, Kuhn M, Halushka MK, Lee DI, and Kass DA
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Animals, Cyclic GMP metabolism, Cyclic GMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 5 metabolism, Disease Models, Animal, Heart Diseases etiology, Humans, Male, Mice, Natriuretic Peptides metabolism, Nitric Oxide metabolism, Phosphodiesterase 5 Inhibitors therapeutic use, Signal Transduction, 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, Heart Diseases drug therapy, MicroRNAs metabolism, Phosphodiesterase 5 Inhibitors pharmacology, RNA Processing, Post-Transcriptional drug effects
- Abstract
MicroRNAs (miRs) posttranscriptionally regulate mRNA and its translation into protein, and are considered master controllers of genes modulating normal physiology and disease. There is growing interest in how miRs change with drug treatment, and leveraging this for precision guided therapy. Here we contrast 2 closely related therapies, inhibitors of phosphodiesterase type 5 or type 9 (PDE5-I, PDE9-I), given to mice subjected to sustained cardiac pressure overload (PO). Both inhibitors augment cyclic guanosine monophosphate (cGMP) to activate protein kinase G, with PDE5-I regulating nitric oxide (NO) and PDE9-I natriuretic peptide-dependent signaling. While both produced strong phenotypic improvement of PO pathobiology, they surprisingly showed binary differences in miR profiles; PDE5-I broadly reduces more than 120 miRs, including nearly half those increased by PO, whereas PDE9-I has minimal impact on any miR (P < 0.0001). The disparity evolves after pre-miR processing and is organ specific. Lastly, even enhancing NO-coupled cGMP by different methods leads to altered miR regulation. Thus, seemingly similar therapeutic interventions can be barcoded by profound differences in miR signatures, and reversing disease-associated miR changes is not required for therapy success.
- Published
- 2018
- Full Text
- View/download PDF
19. Prevention of PKG-1α Oxidation Suppresses Antihypertrophic/Antifibrotic Effects From PDE5 Inhibition but not sGC Stimulation.
- Author
-
Nakamura T, Zhu G, Ranek MJ, Kokkonen-Simon K, Zhang M, Kim GE, Tsujita K, and Kass DA
- Subjects
- Animals, Cyclic GMP-Dependent Protein Kinase Type I metabolism, Mice, Transgenic, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Phosphorylation drug effects, Signal Transduction drug effects, Cardiomegaly drug therapy, Cyclic GMP-Dependent Protein Kinase Type I drug effects, Heart Failure drug therapy, Phosphodiesterase 5 Inhibitors pharmacology, Sildenafil Citrate pharmacology
- Abstract
Background: Stimulation of sGC (soluble guanylate cyclase) or inhibition of PDE5 (phosphodiesterase type 5) activates PKG (protein kinase G)-1α to counteract cardiac hypertrophy and failure. PKG1α acts within localized intracellular domains; however, its oxidation at cysteine 42, linking homomonomers, alters this localization, impairing suppression of pathological cardiac stress. Because PDE5 and sGC reside in separate microdomains, we speculated that PKG1α oxidation might also differentially influence the effects from their pharmacological modulation., Methods and Results: Knock-in mice expressing a redox-dead PKG1α (PKG1α
C42S ) or littermate controls (PKG1αWT ) were subjected to transaortic constriction to induce pressure overload and treated with a PDE5 inhibitor (sildenafil), sGC activator (BAY602770 [BAY]), or vehicle. In PKG1αWT controls, sildenafil and BAY similarly enhanced PKG activity and reduced pathological hypertrophy/fibrosis and cardiac dysfunction after transaortic constriction. However, sildenafil failed to protect the heart in PKG1αC42S , unlike BAY, which activated PKG and thereby facilitated protective effects. This corresponded with minimal PDE5 activation in PKG1αC42S exposed to transaortic constriction versus higher activity in controls and little colocalization of PDE5 with PKG1αC42S (versus colocalization with PKG1αWT ) in stressed myocytes., Conclusions: In the stressed heart and myocytes, PKG1α C42-disulfide formation contributes to PDE5 activation. This augments the pathological role of PDE5 and so in turn enhances the therapeutic impact from its inhibition. PKG1α oxidation does not change the benefits from sGC activation. This finding favors the use of sGC activators regardless of PKG1α oxidation and may help guide precision therapy leveraging the cyclic GMP/PKG pathway to treat heart disease., (© 2018 American Heart Association, Inc.)- Published
- 2018
- Full Text
- View/download PDF
20. The role of heat shock proteins and co-chaperones in heart failure.
- Author
-
Ranek MJ, Stachowski MJ, Kirk JA, and Willis MS
- Subjects
- Animals, Heart Failure physiopathology, Heat-Shock Proteins metabolism, Humans, Mice, Molecular Chaperones genetics, Molecular Chaperones metabolism, Rats, Heart Failure genetics, Heat-Shock Proteins genetics, Myocytes, Cardiac metabolism
- Abstract
The ongoing contractile and metabolic demands of the heart require a tight control over protein quality control, including the maintenance of protein folding, turnover and synthesis. In heart disease, increases in mechanical and oxidative stresses, post-translational modifications (e.g., phosphorylation), for example, decrease protein stability to favour misfolding in myocardial infarction, heart failure or ageing. These misfolded proteins are toxic to cardiomyocytes, directly contributing to the common accumulation found in human heart failure. One of the critical class of proteins involved in protecting the heart against these threats are molecular chaperones, including the heat shock protein70 (HSP70), HSP90 and co-chaperones CHIP (carboxy terminus of Hsp70-interacting protein, encoded by the Stub1 gene) and BAG-3 (BCL2-associated athanogene 3). Here, we review their emerging roles in the maintenance of cardiomyocytes in human and experimental models of heart failure, including their roles in facilitating the removal of misfolded and degraded proteins, inhibiting apoptosis and maintaining the structural integrity of the sarcomere and regulation of nuclear receptors. Furthermore, we discuss emerging evidence of increased expression of extracellular HSP70, HSP90 and BAG-3 in heart failure, with complementary independent roles from intracellular functions with important therapeutic and diagnostic considerations. While our understanding of these major HSPs in heart failure is incomplete, there is a clear potential role for therapeutic modulation of HSPs in heart failure with important contextual considerations to counteract the imbalance of protein damage and endogenous protein quality control systems.This article is part of the theme issue 'Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective'., (© 2017 The Author(s).)
- Published
- 2018
- Full Text
- View/download PDF
21. Mast cells regulate myofilament calcium sensitization and heart function after myocardial infarction.
- Author
-
Ngkelo A, Richart A, Kirk JA, Bonnin P, Vilar J, Lemitre M, Marck P, Branchereau M, Le Gall S, Renault N, Guerin C, Ranek MJ, Kervadec A, Danelli L, Gautier G, Blank U, Launay P, Camerer E, Bruneval P, Menasche P, Heymes C, Luche E, Casteilla L, Cousin B, Rodewald HR, Kass DA, and Silvestre JS
- Subjects
- Animals, Carboxypeptidases A genetics, Carboxypeptidases A metabolism, Cyclic AMP-Dependent Protein Kinases genetics, Cyclic AMP-Dependent Protein Kinases metabolism, Mice, Mice, Knockout, Myocardial Contraction genetics, Myocardial Infarction genetics, Myocardial Infarction pathology, Myocardial Infarction physiopathology, Myocardium pathology, Myofibrils pathology, Proteolysis, Proto-Oncogene Proteins c-kit genetics, Proto-Oncogene Proteins c-kit metabolism, Receptor, PAR-2 genetics, Receptor, PAR-2 metabolism, Calcium metabolism, Calcium Signaling, Mast Cells metabolism, Myocardial Infarction metabolism, Myocardium metabolism, Myofibrils metabolism
- Abstract
Acute myocardial infarction (MI) is a severe ischemic disease responsible for heart failure and sudden death. Inflammatory cells orchestrate postischemic cardiac remodeling after MI. Studies using mice with defective mast/stem cell growth factor receptor c-Kit have suggested key roles for mast cells (MCs) in postischemic cardiac remodeling. Because c-Kit mutations affect multiple cell types of both immune and nonimmune origin, we addressed the impact of MCs on cardiac function after MI, using the c-Kit-independent MC-deficient (Cpa3(Cre/+)) mice. In response to MI, MC progenitors originated primarily from white adipose tissue, infiltrated the heart, and differentiated into mature MCs. MC deficiency led to reduced postischemic cardiac function and depressed cardiomyocyte contractility caused by myofilament Ca(2+) desensitization. This effect correlated with increased protein kinase A (PKA) activity and hyperphosphorylation of its targets, troponin I and myosin-binding protein C. MC-specific tryptase was identified to regulate PKA activity in cardiomyocytes via protease-activated receptor 2 proteolysis. This work reveals a novel function for cardiac MCs modulating cardiomyocyte contractility via alteration of PKA-regulated force-Ca(2+) interactions in response to MI. Identification of this MC-cardiomyocyte cross-talk provides new insights on the cellular and molecular mechanisms regulating the cardiac contractile machinery and a novel platform for therapeutically addressable regulators., (©2016 Ngkelo et al.)
- Published
- 2016
- Full Text
- View/download PDF
22. Genetically induced moderate inhibition of 20S proteasomes in cardiomyocytes facilitates heart failure in mice during systolic overload.
- Author
-
Ranek MJ, Zheng H, Huang W, Kumarapeli AR, Li J, Liu J, and Wang X
- Subjects
- Animals, Aortic Diseases complications, Aortic Diseases enzymology, Cardiomegaly enzymology, Cardiomegaly etiology, Heart Failure, Heart Ventricles enzymology, Heart Ventricles pathology, Mice, Transgenic, Proteolysis, Ubiquitination, Ventricular Pressure, Myocytes, Cardiac enzymology, Proteasome Endopeptidase Complex genetics
- Abstract
The in vivo function status of the ubiquitin-proteasome system (UPS) in pressure overloaded hearts remains undefined. Cardiotoxicity was observed during proteasome inhibitor chemotherapy, especially in those with preexisting cardiovascular conditions; however, proteasome inhibition (PsmI) was also suggested by some experimental studies as a potential therapeutic strategy to curtail cardiac hypertrophy. Here we used genetic approaches to probe cardiac UPS performance and determine the impact of cardiomyocyte-restricted PsmI (CR-PsmI) on cardiac responses to systolic overload. Transgenic mice expressing an inverse reporter of the UPS (GFPdgn) were subject to transverse aortic constriction (TAC) to probe myocardial UPS performance during systolic overload. Mice with or without moderate CR-PsmI were subject to TAC and temporally characterized for cardiac responses to moderate and severe systolic overload. After moderate TAC (pressure gradient: ~40mmHg), cardiac UPS function was upregulated during the first two weeks but turned to functional insufficiency between 6 and 12weeks as evidenced by the dynamic changes in GFPdgn protein levels, proteasome peptidase activities, and total ubiquitin conjugates. Severe TAC (pressure gradients >60mmHg) led to UPS functional insufficiency within a week. Moderate TAC elicited comparable hypertrophic responses between mice with and without genetic CR-PsmI but caused cardiac malfunction in CR-PsmI mice significantly earlier than those without CR-PsmI. In mice subject to severe TAC, CR-PsmI inhibited cardiac hypertrophy but led to rapidly progressed heart failure and premature death, associated with a pronounced increase in cardiomyocyte death. It is concluded that cardiac UPS function is dynamically altered, with the initial brief upregulation of proteasome function being adaptive; and CR-PsmI facilitates cardiac malfunction during systolic overload., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
23. Prevention of PKG1α oxidation augments cardioprotection in the stressed heart.
- Author
-
Nakamura T, Ranek MJ, Lee DI, Shalkey Hahn V, Kim C, Eaton P, and Kass DA
- Subjects
- Animals, Cardiomegaly genetics, Cardiomegaly pathology, Cardiomegaly prevention & control, Cyclic GMP-Dependent Protein Kinase Type I genetics, Disease Models, Animal, Humans, Mice, Mice, Transgenic, Myocytes, Cardiac pathology, Oxidation-Reduction, Protein Transport genetics, TRPC Cation Channels genetics, TRPC Cation Channels metabolism, TRPC6 Cation Channel, Cardiomegaly enzymology, Cyclic GMP-Dependent Protein Kinase Type I metabolism, Myocytes, Cardiac enzymology, Oxidative Stress
- Abstract
The cGMP-dependent protein kinase-1α (PKG1α) transduces NO and natriuretic peptide signaling; therefore, PKG1α activation can benefit the failing heart. Disease modifiers such as oxidative stress may depress the efficacy of PKG1α pathway activation and underlie variable clinical results. PKG1α can also be directly oxidized, forming a disulfide bond between homodimer subunits at cysteine 42 to enhance oxidant-stimulated vasorelaxation; however, the impact of PKG1α oxidation on myocardial regulation is unknown. Here, we demonstrated that PKG1α is oxidized in both patients with heart disease and in rodent disease models. Moreover, this oxidation contributed to adverse heart remodeling following sustained pressure overload or Gq agonist stimulation. Compared with control hearts and myocytes, those expressing a redox-dead protein (PKG1α(C42S)) better adapted to cardiac stresses at functional, histological, and molecular levels. Redox-dependent changes in PKG1α altered intracellular translocation, with the activated, oxidized form solely located in the cytosol, whereas reduced PKG1α(C42S) translocated to and remained at the outer plasma membrane. This altered PKG1α localization enhanced suppression of transient receptor potential channel 6 (TRPC6), thereby potentiating antihypertrophic signaling. Together, these results demonstrate that myocardial PKG1α oxidation prevents a beneficial response to pathological stress, may explain variable responses to PKG1α pathway stimulation in heart disease, and indicate that maintaining PKG1α in its reduced form may optimize its intrinsic cardioprotective properties.
- Published
- 2015
- Full Text
- View/download PDF
24. Functional Amyloid Signaling via the Inflammasome, Necrosome, and Signalosome: New Therapeutic Targets in Heart Failure.
- Author
-
Parry TL, Melehani JH, Ranek MJ, and Willis MS
- Abstract
As the most common cause of death and disability, globally, heart disease remains an incompletely understood enigma. A growing number of cardiac diseases are being characterized by the presence of misfolded proteins underlying their pathophysiology, including cardiac amyloidosis and dilated cardiomyopathy (DCM). At least nine precursor proteins have been implicated in the development of cardiac amyloidosis, most commonly caused by multiple myeloma light chain disease and disease-causing mutant or wildtype transthyretin (TTR). Similarly, aggregates with PSEN1 and COFILIN-2 have been identified in up to one-third of idiopathic DCM cases studied, indicating the potential predominance of misfolded proteins in heart failure. In this review, we present recent evidence linking misfolded proteins mechanistically with heart failure and present multiple lines of new therapeutic approaches that target the prevention of misfolded proteins in cardiac TTR amyloid disease. These include multiple small molecule pharmacological chaperones now in clinical trials designed specifically to support TTR folding by rational design, such as tafamidis, and chaperones previously developed for other purposes, such as doxycycline and tauroursodeoxycholic acid. Last, we present newly discovered non-pathological "functional" amyloid structures, such as the inflammasome and necrosome signaling complexes, which can be activated directly by amyloid. These may represent future targets to successfully attenuate amyloid-induced proteotoxicity in heart failure, as the inflammasome, for example, is being therapeutically inhibited experimentally in autoimmune disease. Together, these studies demonstrate multiple novel points in which new therapies may be used to primarily prevent misfolded proteins or to inhibit their downstream amyloid-mediated effectors, such as the inflammasome, to prevent proteotoxicity in heart failure.
- Published
- 2015
- Full Text
- View/download PDF
25. Phosphodiesterase 9A controls nitric-oxide-independent cGMP and hypertrophic heart disease.
- Author
-
Lee DI, Zhu G, Sasaki T, Cho GS, Hamdani N, Holewinski R, Jo SH, Danner T, Zhang M, Rainer PP, Bedja D, Kirk JA, Ranek MJ, Dostmann WR, Kwon C, Margulies KB, Van Eyk JE, Paulus WJ, Takimoto E, and Kass DA
- Subjects
- 3',5'-Cyclic-AMP Phosphodiesterases antagonists & inhibitors, 3',5'-Cyclic-AMP Phosphodiesterases deficiency, 3',5'-Cyclic-AMP Phosphodiesterases genetics, Animals, Aortic Valve Stenosis complications, Cardiomegaly drug therapy, Cardiomegaly etiology, Humans, Male, Mice, Mice, Inbred C57BL, Muscle Cells enzymology, Myocardium enzymology, Natriuretic Peptides metabolism, Nitric Oxide Synthase, Phosphodiesterase Inhibitors pharmacology, Phosphodiesterase Inhibitors therapeutic use, Pressure, Signal Transduction drug effects, Stress, Physiological, Up-Regulation, 3',5'-Cyclic-AMP Phosphodiesterases metabolism, Cardiomegaly enzymology, Cardiomegaly metabolism, Cyclic GMP metabolism, Nitric Oxide metabolism
- Abstract
Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP, nitric oxide signalling is often depressed by heart disease. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.
- Published
- 2015
- Full Text
- View/download PDF
26. Soluble guanylate cyclase is required for systemic vasodilation but not positive inotropy induced by nitroxyl in the mouse.
- Author
-
Zhu G, Groneberg D, Sikka G, Hori D, Ranek MJ, Nakamura T, Takimoto E, Paolocci N, Berkowitz DE, Friebe A, and Kass DA
- Subjects
- Animals, Aorta drug effects, Cyclic GMP physiology, Cyclic GMP-Dependent Protein Kinase Type I chemistry, Cyclic GMP-Dependent Protein Kinase Type I deficiency, Cyclic GMP-Dependent Protein Kinase Type I genetics, Cysteine chemistry, Guanylate Cyclase deficiency, Guanylate Cyclase genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Muscle, Smooth, Vascular physiopathology, Myocardial Contraction drug effects, Myocardial Contraction physiology, Myocardium metabolism, Nitric Oxide physiology, Nitric Oxide Donors pharmacology, Oxidation-Reduction, Receptors, Cytoplasmic and Nuclear deficiency, Receptors, Cytoplasmic and Nuclear genetics, Second Messenger Systems physiology, Soluble Guanylyl Cyclase, Sulfonamides pharmacology, Vasodilation drug effects, Cardiotonic Agents pharmacology, Guanylate Cyclase physiology, Nitrogen Oxides pharmacology, Receptors, Cytoplasmic and Nuclear physiology, Vasodilation physiology
- Abstract
Nitroxyl (HNO), the reduced and protonated form of nitric oxide (NO·), confers unique physiological effects including vasorelaxation and enhanced cardiac contractility. These features have spawned current pharmaceutical development of HNO donors as heart failure therapeutics. HNO interacts with selective redox sensitive cysteines to effect signaling but is also proposed to activate soluble guanylate cyclase (sGC) in vitro to induce vasodilation and potentially enhance contractility. Here, we tested whether sGC stimulation is required for these HNO effects in vivo and if HNO also modifies a redox-sensitive cysteine (C42) in protein kinase G-1α to control vasorelaxation. Intact mice and isolated arteries lacking the sGC-β subunit (sGCKO, results in full sGC deficiency) or expressing solely a redox-dead C42S mutant protein kinase G-1α were exposed to the pure HNO donor, CXL-1020. CXL-1020 induced dose-dependent systemic vasodilation while increasing contractility in controls; however, vasodilator effects were absent in sGCKO mice whereas contractility response remained. The CXL-1020 dose reversing 50% of preconstricted force in aortic rings was ≈400-fold greater in sGCKO than controls. Cyclic-GMP and cAMP levels were unaltered in myocardium exposed to CXL-1020, despite its inotropic-vasodilator activity. In protein kinase G-1α(C42S) mice, CXL-1020 induced identical vasorelaxation in vivo and in isolated aortic and mesenteric vessels as in littermate controls. In both groups, dilation was near fully blocked by pharmacologically inhibiting sGC. Thus, sGC and cGMP-dependent signaling are necessary and sufficient for HNO-induced vasodilation in vivo but are not required for positive inotropic action. Redox modulation of protein kinase G-1α is not a mechanism for HNO-mediated vasodilation., (© 2014 American Heart Association, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
27. Muscarinic 2 receptors modulate cardiac proteasome function in a protein kinase G-dependent manner.
- Author
-
Ranek MJ, Kost CK Jr, Hu C, Martin DS, and Wang X
- Subjects
- Animals, Animals, Newborn, Blotting, Western, Cyclic GMP-Dependent Protein Kinases genetics, Green Fluorescent Proteins genetics, Mice, Mice, Transgenic, Microscopy, Confocal, Microscopy, Fluorescence, Myocytes, Cardiac cytology, Protein Processing, Post-Translational, Proteolysis, RNA, Messenger genetics, Rats, Real-Time Polymerase Chain Reaction, Receptor, Muscarinic M2 genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Cyclic GMP-Dependent Protein Kinases metabolism, Green Fluorescent Proteins metabolism, Myocytes, Cardiac metabolism, Proteasome Endopeptidase Complex metabolism, Receptor, Muscarinic M2 metabolism, Ubiquitin metabolism
- Abstract
Proteasome function insufficiency and inadequate protein quality control are strongly implicated in a large subset of cardiovascular disease and may play an important role in their pathogenesis. Protein degradation by the ubiquitin proteasome system can be physiologically regulated. Cardiac muscarinic 2 (M2) receptors were pharmacologically interrogated in intact mice and cultured neonatal rat ventricular myocytes (NRVMs). Proteasome-mediated proteolysis was measured with a surrogate misfolded protein, proteasome peptidase assay, and by characterizing key proteasome subunits. Successful M2 receptor manipulation in cardiomyocytes was determined by measuring an endogenous protein substrate, and in mice, the cardiovascular physiological response. M2 receptor stimulation was associated with increased proteasome-mediated proteolysis and enhanced peptidase activities, while M2 receptor inhibition yielded opposing results. Additionally, M2 receptor manipulation did not alter abundance of the key proteasome subunits, Rpt6 and β5, but significantly shifted their isoelectric points. Inhibition of protein kinase G abrogated the stimulatory effects on proteasome-mediated proteolysis from M2 receptor activation. We conclude that M2 receptor stimulation enhances, whereas M2 receptor inhibition reduces, proteasome-mediated proteolysis likely through posttranslational modifications. Protein kinase G appears to be the mediator of the M2 receptors actions., (Copyright © 2014 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
28. Protein kinase g positively regulates proteasome-mediated degradation of misfolded proteins.
- Author
-
Ranek MJ, Terpstra EJ, Li J, Kass DA, and Wang X
- Subjects
- Adenoviridae genetics, Animals, Cardiovascular Diseases genetics, Cardiovascular Diseases metabolism, Cells, Cultured, Cyclic GMP-Dependent Protein Kinase Type I genetics, Cyclic GMP-Dependent Protein Kinase Type I metabolism, Desmin metabolism, Enzyme Activation drug effects, Enzyme Activation physiology, Green Fluorescent Proteins genetics, Humans, Male, Mice, Mice, Transgenic, Myocytes, Cardiac cytology, Phosphodiesterase 5 Inhibitors pharmacology, Piperazines pharmacology, Protein Processing, Post-Translational physiology, Purines pharmacology, RNA, Small Interfering genetics, Rats, Sildenafil Citrate, Sulfones pharmacology, Cyclic GMP-Dependent Protein Kinases genetics, Cyclic GMP-Dependent Protein Kinases metabolism, Myocytes, Cardiac enzymology, Proteasome Endopeptidase Complex metabolism, Proteostasis Deficiencies genetics, Proteostasis Deficiencies metabolism
- Abstract
Background: Proteasome functional insufficiency is implicated in a large subset of cardiovascular diseases and may play an important role in their pathogenesis. The regulation of proteasome function is poorly understood, hindering the development of effective strategies to improve proteasome function., Methods and Results: Protein kinase G (PKG) was manipulated genetically and pharmacologically in cultured cardiomyocytes. Activation of PKG increased proteasome peptidase activities, facilitated proteasome-mediated degradation of surrogate (enhanced green fluorescence protein modified by carboxyl fusion of degron CL1) and bona fide (CryAB(R120G)) misfolded proteins, and attenuated CryAB(R120G) overexpression-induced accumulation of ubiquitinated proteins and cellular injury. PKG inhibition elicited the opposite responses. Differences in the abundance of the key 26S proteasome subunits Rpt6 and β5 between the PKG-manipulated and control groups were not statistically significant, but the isoelectric points were shifted by PKG activation. In transgenic mice expressing a surrogate substrate (GFPdgn), PKG activation by sildenafil increased myocardial proteasome activities and significantly decreased myocardial GFPdgn protein levels. Sildenafil treatment significantly increased myocardial PKG activity and significantly reduced myocardial accumulation of CryAB(R120G), ubiquitin conjugates, and aberrant protein aggregates in mice with CryAB(R120G)-based desmin-related cardiomyopathy. No discernible effect on bona fide native substrates of the ubiquitin-proteasome system was observed from PKG manipulation in vitro or in vivo., Conclusions: PKG positively regulates proteasome activities and proteasome-mediated degradation of misfolded proteins, likely through posttranslational modifications to proteasome subunits. This may be a new mechanism underlying the benefit of PKG stimulation in treating cardiac diseases. Stimulation of PKG by measures such as sildenafil administration is potentially a new therapeutic strategy to treat cardiac proteinopathies.
- Published
- 2013
- Full Text
- View/download PDF
29. Autophagy and p62 in cardiac proteinopathy.
- Author
-
Zheng Q, Su H, Ranek MJ, and Wang X
- Subjects
- Adaptation, Physiological physiology, Animals, Cardiomyopathies genetics, Cardiomyopathies physiopathology, Cells, Cultured, Desmin physiology, Genes, Reporter, Mice, Mice, Transgenic, Microscopy, Electron, Mutagenesis physiology, Myocytes, Cardiac ultrastructure, Proteostasis Deficiencies genetics, Proteostasis Deficiencies physiopathology, Rats, Stress, Physiological physiology, Transcription Factor TFIIH, Transcription Factors genetics, Ubiquitination physiology, alpha-Crystallin B Chain genetics, alpha-Crystallin B Chain physiology, Autophagy physiology, Cardiomyopathies pathology, Desmin genetics, Myocytes, Cardiac pathology, Myocytes, Cardiac physiology, Proteostasis Deficiencies pathology, Transcription Factors physiology
- Abstract
Rationale: Recent studies suggest an important role of autophagy in protection against αB-crystallin-based (CryAB(R120G)) desmin-related cardiomyopathies (DRC), but this has not been demonstrated in a different model of cardiac proteinopathy. Mechanisms underlying the response of cardiomyocytes to proteotoxic stress remain incompletely understood., Objective: Our first objective was to determine whether and how the autophagic activity is changed in a mouse model of desminopathy. We also investigated the role of p62 in the protein quality control of cardiomyocytes., Methods and Results: Using an autophagosome reporter and determining changes in LC3-II protein levels in response to lysosomal inhibition, we found significantly increased autophagic flux in mouse hearts with transgenic overexpression of a DRC-linked mutant desmin. Similarly, autophagic flux was increased in cultured neonatal rat ventricular myocytes (NRVMs) expressing a mutant desmin. Suppression of autophagy by 3-methyladenine increased, whereas enhancement of autophagy by rapamycin reduced the ability of a comparable level of mutant desmin overexpression to accumulate ubiquitinated proteins in NRVMs. Furthermore, p62 mRNA and protein expression was significantly up-regulated in cardiomyocytes by transgenic overexpression of the mutant desmin or CryAB(R120G) both in intact mice and in vitro. The p62 depletion impaired aggresome and autophagosome formation, exacerbated cell injury, and decreased cell viability in cultured NRVMs expressing the misfolded proteins., Conclusions: Autophagic flux is increased in desminopathic hearts, and as previously suggested in CryAB(R120G)-based DRC, this increased autophagic flux serves as an adaptive response to overexpression of misfolded proteins. The p62 is up-regulated in mouse proteinopathic hearts. The p62 promotes aggresome formation and autophagy activation and protects cardiomyocytes against proteotoxic stress.
- Published
- 2011
- Full Text
- View/download PDF
30. Activation of the ubiquitin-proteasome system in doxorubicin cardiomyopathy.
- Author
-
Ranek MJ and Wang X
- Subjects
- Animals, Antibiotics, Antineoplastic adverse effects, Apoptosis drug effects, Cardiomyopathies chemically induced, Cardiomyopathies enzymology, Doxorubicin adverse effects, Humans, Mice, Myocytes, Cardiac drug effects, Signal Transduction, Antibiotics, Antineoplastic pharmacology, Cardiomyopathies physiopathology, Doxorubicin pharmacology, Heart drug effects, Ubiquitin-Protein Ligase Complexes metabolism
- Abstract
Doxorubicin (Dox) is a very potent anticancer agent, but its use is limited by its dose-dependent, irreversible cardiotoxicity. Despite intensive research efforts, the mechanism of Dox cardiotoxicity remains poorly understood, so very limited means are available for its prevention or effective management. Recent studies have revealed that a therapeutic dose of Dox can activate proteolysis in cardiomyocytes that is mediated by the ubiquitin-proteasome system (UPS), and that the UPS-mediated degradation of a number of pivotal cardiac transcription factors and/or survival factors is enhanced by Dox treatment. These findings suggest that Dox-induced UPS activation may represent a new mechanism underlying Dox cardiotoxicity. Notably, recent experimental studies suggest that proteasome activation promotes cardiac remodeling during hypertension. This review surveys the current literature on the impact of Dox on the UPS and the potential mechanisms by which UPS activation may compromise the heart during Dox therapy.
- Published
- 2009
- Full Text
- View/download PDF
31. Protein quality control and degradation in cardiomyocytes.
- Author
-
Wang X, Su H, and Ranek MJ
- Subjects
- Animals, Autophagy, Heart Failure pathology, Humans, Molecular Chaperones metabolism, Myocytes, Cardiac pathology, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism, Heart Failure metabolism, Muscle Proteins metabolism, Myocytes, Cardiac metabolism, Protein Folding
- Abstract
The heart is constantly under stress and cardiomyocytes face enormous challenges to correctly fold nascent polypeptides and keep mature proteins from denaturing. To meet the challenge, cardiomyocytes have developed multi-layered protein quality control (PQC) mechanisms which are carried out primarily by chaperones and ubiquitin-proteasome system mediated proteolysis. Autophagy may also participate in PQC in cardiomyocytes, especially under pathological conditions. Cardiac PQC often becomes inadequate in heart disease, which may play an important role in the development of congestive heart failure.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.